Next Article in Journal
Zinc: From Biological Functions to Therapeutic Potential
Next Article in Special Issue
Epigenetic and Immunological Features of Bladder Cancer
Previous Article in Journal
Spexin2 Is a Novel Food Regulator in Gallus gallus
Previous Article in Special Issue
Cell-Free DNA Fragmentomics: The Novel Promising Biomarker
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Histone Modifications Represent a Key Epigenetic Feature of Epithelial-to-Mesenchyme Transition in Pancreatic Cancer

Abdominal Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(5), 4820; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054820
Submission received: 27 December 2022 / Revised: 25 February 2023 / Accepted: 27 February 2023 / Published: 2 March 2023
(This article belongs to the Special Issue Epigenetic Regulation and Cancers)

Abstract

:
Pancreatic cancer is one of the most lethal malignant diseases due to its high invasiveness, early metastatic properties, rapid disease progression, and typically late diagnosis. Notably, the capacity for pancreatic cancer cells to undergo epithelial–mesenchymal transition (EMT) is key to their tumorigenic and metastatic potential, and is a feature that can explain the therapeutic resistance of such cancers to treatment. Epigenetic modifications are a central molecular feature of EMT, for which histone modifications are most prevalent. The modification of histones is a dynamic process typically carried out by pairs of reverse catalytic enzymes, and the functions of these enzymes are increasingly relevant to our improved understanding of cancer. In this review, we discuss the mechanisms through which histone-modifying enzymes regulate EMT in pancreatic cancer.

1. Introduction

Pancreatic cancer is the seventh leading cause of cancer-related death globally [1]. Patient prognosis is bleak, with a five-year survival rate of only 10% [2,3]. Moreover, the incidence of pancreatic cancer has risen steadily in recent years [4]. By 2025, it is expected to become the second-leading cause of cancer-related mortality in the US [1]. Surgery remains the main therapeutic approach but is only effective during the early stages of disease. The early metastases and rapid progression commonly seen in patients result in diagnosis not occurring until the advanced stages of disease in more than half of all cases, which precludes them from surgery [5,6]. Although systemic combination chemotherapy also forms a crucial component of pancreatic cancer treatment, drug resistance is a significant problem [7]. Therefore, it is essential to understand the molecular mechanisms involved in the carcinogenesis, progression, metastasis, and drug resistance of pancreatic cancer to identify precise diagnostic and therapeutic targets.
Epithelial–mesenchymal transition (EMT) is a fundamental process for embryogenesis, organ development, and tissue repair, as well as for tumor invasion and early metastasis [8]. During EMT, epithelial cells lose their apical–basal polarity and cell–cell adhesion, and are transformed into invasive mesenchymal cells [9]. In addition, EMT can generate cells with stem cell properties that allow them to form tumors more efficiently [10,11]. EMT enables cancer cells at the primary site to acquire motility and invasiveness, which can drive the malignant progression of tumors. Furthermore, EMT also can regulate a variety of cancer features, such as tumor cell stemness, adaptation to microenvironmental alterations, and resistance to therapy [12]. EMT facilitates tumorigenesis and promotes the invasion, metastasis, and therapeutic resistance of pancreatic cancer, and is, therefore, associated with poor patient prognosis [13,14,15].
EMT is triggered by signals sent within the cellular microenvironment. The mechanisms underlying this process are intricate and involve multiple signaling pathways and various EMT-induced transcription factors [16]. Interestingly, the acquisition of mesenchymal properties in cancer cells is transient rather than permanent, and cancer cells can restore their epithelial state through the mesenchymal–epithelial transformation (MET) process [17]. Given the extensive gene expression reprogramming needed to complete the reversible EMT process, epigenetic regulators are likely to have essential functions [18]. Epigenetics describes heritable changes in cellular phenotypes that are independent of alterations in the DNA sequence [19]. Epigenetic changes are associated with aberrant gene functions and altered patterns of gene expression. The major epigenetic regulators include DNA methylation, histone modification, chromatin remodeling, and non-coding RNAs [19,20]. These molecular regulatory mechanisms can alter promoter accessibility and chromatin structure, so as to affect cellular gene expression and influence tissue homeostasis [20]. Indeed, we find that the most significant and complex epigenetic regulatory mechanisms detected within pancreatic cancers comprise histone modifications, which include methylation, acetylation, ubiquitination, phosphorylation, and SUMOylation [21]. Histone modifications can be dynamically regulated by enzymes with reverse catalytic activity, and these enzymes are the primary epigenetic regulators of EMT.
This review provides an overview of EMT in pancreatic cancer, together with the mechanisms of histone modification involved in the regulation of EMT, focusing primarily on methylation, acetylation, and ubiquitination. Furthermore, we discuss the application of histone epigenetic regulators as therapeutics for patients with pancreatic cancer.

