Next Article in Journal
Validation of the Monocyte Activation Test Demonstrating Equivalence to the Rabbit Pyrogen Test
Previous Article in Journal
The Molecular Mechanisms Underlying Dercum’s Disease: Exploring the Intersection of Obesity, Pain, and Inflammation
Previous Article in Special Issue
Dysregulation of MicroRNAs in Hepatocellular Carcinoma: Targeting Oncogenic Signaling Pathways for Innovative Therapies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epithelial to Mesenchymal Transition Transcriptional Regulator ZEB1 in Liver Cancer: Oncogenic Roles and Therapeutic Potential

by
Ester Gonzalez-Sanchez
1,2,3,
Carlos Andres Roldan-Hernandez
1,
Ana Martin-Ramirez
1,
Lucia Garcia-Collado
1,
Laura Fouassier
4 and
Javier Vaquero
1,3,*
1
HepatoBiliary Tumours Lab, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
2
Department of Physiology and Pharmacology, University of Salamanca, 37007 Salamanca, Spain
3
National Biomedical Research Institute on Liver and Gastrointestinal Diseases (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
4
Université Paris Cité, NABI, CNRS UMR8175, Inserm U1334, F-75006 Paris, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(22), 11135; https://doi.org/10.3390/ijms262211135
Submission received: 15 September 2025 / Revised: 8 November 2025 / Accepted: 13 November 2025 / Published: 18 November 2025
(This article belongs to the Special Issue Pathogenesis and Molecular Treatment of Primary Liver Cancer)

Abstract

Zinc finger E-box binding homeobox 1 (ZEB1) is a member of the zinc finger homeodomain transcription factor family, with a pivotal role in regulating the epithelial to mesenchymal transition (EMT) process. Increasing evidence suggests that ZEB1 is overexpressed in liver tumors, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), and it correlates with advanced disease features and reduced overall survival. Here, we examine ZEB1 molecular functions, regulatory networks and contribution to tumorigenesis. We also discuss the emerging therapeutic strategies and future research directions aimed at targeting the ZEB1 molecular network to improve the outcome of liver cancer patients.

1. Introduction

Liver cancer includes hepatocellular carcinoma (HCC), which is the most prevalent form of primary liver cancer and accounts for approximately 75–85% of cases [1], and cholangiocarcinoma (CCA), which arises from the epithelial cells that line the bile ducts, and represents around 10–15% of all liver cancer cases [2]. Despite fundamental differences in etiological factors, genetic drivers and tumor microenvironment (TME) characteristics, both HCC and CCA are characterized by aggressive behavior, poor prognosis and limited therapeutic options, especially in the advanced stages of the disease, which occur in the majority of patients due to frequent late-stage diagnosis of these malignancies [1,2]. Consequently, liver cancer currently ranks as the third leading cause of cancer-related mortality [3].
Among the mechanisms leading to cell dissemination and metastasis of cancer cells, the epithelial to mesenchymal transition (EMT) is probably the most widely studied mechanism in this regard [4]. EMT, originally observed during embryogenesis, is a reversible dynamic process during which epithelial cells gradually adopt the structural and functional characteristics of mesenchymal cells, including motility, invasiveness, stemness and chemoresistance [4,5]. This process is tightly regulated by different environmental signals from major signaling pathways (i.e., transforming growth factor-beta (TGF-β), Fibroblast growth factor (FGF), wingless/integrated (Wnt), etc.) and a group of EMT-inducing transcription factors (EMT-TF) belonging to three main families (zinc finger E-box-binding homeobox 1/2 (ZEB1/2), snail family transcriptional repressor 1/2 (SNAI1/2) and twist family bHLH transcription factor 1/2 (TWIST1/2)) [4,5]. These factors, mainly identified as negative regulators of epithelial markers such as E-cadherin, have been involved in a plethora of molecular mechanisms involved in various biological processes since then [6,7,8].
The ZEB (Zinc finger E-box-binding homeobox) protein family of transcription factors was first discovered in Drosophila melanogaster and is constituted by two members, ZEB1 and ZEB2 [9]. Both of them share structural similarities and contain two zinc finger clusters that allow them to bind DNA regulatory sequences and, therefore, control the expression of their targets through promoter binding [9]. ZEB1 and ZEB2 are known for their ability to stimulate EMT, but some reports indicate that they can have both redundant and opposing functions, even in the same type of cancer [9,10,11].
The most studied ZEB protein is undoubtedly ZEB1, which has been shown to play multiple roles in regulating the aggressivity and chemoresistance of different tumor types [12]. Emerging evidence suggests that ZEB1 is overexpressed in liver tumors compared to normal liver tissue, and that its expression correlates with advanced disease features such as vascular invasion, intrahepatic metastasis and reduced overall survival [13,14]. Thus, understanding ZEB1-related functions in liver cancer may introduce new ways to therapeutically challenge these aggressive malignancies.
In this review, we aim to give a comprehensive overview of the current knowledge on the biological roles of ZEB1 in the two main subtypes of liver cancer, HCC and CCA. We will first introduce the structural and functional characteristics of ZEB1, followed by a detailed examination of its role in tumor initiation, progression and resistance mechanisms in both types of liver cancer. Finally, we will discuss emerging therapeutic strategies and future directions for research, aimed at exploiting ZEB1 as a biomarker and therapeutic target.

2. ZEB1 Structural Features and Functional Domains

The ZEB1 protein (also known as TCF8 and δEF1) is encoded by the homonym gene located on chromosome 10p11.22 [15]. The protein consists of 1117 amino acids and is a transcription factor that features two clusters of C2H2-type zinc finger domains at both the N-terminal and C-terminal regions, which are responsible for binding to the E-box sequences in target gene promoters with a consensus sequence of 5′-CANNTG-3′ (Figure 1) [16]. The middle region contains a homeodomain that contributes to protein–protein interactions. In addition, ZEB1 contains a Smad interaction domain (SID), a CtBP interaction domain (CID) and p300-P/CBP-associated factor (P/CAF) binding domain (CBD) that are key in the control of its transcriptional activity (Figure 1) [17]. ZEB1 may upregulate or downregulate target gene transcription through the recruitment of different coactivators or cosuppressors via SID, CID and CBD [17]. For example, ZEB1 can directly interact with SMADs through SID to regulate TGF-β-responsive genes. Similarly, p300 and P/CAF can be recruited to CBD to activate gene transcription. Conversely, ZEB1 can repress target gene transcription through the recruitment of the CtBP corepressor complex (histone deacetylases HDAC1/2) via CID (Figure 1) [18]. These cofactors allow ZEB1 to modulate transcription with epigenetic mechanisms, including DNA methylation or histone modifications [19,20]. This extensive list of partners allows ZEB1 to play pleiotropic functions in various biological processes, from embryogenesis to cancer.

3. Regulation of ZEB1 Expression in Liver Cancer

3.1. Transcriptional Regulation

ZEB1 is not only a key effector of EMT but is also integrated into complex regulatory networks involving major signaling pathways, which are frequently dysregulated in liver cancers. The mechanisms of regulation of ZEB1 transcription are summarized in Figure 2. For example, TGF-β is able to upregulate ZEB1 expression in HCC cells in a SMAD2/3-dependent mechanism (Figure 2A) [21,22]. On one side, SMAD2/3 are able to upregulate the expression of Tyrosine-protein kinase-like 7 (PTK7), which in turn upregulates ZEB1 expression. On the other side SMAD2/3 are able to directly bind the ZEB1 promoter to promote ZEB1 transcription [21]. Interestingly, SET (suppressor of variegation, enhancer of zeste, trithorax) and MYND (Myeloid-Nervy-DEAF1) domain containing 3 (SMYD3) is required for TGF-β-induced EMT in HCC cells that are resistant to sorafenib. Indeed, SMYD3 interacts with SMAD2/3 and epigenetically promotes the expression of ZEB1 [21].
Another major signaling cascade involved in ZEB1 regulation is the Wnt/β-catenin pathway (Figure 2A). Generally, WNT signaling activation disrupts the binding of Glycogen synthase kinase 3 beta (GSK3β) to β-catenin to allow the later to translocate to the nucleus and activate target gene transduction, including ZEB1, but several other molecules can modulate this cascade in liver cancer. For example, upstream WNT, Zrt- and Irt-like protein 4 (ZIP4), a zinc transporter located at the plasma membrane, inhibits by direct binding the ubiquitination of Ephrin-B1, which in turn activates WNT5A signaling in HCC cells [23]. Downstream of WNT, several mechanisms also regulate the activity of β-catenin in HCC cells. A thioredoxin domain containing 12 (TXNDC12) interacts with β-catenin and stimulates the nuclear translocation and activation of β-catenin [24]. Nicastrin (NCSTN) also facilitates the translocation of β-catenin to the nucleus. More precisely, Nicastrin activates Notch1 cleavage, which in turn activates AKT serine/threonine kinase (AKT) signaling, leading to GSK3β phosphorylation and liberation of β-catenin from the GSK3β/β-catenin complex [25]. Similarly, Sortilin 1 (SORT1) is able to bind p38 to enhance its stability and promote GSK-3β phosphorylation and the liberation of β-catenin [26]. Other factors affect β-catenin signaling in HCC cells. GINS complex subunit 1 (GINS1) promotes EMT and tumor metastasis by favoring β-catenin signaling [27], while Forkhead box O3 (FOXO3a) reduces the binding of β-catenin to the T-cell factor (TCF) and inhibits β-catenin/TCF target gene expression [28]. Although less studied, some other mechanisms regulating β-catenin have been identified in CCA. For example, the ubiquitin thioesterase OTU deubiquitinase, ubiquitin aldehyde binding 2 (OTUB2), is able to inhibit the lysosomal degradation of β-catenin by interacting with the TNF receptor-associated factor 6 (TRAF6), resulting in increased expression and signaling of β-catenin [29]. Another mechanism involves Protein tyrosine phosphatase type 4A1 (PTP4A1), which activates AKT signaling, which in turn phosphorylates GSK3β, liberating β-catenin to translocate to the nucleus and activate its target gene transcription [30].
Another ZEB1 major regulatory cascade is the Mitogen-activated protein kinase (MAPK) signaling pathway (Figure 2A). Activation of different receptors leads to the activation of this pathway, which ends with the translocation of phosphorylated extracellular signal-regulated kinase (ERK) to the nucleus and its binding to the ZEB1 promoter. In HCC cells, the thyroid hormone responsive protein (THRSP) and ATP binding cassette subfamily A member 8 (ABCA8) inhibit ERK signaling and ZEB1-induced EMT [31,32]. On the contrary, claudin-1 activates the MAPK pathway and ZEB1 expression [33].
Other signals can also regulate ZEB1 expression (Figure 2B). In HCC cells, insulin binding to the insulin receptor triggers the phosphorylation of pyruvate dehydrogenase E1 subunit alpha 1 (PDHA-1) and its nuclear translocation in the company of eukaryotic translation elongation factor 1 gamma (eEF-1γ) and pyruvate kinase M2 (PKM2), which together bind to the ZEB1 promoter to activate its expression [34]. Insulin-like growth factor binding protein 2 (IGFBP2), another protein from the insulin family, mediates the nuclear localization of p65 in HCC cells. p65 is then able to activate nuclear factor kappa B (NFκB) to promote ZEB1 transcription [35]. Hematological and neurological expressed 1-like (HN1L) acts in concert with Activator Protein-2 gamma (AP-2γ) to promote the transcription of methyltransferase 13, (METTL13), which then cooperates with MYC to transcriptionally upregulate the expression of ZEB1 in HCC cells [36]. Also in HCC cells, Protein phosphatase 1G (PPM1G) alters the alternative splicing of transducin beta like 1 X-linked (TBL1X) favoring the TBL1X-S isoform, which acts as an oncogene by promoting ZEB1 expression [37]. Other EMT-TF, such as Snail, can regulate ZEB1 expression. Indeed, exposure of HCC cells to TPA (O-tetradecanoyl-phorbol 13-acetate) promotes Snail binding to the ZEB1 promoter in a complex involving early growth response 1 (EGR1) and Specificity protein 1 (SP1) [38]. On the contrary, F-box and WD repeat domain containing 8 (FBXW8) acts as a negative regulator of ZEB1 expression by promoting the proteasomal degradation of Palmitoyl-Protein Thioesterase 1 (PPT1), which is an activator of ZEB1 expression in HCC cells [39]. Another F-box protein, F-Box and WD Repeat Domain Containing 7 (FBXW7) negatively regulates ZEB1 expression by inhibiting the mechanistic target of rapamycin kinase (mTOR) signaling in CCA cells [40].
Most of these regulatory mechanisms involve autocrine loops that activate signaling cascades in HCC or CCA cells, but signals coming from the TME can also regulate ZEB1 expression (Figure 2C). In this regard, C-C motif chemokine ligand 5 (CCL5) regulates ZEB1 in liver cancer. In HCC, cancer-associated fibroblasts (CAF) produce CCL5 that, upon binding to tumor cells, activates signals that interfere with the ubiquitination and degradation by the proteasome of hypoxia-inducible factor 1 alpha (HIF1α). Then, HIF1α translocates to the nucleus and binds to the ZEB1 promoter to increase its expression [41]. In CCA, mesenchymal stem cells (MSC) are also able to produce CCL5 upon exposure to pro-inflammatory signals. CCL5 then activates the expression of ZEB1 through a mechanism involving AKT-NFκB signaling activation.
On top of these signaling cascades, different environmental cues have been shown to regulate the expression of ZEB1. Indeed, some etiologic factors that lead to the development of HCC, such as alcohol, hepatitis virus (HCV and HBV) or high-fat diet consumption, can induce ZEB1 expression [42,43,44,45]. In the case of ethanol, the mechanism has been associated with the induction of long non-coding (lnc) RNA lnc171, which in turn sponges microRNA (miRNA or miR) mir-873-5p, a negative regulator of ZEB1 mRNA expression [43]. Ethanol and hepatitis virus have been linked to ZEB1 through a mechanism involving chromosome 8q24 amplification, which leads to MYC expression, that in turn promotes ZEB1 expression [42]. In the case of a high-fat diet, prolonged exposure to fatty acids increased the levels of TGF-β and β-catenin, important EMT inducers and ZEB1 regulators [44]. Nevertheless, other environmental factors besides etiologic factors can regulate ZEB1. Di(2-ethylhexyl) phthalate (DEHP), a worldwide common plasticizer, is often observed in patients because of the continual usage of plastic medical devices. Interestingly, long-term DEHP exposure induces EMT by increasing the expression of EMT-TF, including ZEB1 [46]. High expression of ZEB1 in response to these environmental signals increases the invasiveness, stemness and even resistance to sorafenib, of HCC cells [43,44,45,46].