2. EMT in Pancreatic Cancer

EMT leads to significant phenotypic alterations in cancer cells that are associated with their invasion, spread, and metastasis. Epithelial cells maintain cell–cell contact through tight junctions, adhesive junctions, desmosomes, and gap junctions [22]. During EMT, these cell–cell junctions are lost, and significant cytoskeletal reorganization occurs. Epithelial cells lose top-basal polarity and are transformed into mesenchymal cells, gaining motility and invasiveness [9,23]. The activation of EMT leads to a downregulation of the expression of epithelial genes, including E-cadherin, ZO-1, and occludin. Concurrently, cells acquire a mesenchymal morphology and express mesenchymal markers, such as N-cadherin, vimentin, and fibronectin [8]. The downregulation of E-cadherin (which binds to β-catenin to form mature adhesion junctions between cells [24,25]) is a key step in EMT and is often associated with the invasive potential and undifferentiated phenotype of tumor cells. Moreover, E-cadherin downregulation can disrupt cell adhesions and activate various EMT-related intracellular signaling pathways [26].
EMT is induced by a variety of extracellular stimuli and intracellular signaling pathways, including the transforming growth factor (TGF), Wnt, and hedgehog (Hh) signaling pathways, etc. [16] (Figure 1). TGF-β can induce EMT through different signaling pathways: those that are dependent on the SMAD (suppressor of mothers against decapentaplegic) transcription factor family, and those that are SMAD-independent [27]. In the SMAD-dependent pathway, TGF-β signals through a tetrameric complex containing type I and type II receptors (TβRI and TβRII) to activate SMAD2 and SMAD3. Phosphorylated SMAD2 and SMAD3 form trimers with SMAD4 to regulate EMT-related transcriptional regulators. In the SMAD-independent pathway, TGF-β activates the extracellular signal-regulated kinase 1/2 (ERK)/mitogen-activated protein (MAP) kinase, Rho GTPase, and phosphoinositide 3 (PI3) kinase/Akt pathways to promote EMT [28]. The Wnt signaling pathway is also crucial for the induction of EMT. In the absence of Wnt signaling, β-catenin is phosphorylated by casein Kinase 1 (CK1) and adenomatous polyposis coli (APC)/AXIN/glycogen synthase kinase-3 beta (GSK3β) complexes, prior to being ubiquitinated and degraded by proteasomes. When Wnt ligands bind to frizzled receptors, this degradation process is inhibited, and accumulated cytoplasmic β-catenin transfers to the nucleus and activates the transcription of EMT-related genes [29]. In Hh signaling, Hh ligands bind to the transmembrane receptor patched-1 to inhibit the transmembrane G protein-coupled receptor Smoothened (SMO), initiating intracellular cascades and activating the glioma-associated oncogene (GLI) transcription factors that induce the expression of EMT-related genes [30]. In addition, tyrosine kinase receptors are also involved in pancreatic cancer EMT through the regulation of the PI3K/AKT and ERK/MAPK signaling pathways [31,32]. These signaling pathways activate EMT transcription factors (EMT-TFs) [12], which include the Snail family (Snail1/Snail and Snail2/Slug), TWIST family (Twist1 and Twist2), and Zinc finger E-box binding homeobox family (Zeb1 and Zeb2) of proteins [33].
The tumor microenvironment (TME) is crucial in EMT. The pancreatic cancer microenvironment consists of cancer cells, stromal cells, and extracellular components [34]. TME-inducible factors secreted by cancer cells and cancer-associated fibroblasts (CAF) create an inflammatory microenvironment by recruiting immune cells [35]. Many of these cells secrete cytokines and chemokines that induce EMT [36]. For example, IL-6 regulates the expression of EMT-related genes in pancreatic cancer cells through the STAT3-mediated signaling pathway [37]. As pancreatic cancer contains abundant stromal cells and extracellular matrix but lacks vascularization, severe hypoxia persists within the tumor, causing broad effects on cellular behavior [38]. The adaptive response of pancreatic cancer cells to hypoxia is mainly mediated by hypoxia-inducible factors (HIFs), which bestow more aggressive and therapeutically resistant phenotypes [39]. During hypoxia, the ubiquitin-proteasome degradation pathway mediated by HIF-1 is inhibited. As a consequence, HIF-1α accumulates in cells and binds to HIF-1β to form a functional transcriptional complex [40]. HIF-1α drives hypoxia-induced EMT programming in pancreatic cancer cells through interaction with the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcriptional complex [41]. In addition, HIF-2α can promote EMT in pancreatic cancer cells by regulating the binding of Twist2 to the E-cadherin promoter [42].
The two most common genetic alterations in pancreatic cancer are mutations in the Kirsten rat sarcoma viral (KRAS) oncogene and inactivation of SMAD4 [43]. KRAS-driven tumors frequently exhibit EMT induction [15,44,45,46]. The most extensively studied mechanism for inducing EMT is the TGF-signaling pathway [47], and SMAD4 is an effector of this pathway [28,48]. The function of SMAD4 in the EMT of pancreatic cancer cells remains controversial. Whilst the majority of studies have demonstrated that EMT requires the entire TGF-β signaling pathway (including SMAD4) [28,49,50,51], others have shown that the inactivation of SMAD4 can induce EMT [52,53]. Nevertheless, it is clear that SMAD4 is a major factor in the EMT seen in pancreatic cancers.
Cancer stem cells (CSCs) are undifferentiated quiescent cells that possess properties of self-renewal and cellular plasticity [54]. The molecular features of CSCs are influenced by cells exhibiting an EMT phenotype, and CSCs themselves show an EMT phenotype [55,56,57]. Pancreatic cancer stem cells (PCSCs) are subsets of pancreatic cancers with specific cell surface markers that have important functions during tumor recurrence and therapeutic resistance [58,59,60]. Key signaling pathways that regulate PCSCs can also activate many EMT-related pathways, promoting pancreatic cancer progression and resistance to therapeutics [61,62].

3. Epigenetics in Pancreatic Cancer

Cancer was initially recognized as a genetic disease. However, it is now widely believed that the initiation and progression of cancer cannot be accounted for by genetic alterations alone, but must also involve epigenetic changes [20]. Epigenetic modifications are reversible changes that regulate gene expression without altering DNA sequences. They include DNA methylation, histone modification, chromatin modification, and alterations in noncoding RNA profiles (Figure 2). Chromatin is the carrier of genetic information, and the nucleosome is its basic unit. Each nucleosome consists of a histone octamer composed of the core histones (two copies each of H2A, H2B, H3, and H4), which is wrapped in approximately 147 bp of DNA [63]. DNA methylation transfers a methyl group to the C5 position of cytosine in CpG dinucleotides to form 5-methylcytosine (5mC). The DNA (cytosine-5)-methyltransferase 3A (DNMT3A) and DNMT3B enzymes are responsible for de novo DNA methylation, which is maintained by DNMT1 [64]. Chromatin-remodeling complexes use ATP as an energy source to mobilize, eject, and exchange histones. The SWI/SNF complex alters nucleosome positioning and structure by sliding and ejecting nucleosomes to make the DNA more accessible to transcription factors and other chromatin regulators [65]. Noncoding RNAs (ncRNAs), including circular RNA (circRNA), long non-coding RNA (lncRNA), and microRNA (miRNA), can regulate other epigenetic processes, and be regulated by them. Some specific lncRNAs are extensively associated with chromatin remodeling and modification complexes, and target them to specific genomic loci to alter DNA methylation and the structure and modification of chromosomes [66].
Histone modifications affect chromatin structure, which plays an important role in gene regulation and carcinogenesis [67]. The most common changes observed in histone modification patterns in pancreatic cancer are methylation, acetylation, and ubiquitination. Multiple histone-modifying enzymes are involved in the dynamic regulation of these changes, and a balance between histone-modifying enzymes is critical for maintaining normal cellular function (Table 1). A common histone modification is methylation, which takes place on arginine, lysine, and histidine residues [68]. Lysine and arginine methyltransferases (KMTs and PRMTs) catalyze the transfer of the methyl group, while lysine demethylases (KDMs) control its removal. All methyltransferases use S-adenosyl methionine (SAM) as a substrate to transfer methyl groups onto the lysine and arginine residues of histones [69,70]. Histone acetylation is regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs). Acetylation can reduce the positive charge of lysine residues, leading to reduced binding between histone tails and negatively charged DNA, leaving the underlying DNA exposed [71]. Histone ubiquitination occurs primarily on H2A and H2B [72]. Ubiquitin (Ub) covalently binds to the target lysine residue under the continuous action of three proteins, the E1 activating enzyme, E2 conjugating enzyme, and E3 ligase [73]. E3 is essential and specifically required for histone ubiquitination [74]. Conversely, the deubiquitinating enzyme (DUB) is responsible for removing Ub [75].