3.2. Post-Transcriptional Regulation

Besides the above-described regulatory signals, ZEB1 is also under tight post-transcriptional regulations at different levels, including mechanisms that regulate the mRNA export to the cytoplasm, the addition of mRNA modifications, and regulation by non-coding RNAs (ncRNAs), which are RNAs that do not encode proteins but play an important role in post-transcriptional control of gene expression. These ncRNA are classified based on length and structure into lncRNA, miRNA and circular RNA (circRNA). All these post-transcriptional mechanisms are summarized in Figure 3. In terms of RNA modifications, N6-methyladenosine (m6A) is the only mRNA modification identified so far in the regulation of ZEB1 in HCC. In this sense, the MYC-associated zinc finger protein (MAZ) is involved in m6A methylation in HCC by targeting the transcriptional regulation of key m6A enzymes [47]. Indeed, MAZ can upregulate the expression of the methylase Methyltransferase-like protein 3 (METTL3) by directly binding to its promoter. In turn, METTL3 can methylate its targets, among which ZEB1 has been identified in HCC. METTL3-mediated m6A methylation of ZEB1 mRNA increases its stabilization [47,48]. Besides writer methylases, the reader YTH domain-containing family protein 3 (YTHDF3) also regulates the stability of ZEB1 in HCC through the m6A modification [49]. Another layer of regulation of this process comes from circ_KIAA1429, which is overexpressed in HCC and facilitates the function of YTHDF3 on ZEB1 mRNA [49]. Thus, mechanisms favoring m6A methylation of ZEB1 mRNA play a role in increasing the invasion and metastasis of HCC by inducing EMT.
Regarding ncRNAs, the most widely described mechanism is the double-negative feedback loop that ZEB1 forms with the miR-200 family, wherein ZEB1 represses the transcription of miR-200 members while being targeted for degradation by the same miRNAs: a mechanism that allows fine-tuned and dynamic regulation of EMT [50]. However, multiple other miRNAs have been described to regulate ZEB1 in recent years. In addition, there are other upstream signals that may regulate the expression of miRNAs, such as lncRNA, which have gained a lot of interest, as they may regulate miRNAs by acting as sponges and, thus, reducing their availability to target ZEB1. The ZEB1 regulatory mechanisms involving non-coding RNAs are summarized in Table 1.
Apart from these mechanisms, ZEB1 can be regulated post-transcriptionally by ncRNAs without its mRNA levels being affected. A non-POU domain containing octamer binding (NONO) is an RNA-binding protein that facilitates ZEB1 mRNA nuclear export. The lncRNA DIO3OS is generally downregulated in HCC patients. This lncRNA represses ZEB1 by interacting with the NONO protein, thereby restricting NONO-mediated nuclear export of ZEB1 mRNA. Thus, reduced DIO3OS leads to increased ZEB1 protein expression, stemness and HCC progression [51]. Conversely, hsa_circ_0007132 has been found to be increased in patients following lenvatinib treatment and HCC lenvatinib-resistant cell lines. Mechanistically, hsa_circ_0007132 binds to NONO and impairs its ubiquitination-mediated degradation, thereby enhancing the NONO-mediated nuclear export of ZEB1 mRNA. Ultimately, ZEB1 increased protein levels and participates in lenvatinib resistance [52].
It is worth mentioning that while we have knowledge about various types of ncRNAs modulating ZEB1 expression in liver cancer for more than 10 years [53,54], and even longer in other tumors, reports on other posttranscriptional regulatory mechanisms such as RNA modifications (i.e., m6A) are as recent as 2025 [47], underscoring the novelty of a growing field in the past few years that will surely help to understand ZEB1’s role, not only in liver cancer but also many other pathological settings.

3.3. Post-Translational Regulation

ZEB1 protein can also undergo post-translational modifications, such as phosphorylation, ubiquitination and acetylation, which influence its stability, localization and transcriptional activity [55]. Very recently, a new form of posttranslational modification has been identified for ZEB1 in HCC, the O-GlcNAcylation. ZEB1 can be O-GlcNAcylated at ser670 by O-GlcNAc transferase (OGT), which increases the protein stability of ZEB1, while O-GlcNAcase (OGA) is able to remove the O-linked N-acetylglucosamine modification. Thus, the expression and activity of OGT and OGA may determine the protein stability of ZEB1 [56]. Nevertheless, in terms of the regulation of ZEB1 expression in liver cancer, most studies are related to ZEB1 degradation by ubiquitination mechanisms (Figure 3). Related to proteasome degradation of ZEB1, deubiquitinase ubiquitin specific peptidase 39 (USP39) and E3 ligase tripartite motif containing 26 (TRIM26) function in an antagonistic pattern in HCC, controlling ZEB1 stability to determine HCC progression. Thus, HCC with a high expression of USP39 show increased ZEB1 expression that determines higher proliferation and migration of HCC cells [57]. Similarly, ubiquitin-specific peptidase 22 (USP22) is involved in the maintenance of ZEB1 stability via its deubiquitinase activity [58]. Another factor involved in ZEB1 degradation by the proteasome is the CRL4-DCAF15 (DDB1 and CUL4 associated factor 15) E3 ubiquitin ligase complex, which recognizes the N-terminal zinc finger domain of ZEB1 to trigger its ubiquitination and posterior degradation, reducing ZEB1 expression and the proliferative and invasive abilities of HCC cells [59]. The ubiquitin-like plant homeodomain (PHD) and really interesting new gene (RING) finger domain-containing protein 1 (UHRF1) protein-associated transcript (UPAT) are also able to promote ZEB1 degradation through the ubiquitin-proteasome pathway in HCC [60]. Besides the proteasome-related mechanisms, ZEB1 protein expression can be reduced by autophagic degradation. In HCC, high mobility group box 1 (HMGB1) promotes homeodomain interacting protein kinase 2 (HIPK2) degradation. HIPK2 is a kinase that plays multifaceted roles in various cellular processes. Among them, HIPK2 regulates ZEB1’s autophagic degradation. Thus, HIPK2 degradation results in protection of ZEB1 from autophagic degradation and increases HCC cell growth. Thus, HMGB1 inhibition can suppress HCC progression via HIPK2-mediated autophagic degradation of ZEB1 [61].
Table 1. Mechanism of regulation of ZEB1 expression involving non-coding RNAs (miRNA and lncRNA).
Table 1. Mechanism of regulation of ZEB1 expression involving non-coding RNAs (miRNA and lncRNA).
TumorFactorRegulatesmiRNARegulatesFactorResultRef.
HCCLnc-RP11-422N16.3NegativelymiR-23b-3pNegativelyZEB1↑ ZEB1[62]
HCCLncRNA-SNHG6NegativelymiR-101-3pNegativelyZEB1↑ ZEB1[63]
HCC--miR-139-5pNegativelyZEB1↓ ZEB1[64]
HCCLncRNA-TUG1NegativelymiR-142-3pNegativelyZEB1↑ ZEB1[65]
HCCLnc-MALAT1NegativelymiR-143-3pNegativelyZEB1↑ ZEB1[66]
HCCcirc-100338NegativelymiR-143-3pNegativelyZEB1↑ ZEB1[67]
HCCELF3NegativelymiR-141-3pNegativelyZEB1↑ ZEB1[68]
HCCATRAPositivelymiR-141-3p
miR-200a-3p
miR-200c-3p
NegativelyZEB1↓ ZEB1[69]
HCCLncRNA-ZFAS1NegativelymiR-150NegativelyZEB1↑ ZEB1[70]
HCCTGF-βPositivelymiR-155PositivelyZEB1↑ ZEB1[71]
HCCLncRNE-PENegativelymiR-200a
miR-200b
NegativelyZEB1↑ ZEB1[72]
HCCGα12NegativelymiR-200a
miR-200b
NegativelyZEB1↑ ZEB1[73]
CCALnc-ZEB1-AS1NegativelymiR-200aNegativelyZEB1↑ ZEB1[74]
HCCLnc-HULCNegativelymiR-200a-3pNegativelyZEB1↑ ZEB1[75]
HCCGenomic deletion, promoter methylationNegativelymiR-200bNegativelyZEB1↑ ZEB1[76]
HCCLnc-CARLo-5NegativelymiR-200bNegativelyZEB1↑ ZEB1[77]
HCCKLF4PositivelymiR-200bNegativelyZEB1↓ ZEB1[78]
HCCYB1NegativelymiR-200b
miR-205
NegativelyZEB1↑ ZEB1[79]
HCCLncRNA-XISTNegativelymiR-200b-3pNegativelyZEB1↑ ZEB1[80]
CCA--miR-200cNegativelyZEB1↓ ZEB1[81]
CCALnc-ATBNegativelymiR-200cNegativelyZEB1↑ ZEB1[82]
HCCp53PositivelymiR-200NegativelyZEB1↓ ZEB1[54]
HCCLncRNA-ATBNegativelymiR-200NegativelyZEB1↑ ZEB1[53]
HCCLINC00273NegativelymiR-200NegativelyZEB1↑ ZEB1[83]
CCALnc-NNT-AS1NegativelymiR-203NegativelyZEB1↑ ZEB1[84]
HCCLnc-SNHG3NegativelymiR-326NegativelyZEB1↑ ZEB1[85]
HCC--miR-369NegativelyZEB1↓ ZEB1[86]
HCCLncRNA-PRNCR1NegativelymiR-411-3pNegativelyZEB1↑ ZEB1[87]
HCCLnc-MAPKAPK5NegativelymiR-429NegativelyZEB1↑ ZEB1[88]
CCADMYPositivelymiR-455-3pNegativelyZEB1↓ ZEB1[89]
HCC--miR-590-3pNegativelyZEB1↓ ZEB1[90]
HCC--miR-708NegativelyZEB1↓ ZEB1[91]
HCCLnc171NegativelymiR-873-5pNegativelyZEB1↑ ZEB1[43]
HCCLnc-HCCL5--PositivelyZEB1↑ ZEB1[92]
HCCLncPNUTS--PositivelyZEB1↑ ZEB1[93]
ATRA, all-trans retinoic acid; CCA, cholangiocarcinoma; DMY, dihydromyricetin; ELF3, E74-like ETS transcription factor 3; HCC, hepatocellular carcinoma; KLF4, Kruppel-like factor 4; TGF-β, transforming growth factor β; YB1, Y-box binding protein 1. ↑ indicates increased ZEB1 expression and ↓ indicates ZEB1 reduced expression.
In addition to the above-described regulatory networks controlling ZEB1, the expression of other proteins has been linked to modified expression of ZEB1 without giving precise specifications of the regulatory mechanism. For example, increased expression of HMGB2, 14-3-3ε, POU class 2 homeobox 1 (POU2F1), fibrinogen gamma chain (FGG), RNA-binding motif, Y chromosome (RBMY), stage-specific embryonic antigen 3 (SSEA3), chromobox 6 (CBX6) has been shown to increase EMT, invasiveness and stemness of HCC cells through increasing ZEB1 expression [94,95,96,97,98,99,100], while for F-actin-capping protein subunit alpha-1 (CAPZA1), transient receptor potential cation channel subfamily V member 1 (TRPV1) and liver kinase B1 (LKB1), high expression correlates with low ZEB1 expression and reduced aggressivity of HCC cells [101,102,103]. Similarly, for adrenomedullin (ADM), cullin 4A (CUL4A) and epidermal growth factor (EGF), high expression correlates with high expression of ZEB1 and induced EMT and metastasis in CCA [104,105,106].