4. Histone Modifications in Pancreatic Cancer EMT

4.1. Histone Methylation in EMT

4.1.1. Histone Methylases

PRMT1 in EMT: Protein arginine methyltransferase 1 (PRMT1) is the principal methyltransferase that catalyzes the methylation of H4R3 and functions as a transcription co-activator [147]. PRMT1 expression is increased in patients with pancreatic cancer, and higher expression is associated with poorer prognosis [77,79]. PRMT1 can bind to the β-catenin promoter region, increasing the expression of the β-catenin protein in pancreatic cancer cells [79]. Stimulation of the Wnt/β-catenin signaling pathway promotes pancreatic cancer proliferation, migration, and invasion while regulating therapeutic resistance [148,149,150,151]. Increased levels of β-catenin in the nucleus can promote EMT-related gene expression. Furthermore, β-catenin can form a complex with E-cadherin to regulate the intercellular connections that are important in cell–cell adhesions [25,152].
The Hh signaling pathway is aberrantly activated during tumorigenesis in the pancreas [153]. Hh signaling induces the expression of EMT-related genes, such as PTCH1, WNT, and SNAI1, by activating Gli transcription factors. The methylation of Gli1 by PRMT1 at R597 improves the capacity of Gli1 to bind to its target gene promoter, thereby enhancing the transcriptional activity of Gli1 [81]. Moreover, ZEB1, a zinc finger E-box binding homeobox transcription factor, is crucial for the EMT process and abnormal expression of ZEB1 has been reported in pancreatic cancer [128]. Overexpression of ZEB1 can significantly reverse the anti-tumor effects induced by PRMT1 downregulation [77].
PRMT5 in EMT: PRMT5 catalyzes the methylation of arginine on H2AR3, H4R3, and H3R8, and acts primarily as a tumor-promoting factor [147,154]. Patients with pancreatic cancer who display higher levels of PRMT5 expression present lower overall survival [83,84]. PRMT5 induces the phosphorylation of the epidermal growth factor receptor (EGFR), upregulates the expression of β-catenin through the EGFR/AKT/β-catenin pathway, and promotes the EMT of pancreatic cancer cells to enhance tumor migration and invasion [83]. EGFR belongs to the membrane-bound ErbB/HER family of receptor tyrosine kinases (RTKs) and plays an important role in the maintenance of epithelial tissues. When EGFR signaling is altered, it becomes the grand orchestrator of epithelial transformation and promotes EGF-induced EMT in pancreatic cancer [155].
SETD2 in EMT: SETD2 is the sole enzyme responsible for H3K36me3 [156]. SETD2 is downregulated in pancreatic cancer, and a low expression of SETD2 is linked to poor clinical prognosis [93,94]. During pancreatic carcinogenesis, loss of SETD2 promotes KRAS-driven acinar-to-ductal metaplasia, as well as EMT and metastasis through sustained AKT activation and loss of α-catenin [94].
KMT5 family in EMT: KMT5A (SET8/PR-SET7) and KMT5B/C (SUV4-20H1/2) belong to the KMT5 family of lysine methyltransferases. H4K20 is monomethylated by KMT5A, before stepwise methylation occurs through the function of KMT5B/C from H4K20me1 to H4K20me2/me3 [21,157]. KMT5A upregulates the expression of stemness and EMT-related genes in pancreatic cancer cells by inducing the expression of the receptor tyrosine kinase-like orphan receptor 1 (ROR1) [99]. ROR1 is a transmembrane protein that induces EMT in breast cancer by enhancing EGFR signaling [158,159]. KMT5C has no direct effect on EMT-related transcription factors but instead regulates EMT by regulating the expression of MET-related transcription factors such as FOXA1, OVOL1, and OVOL2. Targeting KMT5C can activate epithelial transcription programs and inhibit the invasion and migration of pancreatic cancer cells [101].
EZH2 in EMT: Enhancer of zeste homolog 2 (EZH2) is the enzymatic subunit of the polycomb repressor complex 2 (PRC2) that promotes transcriptional silencing by methylating H3K27 [160]. EZH2 is overexpressed in patients with pancreatic cancer and is associated with poorer clinical outcomes [104,106]. The best characterized pathway involved in the induction of EMT is mediated by TGF-β signaling. TGF-β stimulates the expression of the SRY-box transcription factor 4 (SOX4) protein, which is an important developmental transcription factor involved in the regulation of the TGF-β signal transduction pathway [161]. SOX4 not only regulates the expression of EMT-related genes but also reprograms the epigenome to induce EMT by inducing EZH2 expression [162]. In pancreatic cancer, increased expression of SOX4 and EZH2 is associated with poor patient prognosis [116]. The downregulation of E-cadherin is a crucial step in EMT, which promotes metastasis by boosting cancer cell invasion and dissociation. EZH2 can stimulate the migration and invasion of pancreatic cancer by inhibiting the expression of E-cadherin [106]. EZH2 may achieve this through its interaction with the lncRNA MALAT-1, which promotes the proliferation and metastasis of pancreatic cancer [163].

4.1.2. Histone Demethylases

KDM2B in EMT: KDM2B, also known as Ndy1, FBXL10, and JHDM1B, demethylates H3K36 and is overexpressed in human pancreatic cancer cells [164]. A core component of the Hippo signaling pathway is the adaptor protein MOB1 [165], which functions to depress Hippo transcriptional activity by increasing the phosphorylation of the yes-associated protein 1 (YAP) and tafazzin (TAZ) proteins. KDM2B transcriptionally inhibits the expression of MOB1 and promotes pancreatic cancer migration and invasion by promoting Hippo signaling [112]. KDM2B also inhibits the expression of several epithelial marker genes such as CDH1, miR200a, and CGN by modulating histone H1A ubiquitination [109].
KDM3A in EMT: KDM3A catalyzes the demethylation of H3K9me1/me2, which mediates transcriptional activation [166]. KDM3A mediates the upregulation of doublecortin-like kinase 1 (DCLK1), which is critical for the development and progression of pancreatic cancer during hypoxia. Under hypoxia, HIF1α activates KDM3A, which, in turn, increases DCLK1 mRNA expression [113]. DCLK1 activates EMT and promotes the migration and invasion of pancreatic cancer cells [167], while knockdown of DCLK1 reduces the expression of EMT transcription factors in these cells [168].
KDM4B in EMT: KDM4B is a member of the jumonji domain 2 (JMJD2) demethylase family [169], which catalyzes the demethylation of H3K9 and H3K36 [170]. KDM4B positively regulates the EMT process by activating the transcription of ZEB1, which plays a key role in TGF-β-induced EMT [116].
KDM5A in EMT: KDM5A, also known as JARID1A or RBP2, can remove the lysine of H3K4me2 and H3K4me3, resulting in activation or inhibition of transcription. KDM5A is overexpressed in pancreatic cancer cells, and is a member of the JMJC family of oxygen-sensitive enzymes [118,119,171]. KDM5A can be blocked by increased NOX4, which is the main endogenous source of reactive oxygen species (ROS). In pancreatic cancer cells under hypoxia, NOX4 expression is upregulated [172]. NOX4 regulates SNAIL1 transcription to induce EMT and promote the invasion and metastasis of pancreatic cancer cells [117].
KDM6A in EMT: KDM6A, also known as UTX, specifically catalyzes the demethylation of H3K27me3 [173] and is downregulated in pancreatic cancer cells [120,121,122]. The transcriptional activator hepatocyte nuclear factor-1a (HNF-1a) is thought to be a key tumor suppressor in pancreatic cancer [174], and recruits KDM6A to indirectly inhibit the expression of genes involved in tumorigenesis and EMT [122,123]. In pancreatic cancer cells, reductions in KDM6A lead to the increased expression of activin A, a member of the transforming growth factor-β superfamily of cytokines that activates a noncanonical p38 MAPK pathway to induce mesenchymal identity [120] and promote EMT [175,176].

4.2. Histone Acetylation in EMT

4.2.1. Histone Acetylases

The CREB-binding protein (CBP) and p300 mediate histone lysine acetylation. Due to their considerable sequence homology and functional overlap, they are often referred to as a single entity (CBP/p300) [177]. CBP/p300 activates gene transcription through the acetylation of histone H3 lysine 27 (H3K27ac) [177]. Aberrant expression of p300 has been shown in pancreatic cancer cells [178]. p300 maintains the expression of the GATA-binding factor 6 (GATA6), thereby affecting the differentiation program regulated by this transcription factor [179]. GATA6 directly and indirectly inhibits dedifferentiation and EMT in pancreatic ductal adenocarcinoma (PDAC) [180]. P300/CBP-associated factor (PCAF) is a member of the GCN5-related N-acetyltransferase family that promotes transcription as a histone acetyltransferase [181]. PCAF/p300 has a synergistic effect in regulating the transcriptional repression/activation of ZEB1, forming the P300/PCAF/ZEB1 complex on the miR200c/141 promoter. PCAF/P300 induces the lysine acetylation of ZEB1 to activate miR200c transcription [182]. Loss of miR-200 family members and the upregulation of ZEB1 and ZEB2 are involved in the regulation of EMT. Transcriptional repression of the miR-200 family by ZEB1/ZEB2 is a major factor in the maintenance of mesenchymal characteristics and the induction of EMT [183]. Moreover, PCAF functions in the Ras ERK1/2 pathway and promotes the motility of pancreatic cancer cells, suggesting that PCAF is involved in pancreatic cancer EMT through multiple pathways [184].