4. Role of ZEB1 in Hepatocellular Carcinoma (HCC)

Clinical evaluation showed that ZEB1 is highly expressed in malignant HCC cells compared to normal hepatocytes in 23–65.4% of the patients (Table 2) [91,107,108,109,110]. As an EMT-TF, ZEB1 expression correlated with reduced E-cadherin expression [13,107] and increased Vimentin expression [109]. Furthermore, ZEB1 expression was associated with aggressive clinical parameters, including poorly differentiated tumors, advanced TNM stage, increased number of tumors, multifocal metastases, vascular invasion (Table 3). All of these factors contributed to reduced progression-free survival and overall survival in patients with high ZEB1 expression (Table 2) [91,107,108,109,110].
Preclinical in vitro studies showed that ZEB1 can promote the proliferation, colony formation, sphere formation and migration of HCC cells and markedly enhance tumor progression in vivo in diethylnitrosamine (DEN)-induced models and orthotopic xenografts of human cell lines [91,109,110,112,113]. Mechanistically, multiple factors can be regulated by ZEB1 to lead to these aggressive features, which are indicated in Figure 4. As in patient samples, in HCC cell lines, ZEB1 is able to downregulate the expression of E-cadherin and increase the expression of mesenchymal markers such as Vimentin, matrix metallopeptidase 2 (MMP2) and MMP9, contributing to the migration and invasion of these cells [91,109,110]. ZEB1 is also able to activate the Wnt/β-catenin signaling pathway by upregulating the protein expression levels of β-catenin and its downstream targets c-Myc and Cyclin D1 [91]. Interestingly, ZEB1 has been shown to interact with Yes-associated protein (YAP) to become a transcriptional activator in some aggressive cancers [114]. However, in HCC cells, ZEB1 is able to directly increase the expression of YAP and activate YAP downstream signaling with the upregulation of genes like AXL receptor tyrosine kinase (AXL), connective tissue growth factor (CTGF) and serum deprivation response (SDPR), which contribute to the malignant phenotype [102]. Another transcriptional target of ZEB1 is DEAD-Box 56 (DDX56). Once ZEB1 increases the expression of DDX56, it can exert its downstream actions by upregulating the expression of muscle, intestine and stomach expression 1 (MIST1), which in turn negatively regulates Phosphatase and Tensin Homolog (PTEN) to activate AKT signaling, leading to HCC cell proliferation [115]. Besides classical targets, ZEB1 can control the transcription of ncRNAs. For example, ZEB1 activates the expression of the hsa-microRNA-99b/let-7e/microRNA-125a cluster in the HCC cells to promote invasion and advance liver cancer progression [116].
ZEB1 can also influence the metabolism of HCC cells by upregulating the expression of the muscle isoform of PFKM (phosphofructokinase), a rate-limiting enzyme in glycolysis, which stimulates glycolysis and leads to increased malignant phenotypes through promotion of the Warburg effect [113], a well-known mechanism used by cells undergoing EMT to fuel metabolic needs [117,118]. Similarly, ZEB1 activates the transcription of phosphoglycerate dehydrogenase (PHGDH), which in turn augments the serine synthesis pathway to render the cells more proliferative and invasive and resistant to sorafenib [112]. Activation of the serine biosynthetic pathway is involved in the induction of stemness and chemoresistance across tumor types [119,120]. In HCC cells, PHGDH promotes the translation of mitochondrial DNA-encoded proteins and sustains respiration [121], which helps with meeting the metabolic needs of these cells. This metabolic rewiring has been successfully targeted in other cancers but remains unexplored in HCC [119,120]. Curiously, both PFKM and PHGDH are regulated through non-classic ZEB1 binding sites within their promoter regions [112,113].
Regarding chemoresistance, ZEB1 expression has been shown to increase in HCC cells resistant to oxaliplatin, doxorubicin, sorafenib and bortezomib. These cells are usually enriched in the CD44high/CD24low stem cell population and show augmented expression of Protein kinase C alpha (PKCA) [122]. ZEB1-induced EMT has been linked to resistance to sorafenib [21,46,112], lenvatinib [52] and bortezomib [123], but the exact ZEB1-dependent downstream mechanism has not yet been delineated in these studies.
ZEB1 has also been implicated in the interactions between HCC cells and the TME. ZEB1 interacts with USP22 to coactivate transcriptionally vascular endothelial growth factor A (VEGFA). Then, VEGFA is able to act in a paracrine way to induce cell proliferation, migration and vascular mimicry formation in HCC cells and angiogenesis by acting on endothelial cells [58]. ZEB1 is not only expressed in HCC cells, but also in the TME. Indeed, miR-200b-3p can be encapsulated in exosomes that are taken by macrophages from the TME. In these cells, miR-200b-3p reduces ZEB1 expression, which negatively regulates interleukin 4 (IL4). Thus, increased IL4 levels promote a shift to M2 macrophage phenotype, with increased Pim-1 proto-oncogene, serine/threonine kinase (PIM1) and VEGFA production by the macrophages that act on HCC cells to promote proliferation and metastasis [124].

5. Role of ZEB1 in Cholangiocarcinoma (CCA)

Regarding clinical analysis, nuclear ZEB1 expression has been shown to be highly expressed in malignant intrahepatic CCA cells compared to normal non-neoplastic cells in 20–46.1% of the cases (Table 2) [14,111]. According to its role in EMT, ZEB1 expression showed a significant inverse correlation with E-cadherin expression and a positive correlation with vimentin expression [111]. Interestingly, ZEB1 expression has been associated with programmed cell death ligand 1 (PD-L1) expression in CCA [125]. Although this association has been demonstrated in other tumors [126], there is no mechanistic validation yet in CCA. Importantly, ZEB1 expression was associated with aggressive tumor characteristics, including advanced tumor stage, undifferentiated-type histology, lymph node metastasis and vascular invasion (Table 3) [14,111,127]. Furthermore, patients with high ZEB1 expression showed significantly reduced overall survival rates (Table 2) [111,127].
In vitro, ZEB1 has been shown to promote the proliferation, colony formation, sphere formation and migration of CCA cells and markedly enhance tumor progression in vivo [14,127]. ZEB1 can promote these features by several mechanisms, summarized in Figure 5. On one side, ZEB1 promotes cell migration through the activation of EMT, which leads to downregulation of epithelial markers, such as E-cadherin, and upregulation of mesenchymal markers, such as vimentin [14]. Among the molecular mechanisms related to ZEB1-induced EMT, the feedback loop with miR-200 has been identified in CCA cells [128]. On the other side, ZEB1 increases the stemness of CCA cells by altering the balance of the stem cell markers [14]. ZEB1 upregulates the expression of CD44 [14] (and its alternative splicing) [127], while it downregulates CD24 expression [14], generating a population of cells with high stemness [129,130]. Accordingly, subcutaneous implantation of CCA cells with ZEB1 overexpression produced bigger tumors in nude mice [14]. Curiously, tumors formed by ZEB1 overexpressing cells showed a more abundant stroma than their counterparts. The underlying mechanism involved the direct regulation by ZEB1 of CTGF in CCA cells, which in turn activated CAF proliferation, which could be impaired by a neutralizing antibody [14]. Interestingly, ZEB1 expression has also been detected in CAF from CCA and in different hepatic stellate cell lines. Downregulation of ZEB1 in these cells produced a downregulation of typical activation markers, such as Actin alpha 2, smooth muscle (ACTA2) and Collagen 4A1 (COL4A1). ZEB1 expression in the fibroblasts also impacted CCA cell growth [14]. Indeed, conditioned media from fibroblasts with high ZEB1 expression induced CCA cell growth and activation of the intracellular signaling pathways, including the signal transducer and activator of transcription 3 (STAT3), AKT and ERK. Further analysis showed that ZEB1 upregulated the expression of hepatocyte growth factor (HGF) in liver fibroblasts that induced proliferation of both CCA cells and fibroblasts [14]. These data underscore the role of ZEB1 in regulating tumor cell-CAF crosstalk as promoting tumor dedifferentiation and CAF activation, resulting in tumor progression.

6. Future Perspectives and Therapeutic Opportunities

Liver cancer is one of the leading causes of cancer-related mortality [3], mainly due to the fact that at diagnosis, most patients already present advanced stages of the disease and distant metastasis [1,2]. Consequently, understanding the process of EMT, a key driver of the metastatic cascade, is essential to tackle this important issue. In this context, an increasing number of publications is showing that ZEB1 expression is increased in both HCC and CCA, and it promotes the aggressiveness of these tumors by regulating multiple biological processes related to cell plasticity, including not only EMT, but stemness, therapy resistance and several aspects of the TME development. Thus, understanding the complex regulatory network supervised by ZEB1 may have important clinical and therapeutic implications.
The main issue with exploiting ZEB1’s therapeutic potential is that ZEB1-specific inhibitors are still to be developed. Nevertheless, some indirect approximations have been taken in liver cancer preclinical studies to inhibit ZEB1-related functions (Table 4).

6.1. Chemical Compounds

Farrerol, a bioactive constituent of Rhododendron, is able to suppress TGF-β-mediated migration and invasiveness in HCC cells by inhibiting SMAD2/3 phosphorylation (Figure 2). This impaired phosphorylation led to downregulation of different EMT regulators, including ZEB1 [131]. Similarly, cinobufotalin, a bufadienolide isolated from toad venom, downregulates β-catenin-dependent EMT, which is in part mediated by ZEB1 (Figure 2) [132]. Another report shows that salinomycin, an antibacterial and coccidiostat ionophore, activates FOXO3a, which acts as a negative regulator of the β-catenin, with similar results, reducing ZEB1 expression and EMT in HCC cells (Figure 2). Interestingly, this mechanism mediated by salinomycin is able to revert resistance to doxorubicin in these cells [28]. In CCA, dihydromyricetin (DMY), the main bioactive compound isolated from Ampelopsis grossedentata, significantly reduces proliferation and EMT in tumor cells, through a mechanism involving miR-455-3p upregulation, which in turn targets and inhibits ZEB1 expression [89]. Also in CCA, maraviroc, a CCR5 inhibitor, has been used to impair CCL5-regulated functions upstream of ZEB1 [133]. Additionally, post-translational modifications such as O-GlcNAcylation can be exploited to impair ZEB1 functions. Artesunate, a key antimalarial drug, which originated from the traditional Chinese medicinal plant Artemisia annua, promotes OGA function, which is in charge of removing the O-linked N-acetylglucosamine modification at ser 670, reducing ZEB1 protein stability (Figure 3) [56]. Besides targeting ZEB1 expression, PKC inhibitors have been used to inhibit ZEB1-regulated downstream functions that are mediated by PKCA in HCC cells [122]. The main problem of these approaches is that they do not specifically target ZEB1.

6.2. Neutralizing Antibodies

Neutralizing antibodies against soluble signaling molecules is another regulatory mechanism that can be exploited against ZEB1. For example, neutralizing antibodies against CCL5, produced by MSC or CAF, have been used to inhibit CCL5-regulated processes, including ZEB1 expression [133].

6.3. miRNAs

The potential of miRNAs has been assayed in HCC in a study where miR-708 was used therapeutically to target and downregulate ZEB1 expression [91]. The most accurate therapeutic option available is probably miRNA-based therapy targeting ZEB1 expression. Nevertheless, it is necessary to ensure the specificity of the miRNAs for ZEB1 and to further develop the delivery methods and the stability of the therapeutic agents.

7. Conclusions

In conclusion, ZEB1 is a key regulator of transcriptional plasticity in liver cancer, allowing the cells to adapt to challenges from environmental and therapeutic pressures through the activation of EMT, stemness and chemoresistance programs, which contribute to the aggressivity of the tumors and poorer prognosis of liver cancer patients. Future research should aim to untangle the complexity of the ZEB1-regulated network to identify potential new therapeutic targets that could be integrated with current therapies to improve the evolving treatment landscape of liver cancer patients.

Author Contributions

Conceptualization, J.V., validation, all authors; resources, all authors; writing—original draft preparation, all authors; writing—review and editing, all authors; funding acquisition, J.V. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Agencia Estatal de Investigación (AEI), Ministry of Science and Innovation (MICIN), Spain, cofounded by FEDER funds/Development Fund—a way to build Europe, grant numbers PID2022-141984OB-I00 and RYC2021-034121-I, and by Fundación Memoria de D. Samuel Solorzano Barruso, grant number FS/14-2023, to J.V. This work was also funded by «Escalera de Excelencia» of the Education Ministry of the Castilla y León autonomous government, plus the European Regional Development Fund (CLU-2023-2-01). J.V. was funded by AEI and MICIN, through the “Ramon y Cajal” program RYC2021-034121-I. C.A.R.-H. was funded by CSIC through a JAE-PRE fellowship (JAEPR23102). L.G.-C. was funded by AEI, MICIN through the PID2022-141984OB-I00 grant. The CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, is funded by the Instituto de Salud Carlos III, Spain. This article is based upon work from the COST Action CA22125 Precision medicine in biliary tract cancer (Precision-BTC-Network) supported by COST (European Cooperation in Science and Technology: www.cost.eu), in collaboration with the European Network for the Study of Cholangiocarcinoma (ENS-CCA: https://precision-btc.eu/enscca).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ABCA8ATP binding cassette subfamily A member 8
ACTA2Actin alpha 2, smooth muscle
ADMAdrenomedullin
AKTAKT serine/threonine kinase
AP-2γActivator Protein-2 gamma
AXLAXL receptor tyrosine kinase
CAFCancer-associated fibroblasts
CAPZA1F-actin-capping protein subunit alpha-1
CBDP300-P/CBP-associated factor (P/CAF) binding domain
CBX6Chromobox 6
CCACholangiocarcinoma
CCL5C-C motif chemokine ligand 5
CCR5C-C motif chemokine ligand 5 Receptor
CIDCtBP interaction domain
circRNACircular RNA
COL4A1Collagen 4A1
CTGFConnective tissue growth factor
CUL4ACullin 4A
DDX56DEAD-Box 56
DENDiethylnitrosamine
DMYDihydromyricetin
EGFEpidermal growth factor
EGR1Early growth response 1
eEF-1γEukaryotic translation elongation factor 1 gamma
EMTEpithelial–mesenchymal Transition
EMT-TFEMT-inducing transcription factor
ERKExtracellular signal-regulated kinase
FBXW7F-Box and WD repeat domain containing 7
FBXW8F-box and WD repeat domain containing 8
FGFFibroblast growth factor
FGGFibrinogen gamma chain
FOXO3aForkhead box O3
GINS1GINS complex subunit 1
GSK3βGlycogen synthase kinase 3 beta
HCCHepatocellular carcinoma
HDACHistone deacetylase
HGFHepatocyte growth factor
HIF1αHypoxia-inducible factor 1 alpha
HIPK2Homeodomain Interacting Protein Kinase 2
HMGB1High mobility group box 1
HMGB2High mobility group box 1
HN1LHematological and neurological expressed 1-like
IGFBP2Insulin like growth factor binding protein 2
IL4Interleukin 4
LKB1Liver kinase B1
lncRNAlong non-coding RNA
m6AN6-methyladenosine
MAPKMitogen-activated protein kinase
MAZMYC associated zinc finger protein
METTL3Methyltransferase-like protein 3
METTL13Methyltransferase 13
miRNAMicroRNA
MISTMuscle, intestine and stomach expression 1
MMP2Matrix metallopeptidase 2
MSCMesenchymal stem cells
mTORMechanistic target of rapamycin kinase
ncRNAsNon-coding RNAs
NCSTNNicastrin
NFκBNuclear factor kappa B
NONONon-POU domain containing octamer binding
OTUB2OTU deubiquitinase, ubiquitin aldehyde binding 2
PDHA-1Pyruvate dehydrogenase E1 subunit alpha 1
PFKMPhosphofructokinase-1, muscle
PHGDHPhosphoglycerate dehydrogenase
PIM1Pim-1 proto-oncogene, serine/threonine kinase
PKCAProtein kinase C alpha
PKM2Pyruvate kinase M2
POU2F1POU class 2 homeobox 1
PPM1GProtein phosphatase 1G
PPT1Palmitoyl-Protein Thioesterase 1
PTENPhosphatase and tensin homolog
PTK7Tyrosine-protein kinase-like 7
PTP4A1Protein tyrosine phosphatase type 4A1
RBMYRNA-binding motif, Y chromosome
SDPRSerum deprivation response
SETSuppressor of variegation, enhancer of zeste, trithorax
SIDSmad interaction domain
SMYD3MYND (Myeloid-Nervy-DEAF1) domain containing 3
SNAI1/2Snail family transcriptional repressor 1/2
SORT1Sortilin 1
SP1Specificity protein 1
SSEA3Stage-specific embryonic antigen 3
STAT3Signal Transducer and activator of transcription 3
TBL1XTransducin beta like 1 X-linked
TCFT-cell factor
TFTranscription factor
TGF-βTransforming growth factor-beta
THRSPThyroid Hormone Responsive Protein
TMETumor microenvironment
TPAO-tetradecanoyl-phorbol 13-acetate
TRAF6TNF receptor associated factor 6
TRIM26Tripartite motif containing 26
TRPV1Transient receptor potential cation channel subfamily V member 1
TWIST1/2Twist family bHLH transcription factor 1/2
TXNDC12Thioredoxin domain containing 12
UPATUbiquitin-like plant homeodomain (PHD) and really interesting new gene (RING) finger domain-containing protein 1 (UHRF1) protein associated transcript
USP22Ubiquitin specific peptidase 22
USP39Ubiquitin specific peptidase 39
VEGFAVascular endothelial growth factor A
WntWingless/integrated
YAPYes-associated protein
YTHDF3YTH domain-containing family protein 3
ZEB1/2Zinc finger E-box-binding homeobox 1/2
ZIP4Zrt- and irt-like protein 4