4.2.2. Histone Deacetylases

HDACs prevent gene expression. In pancreatic cancer, HDACs regulate proliferation, apoptosis, and metastasis [185]. High levels of HDAC1 and HDAC2 expression are linked to distant pancreatic cancer transitions, and both proteins enhance tumor invasiveness [129]. Pancreatic cancer cells proliferate and migrate more readily because of HDAC1 and HDAC2-mediated inhibition of E-cadherin expression in tumor cells [127,128]. HIF-1α plays a vital role as a transcriptional regulator of hypoxia-induced EMT, and a significant correlation exists between HIF-1α and HDAC1 expression in pancreatic cancer. In addition, high levels of these proteins are associated with a poorer prognosis for malignancies in patients, which may suggest that HDAC1 mediates HIF-1α stability through epigenetic regulation [186].
SIRT1 is a mammalian NAD+-dependent class III HDAC, which can regulate the Wnt/β-catenin signaling pathway by deacetylating β-catenin [187]. In the acinar-to-ductal metaplasia (ADM) of the pancreas, SIRT1 interferes with Wnt/β-catenin signaling by regulating the acetylation of β-catenin [133].

4.3. Histone Ubiquitination in EMT

4.3.1. Histone Ubiquitinases

K119 is the most common ubiquitination checkpoint for H2A. The main E3 ubiquitin ligase of H2AK119ub1 is the catalytic subunit of the polycomb inhibitory complex (PRC1) [72]. This subunit consists of Ring1A and Ring1B and is regulated by Bmi1 (also known as PCGF4) [188]. The transcriptional repressor Snail can induce EMT by inhibiting the transcription of E-cadherin. In pancreatic cancer, Ring1A and Ring1B are critical for this regulatory process. Furthermore, EZH2 promotes Snail to recruit Ring1A and Ring1B more efficiently [189]. Concomitantly, Bmi-1 is overexpressed in pancreatic cancer, and this dysregulation is associated with abnormal expression of miRNAs. Overexpression of Bmi-1 can also promote EMT by downregulating E-cadherin [138,190].

4.3.2. Histone Deubiquitinases

The ubiquitin-specific peptidases (USPs) are the primary members of the deubiquitinase family [191], where USP22 and USP28 are overexpressed in pancreatic cancer cells [139,140,141,142,143,144,145,146]. USP22 regulates the levels of H2Bub1 and H2Aub1 through deubiquitination, thereby promoting the transcription of downstream genes [192]. Focal adhesion kinase (FAK) is a cytoplasmic tyrosine kinase that mediates a variety of signal transduction pathways [193]. USP22 induces the occurrence of EMT in pancreatic cancer cells by activating FAK signaling. Upregulation of USP22 can increase the expression of the ZEB1 and Snail transcription factors, significantly reduce the expression of E-cadherin at cell–cell junctions, and upregulate the expression of mesenchymal markers [143].
USP28 regulates the process of histone H2A deubiquitination [194]. USP28 deubiquitinates and stabilizes the forkhead box M1 (FOXM1) transcription factor, which, in turn, activates the Wnt/β-catenin pathway and subsequently the expression of many EMT-related genes [146,195]. As a key mediator of Wnt/β-catenin signaling, FOXM1 mediates the nuclear accumulation of β-catenin and the ensuing downstream target gene expression in pancreatic cancer cells [196].

5. Histone Epigenetic Modulators in Pancreatic Cancer

Our understanding of the therapeutic potential of drugs targeting epigenetic modifiers has grown immensely in recent years. Histone modifiers are attractive targets for prospective therapy because they contain unique, druggable catalytic domains. Some FDA-approved inhibitors are already used in clinical practice, and others are currently in clinical trials [197]. Indeed, several drugs targeting EZH2 have been developed [198]. 3-Denitrogen-neplanocin A (DZNeP) and GSK126 are two newly discovered inhibitors of EZH2. Combination therapy of DZNeP and gemcitabine can act synergistically to inhibit pancreatic tumor cell growth and migration. DZNeP enhances the anti-proliferation activity of gemcitabine and significantly promotes apoptosis [199]. Similarly, GSK126 and BET bromine domain inhibitors have synergistic antitumor effects in the treatment of pancreatic cancer [200]. Thus, in combination with other therapeutics, EZH2 inhibitors possess great potential in the treatment of pancreatic cancer.
Histone deacetylase inhibitors (HDACIs) are becoming an important class of therapeutics in the treatment of pancreatic cancer. In preclinical studies, HDACIs have shown significant antitumor potential and low toxicity. HDACIs exert antitumor effects by stalling the cell cycle, activating apoptosis, inhibiting angiogenesis, and inhibiting metastasis [201]. However, when used alone, HDACIs are not as effective in the treatment of solid tumors. Many studies have now shown that HDACIs have synergistic effects with traditional cytotoxic and targeted therapeutic drugs against pancreatic cancer. The small molecule Domatinostat (4SC-202) is a class I selective HDACI that induces antitumor effects and sensitization to chemotherapy. It functions by reducing the expression of the transcription factor FOXM1, which impairs the redox homeostasis of cancer stem cells [202]. MPT0E028 is a novel pan-HDACI that targets both classes I and II HDACs. The co-administration of MPT0E028 and mitogen-activated protein kinase (MEK) inhibitors has synergistic effects in KRAS-mutated and KRAS-wild-type pancreatic cancer cells. This combination therapy induces a strong apoptotic response and significantly reduces cancer cell viability [203]. Additionally, the combination of radiation therapy with either CUDC-101 (simultaneously inhibits targets such as HDAC, EGFR, and HER2) or SAHA (inhibits class I and class II HDACs) could enhance radiation-induced cytotoxicity in human pancreatic cells [204].
Close interactions exist between the various histone epigenetic regulators, and many can be combined to synergistically regulate transcription. The use of dual inhibitors targeting different epigenetic regulators is an effective strategy to improve the safety and efficacy of single epigenetic target drugs and to overcome drug resistance. UDC-907 is a dual inhibitor of HDAC and PI3K that inactivates RAF-MEK-ERK signaling in pancreatic cancer cells by inhibiting the PI3K-AKT-mTOR pathway [205]. Metavert is a dual inhibitor of GSK3β and HDAC that induces cancer cell apoptosis and reduces both migration and the expression of stem cell markers, thereby slowing tumor and metastatic growth. Metavert has also been shown to provide synergistic effects with gemcitabine [206]. A new and effective strategy for the treatment of pancreatic cancer is 13A, a dual BET/HDAC inhibitor. 13A is more effective against pancreatic cancer than either BET inhibitors or HDAC inhibitors alone or when they are combined [207]. Finally, XP-524 is a dual-BET/EP300 inhibitor that prevents KRAS-induced tumor transformation. XP-524 can bind to anti-PD-1 antibodies, which reactivates the cytotoxic immune response [208].

6. Conclusions

The role of EMT in pancreatic cancer and the molecular mechanisms by which histone modifications regulate EMT-related molecules have been the subject of intense research over the past few years. The EMT process governs both physiological and pathological development, and abnormally active EMT phenotypes have been associated with pancreatic cancer initiation, progression, and therapeutic resistance. There is a now a large body of evidence demonstrating that many histone changes can reversibly control the expression of EMT markers during EMT and MET. Inhibitors of histone modification enzymes have been shown to raise the sensitivity of pancreatic cancer cells to chemotherapy and immunotherapy, thereby providing potential therapeutics. Combining histone modification enzyme inhibitors with established anti-tumor drugs is a promising treatment method. Thus, the elucidation of the molecular mechanisms by which histone-modifying enzymes regulate EMT in pancreatic cancers will enhance our understanding of the underlying tumorigenesis and metastatic progression, thereby facilitating the development of alternative treatments that may have important implications for disease outcomes.