References

  1. Yang, C.; Zhang, H.; Zhang, L.; Zhu, A.X.; Bernards, R.; Qin, W.; Wang, C. Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 203–222. [Google Scholar] [CrossRef]
  2. Banales, J.M.; Cardinale, V.; Carpino, G.; Marzioni, M.; Andersen, J.B.; Invernizzi, P.; Lind, G.E.; Folseraas, T.; Forbes, S.J.; Fouassier, L.; et al. Expert consensus document: Cholangiocarcinoma: Current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 261–280. [Google Scholar] [CrossRef] [PubMed]
  3. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  4. Huang, Y.; Hong, W.; Wei, X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J. Hematol. Oncol. 2022, 15, 129. [Google Scholar] [CrossRef]
  5. Vaquero, J.; Guedj, N.; Claperon, A.; Nguyen Ho-Bouldoires, T.H.; Paradis, V.; Fouassier, L. Epithelial-mesenchymal transition in cholangiocarcinoma: From clinical evidence to regulatory networks. J. Hepatol. 2017, 66, 424–441. [Google Scholar] [CrossRef]
  6. Derynck, R.; Weinberg, R.A. EMT and Cancer: More Than Meets the Eye. Dev. Cell 2019, 49, 313–316. [Google Scholar] [CrossRef]
  7. Roy, S.; Sunkara, R.R.; Parmar, M.Y.; Shaikh, S.; Waghmare, S.K. EMT imparts cancer stemness and plasticity: New perspectives and therapeutic potential. Front. Biosci. (Landmark Ed.) 2021, 26, 238–265. [Google Scholar] [CrossRef] [PubMed]
  8. Tanabe, S.; Quader, S.; Cabral, H.; Ono, R. Interplay of EMT and CSC in Cancer and the Potential Therapeutic Strategies. Front. Pharmacol. 2020, 11, 904. [Google Scholar] [CrossRef]
  9. Parfenyev, S.E.; Daks, A.A.; Shuvalov, O.Y.; Fedorova, O.A.; Pestov, N.B.; Korneenko, T.V.; Barlev, N.A. Dualistic role of ZEB1 and ZEB2 in tumor progression. Biol. Direct 2025, 20, 32. [Google Scholar] [CrossRef] [PubMed]
  10. Postigo, A.A. Opposing functions of ZEB proteins in the regulation of the TGFbeta/BMP signaling pathway. EMBO J. 2003, 22, 2443–2452. [Google Scholar] [CrossRef]
  11. Soen, B.; Vandamme, N.; Berx, G.; Schwaller, J.; Van Vlierberghe, P.; Goossens, S. ZEB Proteins in Leukemia: Friends, Foes, or Friendly Foes? Hemasphere 2018, 2, e43. [Google Scholar] [CrossRef] [PubMed]
  12. Caramel, J.; Ligier, M.; Puisieux, A. Pleiotropic Roles for ZEB1 in Cancer. Cancer Res. 2018, 78, 30–35. [Google Scholar] [CrossRef] [PubMed]
  13. Hashiguchi, M.; Ueno, S.; Sakoda, M.; Iino, S.; Hiwatashi, K.; Minami, K.; Ando, K.; Mataki, Y.; Maemura, K.; Shinchi, H.; et al. Clinical implication of ZEB-1 and E-cadherin expression in hepatocellular carcinoma (HCC). BMC Cancer 2013, 13, 572. [Google Scholar] [CrossRef]
  14. Lobe, C.; Vallette, M.; Arbelaiz, A.; Gonzalez-Sanchez, E.; Izquierdo, L.; Pellat, A.; Guedj, N.; Louis, C.; Paradis, V.; Banales, J.M.; et al. Zinc Finger E-Box Binding Homeobox 1 Promotes Cholangiocarcinoma Progression Through Tumor Dedifferentiation and Tumor-Stroma Paracrine Signaling. Hepatology 2021, 74, 3194–3212. [Google Scholar] [CrossRef]
  15. Williams, T.M.; Montoya, G.; Wu, Y.; Eddy, R.L.; Byers, M.G.; Shows, T.B. The TCF8 gene encoding a zinc finger protein (Nil-2-a) resides on human chromosome 10p11.2. Genomics 1992, 14, 194–196. [Google Scholar] [CrossRef]
  16. Remacle, J.E.; Kraft, H.; Lerchner, W.; Wuytens, G.; Collart, C.; Verschueren, K.; Smith, J.C.; Huylebroeck, D. New mode of DNA binding of multi-zinc finger transcription factors: DeltaEF1 family members bind with two hands to two target sites. EMBO J. 1999, 18, 5073–5084. [Google Scholar] [CrossRef]
  17. Postigo, A.A.; Depp, J.L.; Taylor, J.J.; Kroll, K.L. Regulation of Smad signaling through a differential recruitment of coactivators and corepressors by ZEB proteins. EMBO J. 2003, 22, 2453–2462. [Google Scholar] [CrossRef]
  18. Shi, Y.; Sawada, J.; Sui, G.; Affar, E.B.; Whetstine, J.R.; Lan, F.; Ogawa, H.; Luke, M.P.; Nakatani, Y.; Shi, Y. Coordinated histone modifications mediated by a CtBP co-repressor complex. Nature 2003, 422, 735–738. [Google Scholar] [CrossRef]
  19. Roche, J.; Nasarre, P.; Gemmill, R.; Baldys, A.; Pontis, J.; Korch, C.; Guilhot, J.; Ait-Si-Ali, S.; Drabkin, H. Global Decrease of Histone H3K27 Acetylation in ZEB1-Induced Epithelial to Mesenchymal Transition in Lung Cancer Cells. Cancers 2013, 5, 334–356, Erratum in Cancers 2016, 8, 114. [Google Scholar] [CrossRef] [PubMed]
  20. Zhang, J.; Zhou, C.; Jiang, H.; Liang, L.; Shi, W.; Zhang, Q.; Sun, P.; Xiang, R.; Wang, Y.; Yang, S. ZEB1 induces ER-alpha promoter hypermethylation and confers antiestrogen resistance in breast cancer. Cell Death Dis. 2017, 8, e2732. [Google Scholar] [CrossRef]
  21. Wang, S.; You, X.; Liu, X.; Fengwei, Z.; Zhou, H.; Shang, X.; Cai, L. SMYD3 induces sorafenib resistance by activating SMAD2/3-mediated epithelial-mesenchymal transition in hepatocellular carcinoma. iScience 2023, 26, 106994. [Google Scholar] [CrossRef]
  22. Wong, T.L.M.; Wong, T.L.; Zhou, L.; Man, K.; Purcell, J.; Lee, T.K.; Yun, J.P.; Ma, S. Protein Tyrosine Kinase 7 (PTK7) Promotes Metastasis in Hepatocellular Carcinoma via SOX9 Regulation and TGF-beta Signaling. Cell. Mol. Gastroenterol. Hepatol. 2023, 15, 13–37. [Google Scholar] [CrossRef]
  23. Guo, H.; Su, R.; Lu, X.; Zhang, H.; Wei, X.; Xu, X. ZIP4 inhibits Ephrin-B1 ubiquitination, activating Wnt5A/JNK/ZEB1 to promote liver cancer metastasis. Genes Dis. 2024, 11, 101312. [Google Scholar] [CrossRef]
  24. Yuan, K.; Xie, K.; Lan, T.; Xu, L.; Chen, X.; Li, X.; Liao, M.; Li, J.; Huang, J.; Zeng, Y.; et al. TXNDC12 promotes EMT and metastasis of hepatocellular carcinoma cells via activation of beta-catenin. Cell Death Differ. 2020, 27, 1355–1368. [Google Scholar] [CrossRef]
  25. Li, H.; Lan, T.; Xu, L.; Liu, H.; Wang, J.; Li, J.; Chen, X.; Huang, J.; Li, X.; Yuan, K.; et al. NCSTN promotes hepatocellular carcinoma cell growth and metastasis via beta-catenin activation in a Notch1/AKT dependent manner. J. Exp. Clin. Cancer Res. 2020, 39, 128, Correction in J. Exp. Clin. Cancer Res. 2023, 42, 47. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, H.; Liao, W.; Jiang, Y.; Liao, G.; Gao, X.; Cen, S.; Liu, L.; Peng, J.; Shaohang, C. SORT1 promote the metastasis and invasion of hepatocellular carcinoma via p38/beta-catenin/ZEB1 signaling pathway. Cell Death Dis. 2025, 16, 582. [Google Scholar] [CrossRef] [PubMed]
  27. Liang, J.; Yao, N.; Deng, B.; Li, J.; Jiang, Y.; Liu, T.; Hu, Y.; Cao, M.; Hong, J. GINS1 promotes ZEB1-mediated epithelial-mesenchymal transition and tumor metastasis via beta-catenin signaling in hepatocellular carcinoma. J. Cell Physiol. 2024, 239, e31237. [Google Scholar] [CrossRef] [PubMed]
  28. Zhou, Y.; Liang, C.; Xue, F.; Chen, W.; Zhi, X.; Feng, X.; Bai, X.; Liang, T. Salinomycin decreases doxorubicin resistance in hepatocellular carcinoma cells by inhibiting the beta-catenin/TCF complex association via FOXO3a activation. Oncotarget 2015, 6, 10350–10365. [Google Scholar] [CrossRef]
  29. Wang, J.; Dong, Y.; Wei, Z.; Zhang, Y.; Wu, N.; Zhang, C.; Zhang, Y.; Zi, R.; Hao, J.; Liang, H.; et al. Deubiquitinase OTUB2 promotes intrahepatic cholangiocarcinoma progression by stabilizing the CTNNB1-ZEB1 axis. Exp. Cell Res. 2023, 425, 113537. [Google Scholar] [CrossRef]
  30. Liu, L.Z.; He, Y.Z.; Dong, P.P.; Ma, L.J.; Wang, Z.C.; Liu, X.Y.; Duan, M.; Yang, L.X.; Shi, J.Y.; Zhou, J.; et al. Protein tyrosine phosphatase PTP4A1 promotes proliferation and epithelial-mesenchymal transition in intrahepatic cholangiocarcinoma via the PI3K/AKT pathway. Oncotarget 2016, 7, 75210–75220. [Google Scholar] [CrossRef]
  31. Cui, Y.; Liang, S.; Zhang, S.; Zhang, C.; Zhao, Y.; Wu, D.; Wang, J.; Song, R.; Wang, J.; Yin, D.; et al. ABCA8 is regulated by miR-374b-5p and inhibits proliferation and metastasis of hepatocellular carcinoma through the ERK/ZEB1 pathway. J. Exp. Clin. Cancer Res. 2020, 39, 90. [Google Scholar] [CrossRef]
  32. Hu, Q.; Ma, X.; Li, C.; Zhou, C.; Chen, J.; Gu, X. Downregulation of THRSP Promotes Hepatocellular Carcinoma Progression by Triggering ZEB1 Transcription in an ERK-dependent Manner. J. Cancer 2021, 12, 4247–4256. [Google Scholar] [CrossRef]
  33. Suh, Y.; Yoon, C.H.; Kim, R.K.; Lim, E.J.; Oh, Y.S.; Hwang, S.G.; An, S.; Yoon, G.; Gye, M.C.; Yi, J.M.; et al. Claudin-1 induces epithelial-mesenchymal transition through activation of the c-Abl-ERK signaling pathway in human liver cells. Oncogene 2013, 32, 4873–4882, Correction in Oncogene 2017, 36, 1167–1168. [Google Scholar] [CrossRef]
  34. Hossain, A.J.; Hamza, A.; Islam, R.; Dogsom, O.; Park, J.B. Function of eEF-1gamma in the nucleus in response to insulin in hepatocellular carcinoma cells. Commun. Biol. 2025, 8, 826. [Google Scholar] [CrossRef]
  35. Guo, Q.; Yu, D.Y.; Yang, Z.F.; Liu, D.Y.; Cao, H.Q.; Liao, X.W. IGFBP2 upregulates ZEB1 expression and promotes hepatocellular carcinoma progression through NF-kappaB signaling pathway. Dig. Liver Dis. 2020, 52, 573–581. [Google Scholar] [CrossRef]
  36. Li, L.; Zheng, Y.L.; Jiang, C.; Fang, S.; Zeng, T.T.; Zhu, Y.H.; Li, Y.; Xie, D.; Guan, X.Y. HN1L-mediated transcriptional axis AP-2gamma/METTL13/TCF3-ZEB1 drives tumor growth and metastasis in hepatocellular carcinoma. Cell Death Differ. 2019, 26, 2268–2283. [Google Scholar] [CrossRef] [PubMed]
  37. Hu, L.; Shi, X.; Yuan, X.; Liu, D.; Zheng, D.; Li, Y.; Shi, F.; Zhang, M.; Su, S.G.; Zhang, C.Z. PPM1G-mediated TBL1X mRNA splicing promotes cell migration in hepatocellular carcinoma. Cancer Sci. 2025, 116, 67–80. [Google Scholar] [CrossRef] [PubMed]
  38. Wu, W.S.; You, R.I.; Cheng, C.C.; Lee, M.C.; Lin, T.Y.; Hu, C.T. Snail collaborates with EGR-1 and SP-1 to directly activate transcription of MMP 9 and ZEB1. Sci. Rep. 2017, 7, 17753, Correction in Sci. Rep. 2018, 8, 14226. [Google Scholar] [CrossRef] [PubMed]
  39. He, J.; Zou, X.; Wang, D.; Zhou, L.; Li, X.; Ma, S.; Shen, X.; Liu, S.; Liu, B. FBXW8-mediated degradation of PPT1 suppresses epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma. Biochim. Biophys. Acta Mol. Basis Dis. 2025, 1871, 167985. [Google Scholar] [CrossRef]
  40. Yang, H.; Lu, X.; Liu, Z.; Chen, L.; Xu, Y.; Wang, Y.; Wei, G.; Chen, Y. FBXW7 suppresses epithelial-mesenchymal transition, stemness and metastatic potential of cholangiocarcinoma cells. Oncotarget 2015, 6, 6310–6325. [Google Scholar] [CrossRef]
  41. Xu, H.; Zhao, J.; Li, J.; Zhu, Z.; Cui, Z.; Liu, R.; Lu, R.; Yao, Z.; Xu, Q. Cancer associated fibroblast-derived CCL5 promotes hepatocellular carcinoma metastasis through activating HIF1alpha/ZEB1 axis. Cell Death Dis. 2022, 13, 478. [Google Scholar] [CrossRef]
  42. Chahine, J.J.; Davis, S.S.; Culfaci, S.; Kallakury, B.V.; Tuma, P.L. Chromosome 8q24 amplification associated with human hepatocellular carcinoma predicts MYC/ZEB1/MIZ1 transcriptional regulation. Sci. Rep. 2024, 14, 24488. [Google Scholar] [CrossRef]
  43. Huang, S.; Liao, Z.; He, X.; Song, Z.; Fang, X.; Wen, S.; Yang, L.; Li, H.; Zhang, Q.; Mo, W.; et al. Ethanol responsive lnc171 promotes migration and invasion of HCC cells via mir-873-5p/ZEB1 axis. BMC Cancer 2024, 24, 550. [Google Scholar] [CrossRef] [PubMed]
  44. Kwapisz, O.; Gorka, J.; Korlatowicz, A.; Kotlinowski, J.; Waligorska, A.; Marona, P.; Pydyn, N.; Dobrucki, J.W.; Jura, J.; Miekus, K. Fatty Acids and a High-Fat Diet Induce Epithelial-Mesenchymal Transition by Activating TGFbeta and beta-Catenin in Liver Cells. Int. J. Mol. Sci. 2021, 22, 1272. [Google Scholar] [CrossRef]
  45. Yu, W.; Ma, Y.; Shrivastava, S.K.; Srivastava, R.K.; Shankar, S. Chronic alcohol exposure induces hepatocyte damage by inducing oxidative stress, SATB2 and stem cell-like characteristics, and activating lipogenesis. J. Cell Mol. Med. 2022, 26, 2119–2131. [Google Scholar] [CrossRef]
  46. Shih, M.S.; Suk, F.M.; Chiu, W.C.; Lee, C.Y.; Hsu, F.Y.; Liao, Y.J. Long-term di-(2-ethylhexyl) phthalate exposure reduces sorafenib treatment efficacy by enhancing mesenchymal transition in hepatocellular carcinoma. Ecotoxicol. Environ. Saf. 2024, 273, 116161. [Google Scholar] [CrossRef]
  47. Li, D.; Xu, L.; Liu, R.; Yao, Z.; Zheng, C.; Jin, S.; Guo, X.; Zhang, Z.; Tan, S.; Zhu, X. MAZ-mediated N6-methyladenosine modification of ZEB1 promotes hepatocellular carcinoma progression by regulating METTL3. J. Transl. Med. 2025, 23, 265. [Google Scholar] [CrossRef] [PubMed]