Author Contributions

Conceptualization, Y.X.; writing—original draft preparation, Y.X.; writing—review and editing, Q.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by 1.3.5 Project for Disciplines of Excellence, West China Hospital (ZYJC21042), Sichuan University for Qing Zhu.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Bengtsson, A.; Andersson, R.; Ansari, D. The actual 5-year survivors of pancreatic ductal adenocarcinoma based on real-world data. Sci. Rep. 2020, 10, 16425. [Google Scholar] [CrossRef] [PubMed]
  3. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  4. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [Green Version]
  5. Gobbi, P.G.; Bergonzi, M.; Comelli, M.; Villano, L.; Pozzoli, D.; Vanoli, A.; Dionigi, P. The prognostic role of time to diagnosis and presenting symptoms in patients with pancreatic cancer. Cancer Epidemiol. 2013, 37, 186–190. [Google Scholar] [CrossRef]
  6. Grossberg, A.J.; Chu, L.C.; Deig, C.R.; Fishman, E.K.; Hwang, W.L.; Maitra, A.; Marks, D.L.; Mehta, A.; Nabavizadeh, N.; Simeone, D.M.; et al. Multidisciplinary standards of care and recent progress in pancreatic ductal adenocarcinoma. CA Cancer J. Clin. 2020, 70, 375–403. [Google Scholar] [CrossRef]
  7. Tempero, M.A.; Malafa, M.P.; Al-Hawary, M.; Behrman, S.W.; Benson, A.B.; Cardin, D.B.; Chiorean, E.G.; Chung, V.; Czito, B.; Del Chiaro, M.; et al. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2021, 19, 439–457. [Google Scholar] [CrossRef]
  8. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
  9. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell. Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
  10. Du, B.; Shim, J.S. Targeting Epithelial-Mesenchymal Transition (EMT) to Overcome Drug Resistance in Cancer. Molecules 2016, 21, 965. [Google Scholar] [CrossRef] [Green Version]
  11. 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] [Green Version]
  12. Brabletz, S.; Schuhwerk, H.; Brabletz, T.; Stemmler, M.P. Dynamic EMT: A multi-tool for tumor progression. EMBO J. 2021, 40, e108647. [Google Scholar] [CrossRef] [PubMed]
  13. Aiello, N.M.; Brabletz, T.; Kang, Y.; Nieto, M.A.; Weinberg, R.A.; Stanger, B.Z. Upholding a role for EMT in pancreatic cancer metastasis. Nature 2017, 547, E7–E8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Luu, T. Epithelial-Mesenchymal Transition and Its Regulation Mechanisms in Pancreatic Cancer. Front. Oncol. 2021, 11, 646399. [Google Scholar] [CrossRef] [PubMed]
  15. Rhim, A.D.; Mirek, E.T.; Aiello, N.M.; Maitra, A.; Bailey, J.M.; McAllister, F.; Reichert, M.; Beatty, G.L.; Rustgi, A.K.; Vonderheide, R.H.; et al. EMT and dissemination precede pancreatic tumor formation. Cell 2012, 148, 349–361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Loh, C.Y.; Chai, J.Y.; Tang, T.F.; Wong, W.F.; Sethi, G.; Shanmugam, M.K.; Chong, P.P.; Looi, C.Y. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019, 8, 1118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Bakir, B.; Chiarella, A.M.; Pitarresi, J.R.; Rustgi, A.K. EMT, MET, Plasticity, and Tumor Metastasis. Trends Cell Biol. 2020, 30, 764–776. [Google Scholar] [CrossRef] [PubMed]
  18. Tam, W.L.; Weinberg, R.A. The epigenetics of epithelial-mesenchymal plasticity in cancer. Nat. Med. 2013, 19, 1438–1449. [Google Scholar] [CrossRef] [Green Version]
  19. Dawson, M.A.; Kouzarides, T. Cancer epigenetics: From mechanism to therapy. Cell 2012, 150, 12–27. [Google Scholar] [CrossRef] [Green Version]
  20. Recillas-Targa, F. Cancer Epigenetics: An Overview. Arch. Med. Res. 2022, 53, 732–740. [Google Scholar] [CrossRef]
  21. Zhang, Y.; Sun, Z.; Jia, J.; Du, T.; Zhang, N.; Tang, Y.; Fang, Y.; Fang, D. Overview of Histone Modification. Adv. Exp. Med. Biol. 2021, 1283, 1–16. [Google Scholar] [PubMed]
  22. Garcia, M.A.; Nelson, W.J.; Chavez, N. Cell-Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb. Perspect. Biol. 2018, 10, a029181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Jayachandran, J.; Srinivasan, H.; Mani, K.P. Molecular mechanism involved in epithelial to mesenchymal transition. Arch. Biochem. Biophys. 2021, 710, 108984. [Google Scholar] [CrossRef]
  24. Wong, S.H.M.; Fang, C.M.; Chuah, L.H.; Leong, C.O.; Ngai, S.C. E-cadherin: Its dysregulation in carcinogenesis and clinical implications. Crit. Rev. Oncol. Hematol. 2018, 121, 11–22. [Google Scholar] [CrossRef] [PubMed]
  25. Bruner, H.C.; Derksen, P.W.B. Loss of E-Cadherin-Dependent Cell-Cell Adhesion and the Development and Progression of Cancer. Cold Spring Harb. Perspect. Biol. 2018, 10, a029330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sommariva, M.; Gagliano, N. E-Cadherin in Pancreatic Ductal Adenocarcinoma: A Multifaceted Actor during EMT. Cells 2020, 9, 1040. [Google Scholar] [CrossRef]
  27. Hao, Y.; Baker, D.; Ten Dijke, P. TGF-beta-Mediated Epithelial-Mesenchymal Transition and Cancer Metastasis. Int. J. Mol. Sci. 2019, 20, 2767. [Google Scholar