  48. Luo, W.; Zhu, X.; Liu, W.; Ren, Y.; Bei, C.; Qin, L.; Miao, X.; Tang, F.; Tang, G.; Tan, S. MYC associated zinc finger protein promotes the invasion and metastasis of hepatocellular carcinoma by inducing epithelial mesenchymal transition. Oncotarget 2016, 7, 86420–86432. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, M.; Yang, Y.; Yang, J.; Yang, J.; Han, S. circ_KIAA1429 accelerates hepatocellular carcinoma advancement through the mechanism of m(6)A-YTHDF3-Zeb1. Life Sci. 2020, 257, 118082. [Google Scholar] [CrossRef]
  50. Gorecki, I.; Rak, B. The role of microRNAs in epithelial to mesenchymal transition and cancers; focusing on mir-200 family. Cancer Treat. Res. Commun. 2021, 28, 100385. [Google Scholar] [CrossRef]
  51. Hou, Y.R.; Diao, L.T.; Hu, Y.X.; Zhang, Q.Q.; Lv, G.; Tao, S.; Xu, W.Y.; Xie, S.J.; Zhang, Q.; Xiao, Z.D. The Conserved LncRNA DIO3OS Restricts Hepatocellular Carcinoma Stemness by Interfering with NONO-Mediated Nuclear Export of ZEB1 mRNA. Adv. Sci. 2023, 10, e2301983. [Google Scholar] [CrossRef]
  52. Cao, M.; Li, Y.; Su, X.; Tang, Y.; Yuan, F.; Ren, Y.; Deng, M.; Yao, Z. Exosome-derived hsa_circ_0007132 promotes lenvatinib resistance by inhibiting the ubiquitin-mediated degradation of NONO. Noncoding RNA Res. 2025, 14, 1–13. [Google Scholar] [CrossRef]
  53. Yuan, J.H.; Yang, F.; Wang, F.; Ma, J.Z.; Guo, Y.J.; Tao, Q.F.; Liu, F.; Pan, W.; Wang, T.T.; Zhou, C.C.; et al. A long noncoding RNA activated by TGF-beta promotes the invasion-metastasis cascade in hepatocellular carcinoma. Cancer Cell 2014, 25, 666–681. [Google Scholar] [CrossRef] [PubMed]
  54. Kim, T.; Veronese, A.; Pichiorri, F.; Lee, T.J.; Jeon, Y.J.; Volinia, S.; Pineau, P.; Marchio, A.; Palatini, J.; Suh, S.S.; et al. p53 regulates epithelial-mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J. Exp. Med. 2011, 208, 875–883. [Google Scholar] [CrossRef] [PubMed]
  55. Park, M.K.; Lee, H.; Lee, C.H. Post-Translational Modification of ZEB Family Members in Cancer Progression. Int. J. Mol. Sci. 2022, 23, 15127. [Google Scholar] [CrossRef] [PubMed]
  56. Li, Y.; Zhang, Y.; Lu, W.; Chen, Y.; Qian, X. Artesunate inhibits hepatocellular carcinoma cell migration and invasion through OGA-mediated O-GlcNAcylation of ZEB1. Open Life Sci. 2025, 20, 20251109. [Google Scholar] [CrossRef]
  57. Li, X.; Yuan, J.; Song, C.; Lei, Y.; Xu, J.; Zhang, G.; Wang, W.; Song, G. Deubiquitinase USP39 and E3 ligase TRIM26 balance the level of ZEB1 ubiquitination and thereby determine the progression of hepatocellular carcinoma. Cell Death Differ. 2021, 28, 2315–2332. [Google Scholar] [CrossRef]
  58. Zeng, K.; Xie, W.; Wang, C.; Wang, S.; Liu, W.; Su, Y.; Lin, L.; Zou, R.; Sun, G.; Zhou, B.; et al. USP22 upregulates ZEB1-mediated VEGFA transcription in hepatocellular carcinoma. Cell Death Dis. 2023, 14, 194. [Google Scholar] [CrossRef]
  59. Dong, X.; Han, Y.; Zhang, E.; Wang, Y.; Zhang, P.; Wang, C.; Zhong, L.; Li, Q. Tumor suppressor DCAF15 inhibits epithelial-mesenchymal transition by targeting ZEB1 for proteasomal degradation in hepatocellular carcinoma. Aging 2021, 13, 10603–10618. [Google Scholar] [CrossRef]
  60. Xiang, L.; Huang, X.; Wang, S.; Ou, H.; Chen, Z.; Hu, Z.; Huang, Y.; Li, X.; Yuan, Y.; Yang, D. Deficiency of pseudogene UPAT leads to hepatocellular carcinoma progression and forms a positive feedback loop with ZEB1. Cancer Sci. 2020, 111, 4102–4117. [Google Scholar] [CrossRef]
  61. Zhu, W.; Li, J.; Zhang, Y.; Zhu, Z.; Liu, H.; Lin, Y.; Hu, A.; Zhou, J.; Ren, H.; Shi, X. Inhibition of HMGB1 Suppresses Hepatocellular Carcinoma Progression via HIPK2-Mediated Autophagic Degradation of ZEB1. Front. Oncol. 2021, 11, 599124. [Google Scholar] [CrossRef]
  62. Sun, Y.; Zhou, Q.; Li, J.; Zhao, C.; Yu, Z.; Zhu, Q. LncRNA RP11-422N16.3 Inhibits Cell Proliferation and EMT, and Induces Apoptosis in Hepatocellular Carcinoma Cells by Sponging miR-23b-3p. Onco Targets Ther. 2019, 12, 10943–10961. [Google Scholar] [CrossRef]
  63. Chang, L.; Yuan, Y.; Li, C.; Guo, T.; Qi, H.; Xiao, Y.; Dong, X.; Liu, Z.; Liu, Q. Upregulation of SNHG6 exhibits an oncogenic effect, regulating ZEB1 expression by competitively binding miR-101-3p and interacting with UPF1 in hepatocellular carcinoma. Cancer Lett. 2016, 383, 183–194, Corrigendum in Cancer Lett. 2023, 555, 216045. [Google Scholar] [CrossRef]
  64. Qiu, G.; Lin, Y.; Zhang, H.; Wu, D. miR-139-5p inhibits epithelial-mesenchymal transition, migration and invasion of hepatocellular carcinoma cells by targeting ZEB1 and ZEB2. Biochem. Biophys. Res. Commun. 2015, 463, 315–321. [Google Scholar] [CrossRef]
  65. He, C.; Liu, Z.; Jin, L.; Zhang, F.; Peng, X.; Xiao, Y.; Wang, X.; Lyu, Q.; Cai, X. lncRNA TUG1-Mediated Mir-142-3p Downregulation Contributes to Metastasis and the Epithelial-to-Mesenchymal Transition of Hepatocellular Carcinoma by Targeting ZEB1. Cell Physiol. Biochem. 2018, 48, 1928–1941. [Google Scholar] [CrossRef]
  66. Chen, L.; Yao, H.; Wang, K.; Liu, X. Long Non-Coding RNA MALAT1 Regulates ZEB1 Expression by Sponging miR-143-3p and Promotes Hepatocellular Carcinoma Progression. J. Cell Biochem. 2017, 118, 4836–4843. [Google Scholar] [CrossRef]
  67. Cheng, X.; Tian, P.; Zheng, W.; Yan, X. Piplartine attenuates the proliferation of hepatocellular carcinoma cells via regulating hsa_circ_100338 expression. Cancer Med. 2020, 9, 4265–4273. [Google Scholar] [CrossRef] [PubMed]
  68. Zheng, L.; Xu, M.; Xu, J.; Wu, K.; Fang, Q.; Liang, Y.; Zhou, S.; Cen, D.; Ji, L.; Han, W.; et al. ELF3 promotes epithelial-mesenchymal transition by protecting ZEB1 from miR-141-3p-mediated silencing in hepatocellular carcinoma. Cell Death Dis. 2018, 9, 387. [Google Scholar] [CrossRef] [PubMed]
  69. Cui, J.; Gong, M.; Fang, S.; Hu, C.; Wang, Y.; Zhang, J.; Tang, N.; He, Y. All-trans retinoic acid reverses malignant biological behavior of hepatocarcinoma cells by regulating miR-200 family members. Genes Dis. 2021, 8, 509–520. [Google Scholar] [CrossRef] [PubMed]
  70. Li, T.; Xie, J.; Shen, C.; Cheng, D.; Shi, Y.; Wu, Z.; Deng, X.; Chen, H.; Shen, B.; Peng, C.; et al. Amplification of Long Noncoding RNA ZFAS1 Promotes Metastasis in Hepatocellular Carcinoma. Cancer Res. 2015, 75, 3181–3191. [Google Scholar] [CrossRef]
  71. Li, D.P.; Fan, J.; Wu, Y.J.; Xie, Y.F.; Zha, J.M.; Zhou, X.M. MiR-155 up-regulated by TGF-beta promotes epithelial-mesenchymal transition, invasion and metastasis of human hepatocellular carcinoma cells in vitro. Am. J. Transl. Res. 2017, 9, 2956–2965. [Google Scholar]
  72. Shen, Y.; Liu, S.; Yuan, H.; Ying, X.; Fu, H.; Zheng, X. A long non-coding RNA lncRNA-PE promotes invasion and epithelial-mesenchymal transition in hepatocellular carcinoma through the miR-200a/b-ZEB1 pathway. Tumour Biol. 2017, 39, 1010428317705756. [Google Scholar] [CrossRef] [PubMed]
  73. Yang, Y.M.; Lee, W.H.; Lee, C.G.; An, J.; Kim, E.S.; Kim, S.H.; Lee, S.K.; Lee, C.H.; Dhanasekaran, D.N.; Moon, A.; et al. Galpha12 gep oncogene deregulation of p53-responsive microRNAs promotes epithelial-mesenchymal transition of hepatocellular carcinoma. Oncogene 2015, 34, 2910–2921. [Google Scholar] [CrossRef]
  74. Jiao, M.; Ning, S.; Chen, J.; Chen, L.; Jiao, M.; Cui, Z.; Guo, L.; Mu, W.; Yang, H. Long non-coding RNA ZEB1-AS1 predicts a poor prognosis and promotes cancer progression through the miR-200a/ZEB1 signaling pathway in intrahepatic cholangiocarcinoma. Int. J. Oncol. 2020, 56, 1455–1467. [Google Scholar] [CrossRef]
  75. Li, S.P.; Xu, H.X.; Yu, Y.; He, J.D.; Wang, Z.; Xu, Y.J.; Wang, C.Y.; Zhang, H.M.; Zhang, R.X.; Zhang, J.J.; et al. LncRNA HULC enhances epithelial-mesenchymal transition to promote tumorigenesis and metastasis of hepatocellular carcinoma via the miR-200a-3p/ZEB1 signaling pathway. Oncotarget 2016, 7, 42431–42446. [Google Scholar] [CrossRef] [PubMed]
  76. Tsai, S.C.; Lin, C.C.; Shih, T.C.; Tseng, R.J.; Yu, M.C.; Lin, Y.J.; Hsieh, S.Y. The miR-200b-ZEB1 circuit regulates diverse stemness of human hepatocellular carcinoma. Mol. Carcinog. 2017, 56, 2035–2047. [Google Scholar] [CrossRef] [PubMed]
  77. Dou, C.; Sun, L.; Jin, X.; Han, M.; Zhang, B.; Jiang, X.; Lv, J.; Li, T. Long non-coding RNA CARLo-5 promotes tumor progression in hepatocellular carcinoma via suppressing miR-200b expression. Oncotarget 2017, 8, 70172–70182. [Google Scholar] [CrossRef]
  78. Li, Q.; Song, W.; Wang, W.; Yao, S.; Tian, C.; Cai, X.; Wang, L. Suppression of epithelial-mesenchymal transition in hepatocellular carcinoma cells by Kruppel-like factor 4. Oncotarget 2016, 7, 29749–29760. [Google Scholar] [CrossRef]
  79. Liu, X.; Chen, D.; Chen, H.; Wang, W.; Liu, Y.; Wang, Y.; Duan, C.; Ning, Z.; Guo, X.; Otkur, W.; et al. YB1 regulates miR-205/200b-ZEB1 axis by inhibiting microRNA maturation in hepatocellular carcinoma. Cancer Commun. 2021, 41, 576–595. [Google Scholar] [CrossRef]
  80. Liu, L.; Jiang, H.; Pan, H.; Zhu, X. LncRNA XIST promotes liver cancer progression by acting as a molecular sponge of miR-200b-3p to regulate ZEB1/2 expression. J. Int. Med. Res. 2021, 49, 3000605211016211. [Google Scholar] [CrossRef]
  81. Chu, C.H.; Chou, W.; Wang, F.; Yeh, C.N.; Chen, T.C.; Yeh, T.S. Expression profile of microRNA-200 family in cholangiocarcinoma arising from choledochal cyst. J. Gastroenterol. Hepatol. 2016, 31, 1052–1059. [Google Scholar] [CrossRef] [PubMed]
  82. Lin, H.; Yang, L.; Tian, F.; Nie, S.; Zhou, H.; Liu, J.; Chen, W. Up-regulated LncRNA-ATB regulates the growth and metastasis of cholangiocarcinoma via miR-200c signals. Onco Targets Ther. 2019, 12, 7561–7571. [Google Scholar] [CrossRef]
  83. Hossain, A.J.; Islam, R.; Kim, J.G.; Dogsom, O.; Cap, K.C.; Park, J.B. Pyruvate Dehydrogenase A1 Phosphorylated by Insulin Associates with Pyruvate Kinase M2 and Induces LINC00273 through Histone Acetylation. Biomedicines 2022, 10, 1256. [Google Scholar] [CrossRef]
  84. Gu, Y.; Zhu, Z.; Pei, H.; Xu, D.; Jiang, Y.; Zhang, L.; Xiao, L. Long non-coding RNA NNT-AS1 promotes cholangiocarcinoma cells proliferation and epithelial-to-mesenchymal transition through down-regulating miR-203. Aging 2020, 12, 2333–2346. [Google Scholar] [CrossRef]
  85. Zhao, Q.; Wu, C.; Wang, J.; Li, X.; Fan, Y.; Gao, S.; Wang, K. LncRNA SNHG3 Promotes Hepatocellular Tumorigenesis by Targeting miR-326. Tohoku J. Exp. Med. 2019, 249, 43–56. [Google Scholar] [CrossRef] [PubMed]
  86. Dong, Y.; Li, F.; Wang, J.; Hu, J.; Li, Z.; Gu, Y.; Feng, Y. miR-369 inhibits Liver Cancer progression by targeting ZEB1 pathway and predicts the prognosis of HCC patients. J. Cancer 2021, 12, 3067–3076. [Google Scholar] [CrossRef] [PubMed]
  87. Zhou, P.; Liu, Y.; Wu, G.; Lu, K.; Zhao, T.; Yang, L. LincRNA PRNCR1 activates the Wnt/beta-catenin pathway to drive the deterioration of hepatocellular carcinoma via regulating miR-411-3p/ZEB1 axis. Biotechnol. Genet. Eng. Rev. 2024, 40, 4809–4824. [Google Scholar] [CrossRef]
  88. Peng, Z.; Ouyang, X.; Wang, Y.; Fan, Q. MAPKAPK5-AS1 drives the progression of hepatocellular carcinoma via regulating miR-429/ZEB1 axis. BMC Mol. Cell Biol. 2022, 23, 21. [Google Scholar] [CrossRef]
  89. Li, X.; Yang, Z.S.; Cai, W.W.; Deng, Y.; Chen, L.; Tan, S.L. Dihydromyricetin Inhibits Tumor Growth and Epithelial-Mesenchymal Transition through regulating miR-455-3p in Cholangiocarcinoma. J. Cancer 2021, 12, 6058–6070. [Google Scholar] [CrossRef]
  90. Youssef, A.I.; Khaled, G.M.; Amleh, A. Functional role and epithelial to mesenchymal transition of the miR-590-3p/MDM2 axis in hepatocellular carcinoma. BMC Cancer 2023, 23, 396. [Google Scholar] [CrossRef]
  91. Li, L.Y.; Yang, J.F.; Rong, F.; Luo, Z.P.; Hu, S.; Fang, H.; Wu, Y.; Yao, R.; Kong, W.H.; Feng, X.W.; et al. ZEB1 serves an oncogenic role in the tumourigenesis of HCC by promoting cell proliferation, migration, and inhibiting apoptosis via Wnt/beta-catenin signaling pathway. Acta Pharmacol. Sin. 2021, 42, 1676–1689. [Google Scholar] [CrossRef]
  92. Peng, L.; Jiang, B.; Yuan, X.; Qiu, Y.; Peng, J.; Huang, Y.; Zhang, C.; Zhang, Y.; Lin, Z.; Li, J.; et al. Super-Enhancer-Associated Long Noncoding RNA HCCL5 Is Activated by ZEB1 and Promotes the Malignancy of Hepatocellular Carcinoma. Cancer Res. 2019, 79, 572–584. [Google Scholar] [CrossRef]
  93. Zhao, H.; Liu, C.; Zhao, C.; Che, C.; Liu, W.; Mei, Y. Alternatively-spliced lncRNA-PNUTS promotes HCC cell EMT via regulating ZEB1 expression. Tumori J. 2023, 109, 28–37. [Google Scholar] [CrossRef] [PubMed]
  94. Liu, T.A.; Jan, Y.J.; Ko, B.S.; Liang, S.M.; Chen, S.C.; Wang, J.; Hsu, C.; Wu, Y.M.; Liou, J.Y. 14-3-3epsilon overexpression contributes to epithelial-mesenchymal transition of hepatocellular carcinoma. PLoS ONE 2013, 8, e57968. [Google Scholar] [CrossRef]
  95. Lu, K.; Zhao, T.; Yang, L.; Liu, Y.; Ruan, X.; Cui, L.; Zhang, Y. HMGB2 upregulation promotes the progression of hepatocellular carcinoma cells through the activation of ZEB1/vimentin axis. J. Gastrointest. Oncol. 2023, 14, 2178–2191. [Google Scholar] [CrossRef] [PubMed]
  96. Zhong, Y.; Huang, H.; Chen, M.; Huang, J.; Wu, Q.; Yan, G.R.; Chen, D. POU2F1 over-expression correlates with poor prognoses and promotes cell growth and epithelial-to-mesenchymal transition in hepatocellular carcinoma. Oncotarget 2017, 8, 44082–44095. [Google Scholar] [CrossRef]
  97. Chua, H.H.; Chang, M.H.; Chen, Y.H.; Tsuei, D.J.; Jeng, Y.M.; Lee, P.H.; Ni, Y.H. PIM1-Induced Cytoplasmic Expression of RBMY Mediates Hepatocellular Carcinoma Metastasis. Cell. Mol. Gastroenterol. Hepatol. 2023, 15, 121–152. [Google Scholar] [CrossRef] [PubMed]
  98. Hung, T.H.; Huang, Y.; Yeh, C.T.; Yeh, C.N.; Yu, J.; Lin, C.C.; Chiou, S.P.; Chiang, P.Y.; Hung, J.T.; Yu, A.L. High expression of embryonic stem cell marker SSEA3 confers poor prognosis and promotes epithelial mesenchymal transition in hepatocellular carcinoma. Biomed. J. 2024, 47, 100612. [Google Scholar] [CrossRef]
  99. Wang, J.; He, H.; Jiang, Q.; Wang, Y.; Jia, S. CBX6 Promotes HCC Metastasis Via Transcription Factors Snail/Zeb1-Mediated EMT Mechanism. Onco Targets Ther. 2020, 13, 12489–12500. [Google Scholar] [CrossRef]
  100. Zhang, X.; Wang, F.; Huang, Y.; Ke, K.; Zhao, B.; Chen, L.; Liao, N.; Wang, L.; Li, Q.; Liu, X.; et al. FGG promotes migration and invasion in hepatocellular carcinoma cells through activating epithelial to mesenchymal transition. Cancer Manag. Res. 2019, 11, 1653–1665. [Google Scholar] [CrossRef]
  101. Huang, D.; Cao, L.; Zheng, S. CAPZA1 modulates EMT by regulating actin cytoskeleton remodelling in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2017, 36, 13. [Google Scholar] [CrossRef] [PubMed]
  102. Qiu, B.; Wei, W.; Zhu, J.; Fu, G.; Lu, D. EMT induced by loss of LKB1 promotes migration and invasion of liver cancer cells through ZEB1-induced YAP signaling. Oncol. Lett. 2018, 16, 6465–6471. [Google Scholar] [CrossRef] [PubMed]
  103. Xie, C.; Liu, G.; Li, M.; Fang, Y.; Qian, K.; Tang, Y.; Wu, X.; Lei, X.; Li, X.; Liu, Q.; et al. Targeting TRPV1 on cellular plasticity regulated by Ovol 2 and Zeb 1 in hepatocellular carcinoma. Biomed. Pharmacother. 2019, 118, 109270. [Google Scholar] [CrossRef]
  104. Claperon, A.; Mergey, M.; Nguyen Ho-Bouldoires, T.H.; Vignjevic, D.; Wendum, D.; Chretien, Y.; Merabtene, F.; Frazao, A.; Paradis, V.; Housset, C.; et al. EGF/EGFR axis contributes to the progression of cholangiocarcinoma through the induction of an epithelial-mesenchymal transition. J. Hepatol. 2014, 61, 325–332. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, T.J.; Xue, D.; Zhang, C.D.; Zhang, Z.D.; Liu, Q.R.; Wang, J.Q. Cullin 4A is associated with epithelial to mesenchymal transition and poor prognosis in perihilar cholangiocarcinoma. World J. Gastroenterol. 2017, 23, 2318–2329. [Google Scholar] [CrossRef]
  106. Zhou, C.; Zheng, Y.; Li, L.; Zhai, W.; Li, R.; Liang, Z.; Zhao, L. Adrenomedullin promotes intrahepatic cholangiocellular carcinoma metastasis and invasion by inducing epithelial-mesenchymal transition. Oncol. Rep. 2015, 34, 610–616. [Google Scholar] [CrossRef]
  107. Abouhashem, N.S.; Elwan, A.; El Hefnawy, A.S.; Atwa, H.A. Expression of HnRNP A1, ZEB1, and E-cadherin in Hepatocellular carcinoma and their impact on patients’ prognosis and survival. Indian J. Pathol. Microbiol. 2022, 65, 589–597. [Google Scholar] [CrossRef]
  108. Chen, J.; Zhang, L.; Ma, S.; Lu, G.; Wang, D. The aberrant expressions of MACC1, ZEB1, and KLF4 in hepatocellular carcinoma and their clinical significance. Int. J. Clin. Exp. Pathol. 2019, 12, 3653–3661. [Google Scholar]
  109. Qin, Y.; Yu, J.; Zhang, M.; Qin, F.; Lan, X. ZEB1 promotes tumorigenesis and metastasis in hepatocellular carcinoma by regulating the expression of vimentin. Mol. Med. Rep. 2019, 19, 2297–2306. [Google Scholar] [CrossRef]
  110. Zhou, Y.M.; Cao, L.; Li, B.; Zhang, R.X.; Sui, C.J.; Yin, Z.F.; Yang, J.M. Clinicopathological significance of ZEB1 protein in patients with hepatocellular carcinoma. Ann. Surg. Oncol. 2012, 19, 1700–1706. [Google Scholar] [CrossRef]
  111. Terashita, K.; Chuma, M.; Hatanaka, Y.; Hatanaka, K.; Mitsuhashi, T.; Yokoo, H.; Ohmura, T.; Ishizu, H.; Muraoka, S.; Nagasaka, A.; et al. ZEB1 expression is associated with prognosis of intrahepatic cholangiocarcinoma. J. Clin. Pathol. 2016, 69, 593–599. [Google Scholar] [CrossRef]
  112. Wang, H.; Lin, F.; Xu, Z.; Yu, S.; Li, G.; Liao, S.; Zhao, W.; Zhang, F.; Wang, J.; Wang, S.; et al. ZEB1 Transcriptionally Activates PHGDH to Facilitate Carcinogenesis and Progression of HCC. Cell. Mol. Gastroenterol. Hepatol. 2023, 16, 541–556. [Google Scholar] [CrossRef] [PubMed]
  113. Zhou, Y.; Lin, F.; Wan, T.; Chen, A.; Wang, H.; Jiang, B.; Zhao, W.; Liao, S.; Wang, S.; Li, G.; et al. ZEB1 enhances Warburg effect to facilitate tumorigenesis and metastasis of HCC by transcriptionally activating PFKM. Theranostics 2021, 11, 5926–5938. [Google Scholar] [CrossRef]
  114. Lehmann, W.; Mossmann, D.; Kleemann, J.; Mock, K.; Meisinger, C.; Brummer, T.; Herr, R.; Brabletz, S.; Stemmler, M.P.; Brabletz, T. ZEB1 turns into a transcriptional activator by interacting with YAP1 in aggressive cancer types. Nat. Commun. 2016, 7, 10498. [Google Scholar] [CrossRef] [PubMed]
  115. Zhou, H.; Du, Y.; Wei, X.; Song, C.; Song, J.; Xu, N.; Huang, W.; Chen, L.; Yao, F.; Du, D.; et al. DDX56 transcriptionally activates MIST1 to facilitate tumorigenesis of HCC through PTEN-AKT signaling. Theranostics 2022, 12, 6069–6087. [Google Scholar] [CrossRef]
  116. Wang, X.F.; Liu, J.Y.; Li, J.L.; Wang, F.Z.; Sun, B.L. ZEB1 causes the production of hsa-microRNA-99b/let-7e/microRNA-125a cluster and promotes invasion of liver cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 1468–1475. [Google Scholar] [CrossRef] [PubMed]
  117. Fedele, M.; Sgarra, R.; Battista, S.; Cerchia, L.; Manfioletti, G. The Epithelial-Mesenchymal Transition at the Crossroads between Metabolism and Tumor Progression. Int. J. Mol. Sci. 2022, 23, 800. [Google Scholar] [CrossRef]
  118. Hua, Q.; Mi, B.; Huang, G. The emerging co-regulatory role of long noncoding RNAs in epithelial-mesenchymal transition and the Warburg effect in aggressive tumors. Crit. Rev. Oncol. Hematol. 2018, 126, 112–120. [Google Scholar] [CrossRef]
  119. Gong, K.; Huang, Y.; Zheng, Y.; Zhu, Y.; Hao, W.; Shi, K. Preclinical efficacy of CBR-5884 against epithelial ovarian cancer cells by targeting the serine synthesis pathway. Discov. Oncol. 2024, 15, 154. [Google Scholar] [CrossRef]
  120. Liu, X.; Liu, B.; Wang, J.; Liu, H.; Wu, J.; Qi, Y.; Liu, Y.; Zhu, H.; Li, C.; Yang, L.; et al. PHGDH activation fuels glioblastoma progression and radioresistance via serine synthesis pathway. J. Exp. Clin. Cancer Res. 2025, 44, 99. [Google Scholar] [CrossRef]
  121. Shu, Y.; Hao, Y.; Feng, J.; Liu, H.; Li, S.T.; Feng, J.; Jiang, Z.; Ye, L.; Zhou, Y.; Sun, Y.; et al. Non-canonical phosphoglycerate dehydrogenase activity promotes liver cancer growth via mitochondrial translation and respiratory metabolism. EMBO J. 2022, 41, e111550. [Google Scholar] [CrossRef]
  122. Sreekumar, R.; Emaduddin, M.; Al-Saihati, H.; Moutasim, K.; Chan, J.; Spampinato, M.; Bhome, R.; Yuen, H.M.; Mescoli, C.; Vitale, A.; et al. Protein kinase C inhibitors override ZEB1-induced chemoresistance in HCC. Cell Death Dis. 2019, 10, 703. [Google Scholar] [CrossRef]
  123. Xiang, Y.; Jia, M.; Gao, Y.; Yang, F.; Wang, T.; Dai, R.; Wang, M.; Miao, H. Cisplatin Disrupts Proteasome Bounce-Back Effect through Suppressing ZEB1/Nfe2l1 in Cholangiocarcinoma. Front. Biosci. (Landmark Ed.) 2024, 29, 106. [Google Scholar] [CrossRef]
  124. Xu, Y.; Luan, G.; Liu, F.; Zhang, Y.; Li, Z.; Liu, Z.; Yang, T. Exosomal miR-200b-3p induce macrophage polarization by regulating transcriptional repressor ZEB1 in hepatocellular carcinoma. Hepatol. Int. 2023, 17, 889–903, Correction in Hepatol. Int. 2024, 18, 1064. [Google Scholar] [CrossRef] [PubMed]
  125. Ueno, T.; Tsuchikawa, T.; Hatanaka, K.C.; Hatanaka, Y.; Mitsuhashi, T.; Nakanishi, Y.; Noji, T.; Nakamura, T.; Okamura, K.; Matsuno, Y.; et al. Prognostic impact of programmed cell death ligand 1 (PD-L1) expression and its association with epithelial-mesenchymal transition in extrahepatic cholangiocarcinoma. Oncotarget 2018, 9, 20034–20047. [Google Scholar] [CrossRef]
  126. Wirbel, C.; Durand, S.; Boivin, F.; Plaschka, M.; Benboubker, V.; Grimont, M.; Barbollat-Boutrand, L.; Tondeur, G.; Balme, B.; Harou, O.; et al. ZEB1 transcription factor induces tumor cell PD-L1 expression in melanoma. Cancer Immunol. Immunother. 2025, 74, 141. [Google Scholar] [CrossRef] [PubMed]
  127. Haruna, T.; Kudo, M.; Ishino, K.; Ueda, J.; Shintani-Domoto, Y.; Yoshimori, D.; Fuji, T.; Kawamoto, Y.; Teduka, K.; Kitamura, T.; et al. Molecular biological role of epithelial splicing regulatory protein 1 in intrahepatic cholangiocarcinoma. Hepatol. Res. 2024, 55, 58–68. [Google Scholar] [CrossRef]
  128. Mizuguchi, Y.; Isse, K.; Specht, S.; Lunz, J.G., 3rd; Corbitt, N.; Takizawa, T.; Demetris, A.J. Small proline rich protein 2a in benign and malignant liver disease. Hepatology 2014, 59, 1130–1143. [Google Scholar] [CrossRef]
  129. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [PubMed]
  130. Vaquero, J.; Lobe, C.; Tahraoui, S.; Claperon, A.; Mergey, M.; Merabtene, F.; Wendum, D.; Coulouarn, C.; Housset, C.; Desbois-Mouthon, C.; et al. The IGF2/IR/IGF1R Pathway in Tumor Cells and Myofibroblasts Mediates Resistance to EGFR Inhibition in Cholangiocarcinoma. Clin. Cancer Res. 2018, 24, 4282–4296. [Google Scholar] [CrossRef]
  131. Hao, Y.; Long, Z.; Gu, X. Farrerol suppresses epithelial-mesenchymal transition in hepatocellular carcinoma via suppression of TGF-beta1/Smad2/3 signaling. Pathol. Res. Pract. 2024, 264, 155719. [Google Scholar] [CrossRef] [PubMed]
  132. Li, W.; Pei, S.; Zhang, X.; Qi, D.; Zhang, W.; Dou, Y.; Yang, R.; Yao, X.; Zhang, Z.; Xie, S.; et al. Cinobufotalin inhibits the epithelial-mesenchymal transition of hepatocellular carcinoma cells through down-regulate beta-catenin in vitro and in vivo. Eur. J. Pharmacol. 2022, 922, 174886. [Google Scholar] [CrossRef] [PubMed]
  133. Zhong, W.; Tong, Y.; Li, Y.; Yuan, J.; Hu, S.; Hu, T.; Song, G. Mesenchymal stem cells in inflammatory microenvironment potently promote metastatic growth of cholangiocarcinoma via activating Akt/NF-kappaB signaling by paracrine CCL5. Oncotarget 2017, 8, 73693–73704. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic structure of ZEB1. ZEB1 is formed by two DNA-binding zinc finger domains located near the N-terminal (NZF) and C-terminal (CZF) regions, a central homeodomain, binding sites for SMADs (SID), CtBP (CID) and the p300-P/CBP-associated factor binding domain (CBD).
Figure 1. Schematic structure of ZEB1. ZEB1 is formed by two DNA-binding zinc finger domains located near the N-terminal (NZF) and C-terminal (CZF) regions, a central homeodomain, binding sites for SMADs (SID), CtBP (CID) and the p300-P/CBP-associated factor binding domain (CBD).
Ijms 26 11135 g001
Figure 2. Mechanisms of transcriptional regulation of ZEB1 expression in liver cancer. The different mechanisms involved in the regulation of the ZEB1 promoter are depicted in this figure and detailed in the text. (A) Main signaling pathways regulating ZEB1 transcription. (B) ZEB1 transcriptional regulation by other pathways. (C) Impact of tumor microenvironment in ZEB1 transcriptional regulation. ABCA8, ATP binding cassette subfamily A member 8; AKT, AKT serine/threonine kinase; AP-2, Activator Protein-2 gamma; CCL5, C-C motif chemokine ligand 5; CCR5, C-C motif chemokine ligand 5 receptor; eEF-1γ, eukaryotic translation elongation factor 1 gamma; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; EGR1, early growth response 1; ERK, extracellular signal-regulated kinase; FBXW7, F-box and WD repeat domain containing 7; FOXO3A, forkhead box O3; GINS1, GINS complex subunit 1; GSK3β, glycogen synthase kinase 3 beta; HF1α, hypoxia inducible factor 1 subunit alpha; HN1L, hematological and neurological expressed 1-like; IGFBP2, Insulin like growth factor binding protein 2; IRS, insulin receptor substrate; METTL13, methyltransferase 13; mTOR, mechanistic target of rapamycin kinase; NFκB, nuclear factor kappa B; NICD, notch intracellular domain; NOTCH, Neurogenic locus notch homolog protein; OTUB2, OTU deubiquitinase, ubiquitin aldehyde binding 2; PDHA1, pyruvate dehydrogenase E1 subunit alpha 1; PKM2, pyruvate kinase M2; PPM1G, Protein phosphatase 1G; PPT1, palmitoyl-protein thioesterase 1; PTK7, Tyrosine-protein kinase-like 7; PTP4A1, Protein Tyrosine Phosphatase Type 4A1; RTK, receptor tyrosine kinase; SMYD3, SET and MYND domain containing 3; SNAIL, snail family transcriptional repressor 1; SORT1, Sortilin 1; SP1, Specificity protein 1; TBL1X, transducin beta like 1 X-linked; TGF-β, transforming growth factor-beta; TGFβR1, transforming growth factor-beta receptor 1; THRSP, thyroid hormone responsive protein; TXNDC12, thioredoxin domain containing 12; TRAF6, TNF receptor associated factor 6; ZEB1, Zinc finger E-box-binding homeobox 1; ZIP4, Zrt- and Irt-like protein 4.
Figure 2. Mechanisms of transcriptional regulation of ZEB1 expression in liver cancer. The different mechanisms involved in the regulation of the ZEB1 promoter are depicted in this figure and detailed in the text. (A) Main signaling pathways regulating ZEB1 transcription. (B) ZEB1 transcriptional regulation by other pathways. (C) Impact of tumor microenvironment in ZEB1 transcriptional regulation. ABCA8, ATP binding cassette subfamily A member 8; AKT, AKT serine/threonine kinase; AP-2, Activator Protein-2 gamma; CCL5, C-C motif chemokine ligand 5; CCR5, C-C motif chemokine ligand 5 receptor; eEF-1γ, eukaryotic translation elongation factor 1 gamma; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; EGR1, early growth response 1; ERK, extracellular signal-regulated kinase; FBXW7, F-box and WD repeat domain containing 7; FOXO3A, forkhead box O3; GINS1, GINS complex subunit 1; GSK3β, glycogen synthase kinase 3 beta; HF1α, hypoxia inducible factor 1 subunit alpha; HN1L, hematological and neurological expressed 1-like; IGFBP2, Insulin like growth factor binding protein 2; IRS, insulin receptor substrate; METTL13, methyltransferase 13; mTOR, mechanistic target of rapamycin kinase; NFκB, nuclear factor kappa B; NICD, notch intracellular domain; NOTCH, Neurogenic locus notch homolog protein; OTUB2, OTU deubiquitinase, ubiquitin aldehyde binding 2; PDHA1, pyruvate dehydrogenase E1 subunit alpha 1; PKM2, pyruvate kinase M2; PPM1G, Protein phosphatase 1G; PPT1, palmitoyl-protein thioesterase 1; PTK7, Tyrosine-protein kinase-like 7; PTP4A1, Protein Tyrosine Phosphatase Type 4A1; RTK, receptor tyrosine kinase; SMYD3, SET and MYND domain containing 3; SNAIL, snail family transcriptional repressor 1; SORT1, Sortilin 1; SP1, Specificity protein 1; TBL1X, transducin beta like 1 X-linked; TGF-β, transforming growth factor-beta; TGFβR1, transforming growth factor-beta receptor 1; THRSP, thyroid hormone responsive protein; TXNDC12, thioredoxin domain containing 12; TRAF6, TNF receptor associated factor 6; ZEB1, Zinc finger E-box-binding homeobox 1; ZIP4, Zrt- and Irt-like protein 4.
Ijms 26 11135 g002
Figure 3. Mechanisms involved in the regulation of ZEB1 expression in liver cancer at the post-transcriptional (A) and post-translational (B) level. (A) Post-transcriptional mechanisms include mRNA modifications and non-coding RNA regulation. (B) Post-transcriptional mechanisms include protein modifications that regulate protein stability and ZEB1 degradation by the proteasome or by autophagy. circRNA, circular RNA; CRL4-DCAF15, DDB1 and CUL4 associated factor 15; HIPK2, Homeodomain Interacting Protein Kinase 2; HMGB1, high mobility group box 1; lncRNA, long non-coding RNA; MAZ, MYC associated zinc finger protein; METTL3, Methyltransferase-like protein 3; iRNA, microRNA; NONO, non-POU domain containing octamer binding; OGA, O-GlcNAcase; OGT, O-GlcNAc transferase; TRIM26, tripartite motif containing 26; UPAT, ubiquitin-like plant homeodomain (PHD) and really interesting new gene (RING) finger domain containing protein 1 (UHRF1) protein-associated transcript; USP22, ubiquitin specific peptidase 22; USP39, ubiquitin specific peptidase 39; YTHDF3, YTH domain-containing family protein 3; ZEB1, Zinc finger E-box-binding homeobox 1.
Figure 3. Mechanisms involved in the regulation of ZEB1 expression in liver cancer at the post-transcriptional (A) and post-translational (B) level. (A) Post-transcriptional mechanisms include mRNA modifications and non-coding RNA regulation. (B) Post-transcriptional mechanisms include protein modifications that regulate protein stability and ZEB1 degradation by the proteasome or by autophagy. circRNA, circular RNA; CRL4-DCAF15, DDB1 and CUL4 associated factor 15; HIPK2, Homeodomain Interacting Protein Kinase 2; HMGB1, high mobility group box 1; lncRNA, long non-coding RNA; MAZ, MYC associated zinc finger protein; METTL3, Methyltransferase-like protein 3; iRNA, microRNA; NONO, non-POU domain containing octamer binding; OGA, O-GlcNAcase; OGT, O-GlcNAc transferase; TRIM26, tripartite motif containing 26; UPAT, ubiquitin-like plant homeodomain (PHD) and really interesting new gene (RING) finger domain containing protein 1 (UHRF1) protein-associated transcript; USP22, ubiquitin specific peptidase 22; USP39, ubiquitin specific peptidase 39; YTHDF3, YTH domain-containing family protein 3; ZEB1, Zinc finger E-box-binding homeobox 1.
Ijms 26 11135 g003
Figure 4. Schematic representation of the mechanisms governing HCC tumor progression and chemoresistance that are regulated by ZEB1 in tumor and tumor microenvironment cells. AKT, AKT serine/threonine kinase; CTGF, connective tissue growth factor; DDX56, DEAD-Box 56; MIST1, muscle, intestine and stomach expression 1; PFKM, phosphofructokinase-1; PHGDH, phosphoglycerate dehydrogenase; PKCA, Protein kinase C alpha; PTEN, phosphatase and tensin homolog; SDPR, serum deprivation response; UPAT, ubiquitin-like plant homeodomain (PHD) and really interesting new gene (RING) finger domain-containing protein 1 (UHRF1) protein-associated transcript; USP22, ubiquitin specific peptidase 22; VEGFA, vascular endothelial growth factor A; YAP, Yes-Associated Protein; ZEB1, Zinc finger E-box binding homeobox 1.
Figure 4. Schematic representation of the mechanisms governing HCC tumor progression and chemoresistance that are regulated by ZEB1 in tumor and tumor microenvironment cells. AKT, AKT serine/threonine kinase; CTGF, connective tissue growth factor; DDX56, DEAD-Box 56; MIST1, muscle, intestine and stomach expression 1; PFKM, phosphofructokinase-1; PHGDH, phosphoglycerate dehydrogenase; PKCA, Protein kinase C alpha; PTEN, phosphatase and tensin homolog; SDPR, serum deprivation response; UPAT, ubiquitin-like plant homeodomain (PHD) and really interesting new gene (RING) finger domain-containing protein 1 (UHRF1) protein-associated transcript; USP22, ubiquitin specific peptidase 22; VEGFA, vascular endothelial growth factor A; YAP, Yes-Associated Protein; ZEB1, Zinc finger E-box binding homeobox 1.
Ijms 26 11135 g004
Figure 5. Schematic representation of the mechanisms governing CCA tumor progression that are regulated by ZEB1 in tumor cells and the tumor microenvironment. ACTA2, Actin alpha 2, smooth muscle; CDH1, E-cadherin; COL4A1, collagen 4A1; CTGF, connective tissue growth factor; HGF, hepatocyte growth factor; VIM, vimentin; ZEB1, zinc finger E-box binding homeobox 1.
Figure 5. Schematic representation of the mechanisms governing CCA tumor progression that are regulated by ZEB1 in tumor cells and the tumor microenvironment. ACTA2, Actin alpha 2, smooth muscle; CDH1, E-cadherin; COL4A1, collagen 4A1; CTGF, connective tissue growth factor; HGF, hepatocyte growth factor; VIM, vimentin; ZEB1, zinc finger E-box binding homeobox 1.
Ijms 26 11135 g005
Table 2. Clinical relevance of ZEB1 in the survival of patients with liver cancer.
Table 2. Clinical relevance of ZEB1 in the survival of patients with liver cancer.
TumorSample
Size
Detection
Method
Cut Off% ZEB1 Positive/
High Expression
PFS/OSHR95%CIp-ValueRef.
HCC54IHCExpression > 1%29.6PFS (MV)15.510.1–20.9<0.001[107]
OS (MV)9.64.34–14.86<0.001
HCC153IHCScore > 259.4OS (MV)1.8121.060–3.0970.030[108]
HCC110WB-65.4PFS (MV)1.8141.002–3.2840.048[110]
OS (MV)2.2221.097–4.5030.027
HCC108IHC-23.0OS (MV)1.451.02–2.000.037[13]
OS (UV)1.200.83–1.710.320
CCA102IHCScore 0–30046.1OS (UV)0.9110.493–1.6860.027[111]
CCA, cholangiocarcinoma; CI, confidence interval; HCC, hepatocellular carcinoma; HR, hazard ratio; IHC, immunohistochemistry; MV, multivariate; OS, overall survival, PFS; progression free survival; UV, univariate; WB, Western blot.
Table 3. Association of ZEB1 expression with clinicopathological parameters in patients with liver cancer.
Table 3. Association of ZEB1 expression with clinicopathological parameters in patients with liver cancer.
TumorParameterSample SizeZEB1 NegativeZEB1 Positivep-ValueRef.
HCCGrade543816 [107]
I121110.006
II28217
III1468
Staging543816
I9810.001
II20191
III1679
IV945
Vascular Invasion543816
No211920.01
Yes331914
Number of metastases543816
Uni-focal282620.001
Multi-focal261214
Child pough class543816
A191810.003
B281711
C734
HCCNumber of tumors1536291 [108]
1825824<0.001
>171467
Grade1536291
Well + moderate774433<0.001
Poor761858
Staging1536291
I52448<0.001
II64757
III421120
IV606
HCCStaging803842 [109]
I–II4314290.04
III–IV372413
HCCStaging [110]
I12840.010
II301317
III–IV681751
Tumor size
<53718190.027
>5732053
Intrahepatic metastases
Absent8233490.031
Present28523
Vascular invasion
Absent4723240.006
Present631548
Early recurrence
Yes346280.013
No763244
HCCVascular invasion108 [13]
Absent7010600.016
Present381325
Staging108
I + II + III8815730.023
IV20812
CCASex1025547 [111]
Men6329340.042
Women392613
Age (years)1025547
<645120310.003
>64513516
Staging1025547
I + II + III5937220.037
IV431825
Differentiation1025547
Well201640.017
Moderately583048
Poorly24915
Lymph node metastasis1025547
Absent7043270.024
Present321220
Portal vein invasion1025547
Absent5937220.037
Present431825
CCAGrade453510 [14]
I17152<0.05
II18153
III1055
Vascular Invasion453510
Absent13130<0.05
Present322210
CCA, cholangiocarcinoma, HCC, hepatocellular carcinoma.
Table 4. Therapeutic strategies against ZEB1 regulated functions in liver cancer.
Table 4. Therapeutic strategies against ZEB1 regulated functions in liver cancer.
TumorStrategyMoleculeTargetRef.
HCCChemical compoundFarrerolSMAD2/3[131]
HCCChemical compoundCinobufotalinβ-catenin[132]
HCCChemical compoundSalinomycinFOXO3a[28]
CCAChemical compoundDihydromyricetinmiR-455-3p[89]
HCCChemical compoundArtesunateO-GlcNAcase[56]
HCCChemical compoundPKC inhibitorsPKCA[122]
CCAChemical compoundMaravirocCCR5[133]
CCANeutralizing antibody-CCL5[133]
HCCmiRNAmiR-708ZEB1[91]
CCA, cholangiocarcinoma; CCL5, C-C motif chemokine ligand 5; CCR5, C-C motif chemokine ligand 5 receptor; FOXO3a, forkhead box O3; HCC, hepatocellular carcinoma; PKCA, Protein kinase C alpha; ZEB1, zinc finger E-box binding homeobox 1.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gonzalez-Sanchez, E.; Roldan-Hernandez, C.A.; Martin-Ramirez, A.; Garcia-Collado, L.; Fouassier, L.; Vaquero, J. Epithelial to Mesenchymal Transition Transcriptional Regulator ZEB1 in Liver Cancer: Oncogenic Roles and Therapeutic Potential. Int. J. Mol. Sci. 2025, 26, 11135. https://doi.org/10.3390/ijms262211135

AMA Style

Gonzalez-Sanchez E, Roldan-Hernandez CA, Martin-Ramirez A, Garcia-Collado L, Fouassier L, Vaquero J. Epithelial to Mesenchymal Transition Transcriptional Regulator ZEB1 in Liver Cancer: Oncogenic Roles and Therapeutic Potential. International Journal of Molecular Sciences. 2025; 26(22):11135. https://doi.org/10.3390/ijms262211135

Chicago/Turabian Style

Gonzalez-Sanchez, Ester, Carlos Andres Roldan-Hernandez, Ana Martin-Ramirez, Lucia Garcia-Collado, Laura Fouassier, and Javier Vaquero. 2025. "Epithelial to Mesenchymal Transition Transcriptional Regulator ZEB1 in Liver Cancer: Oncogenic Roles and Therapeutic Potential" International Journal of Molecular Sciences 26, no. 22: 11135. https://doi.org/10.3390/ijms262211135

APA Style

Gonzalez-Sanchez, E., Roldan-Hernandez, C. A., Martin-Ramirez, A., Garcia-Collado, L., Fouassier, L., & Vaquero, J. (2025). Epithelial to Mesenchymal Transition Transcriptional Regulator ZEB1 in Liver Cancer: Oncogenic Roles and Therapeutic Potential. International Journal of Molecular Sciences, 26(22), 11135. https://doi.org/10.3390/ijms262211135

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop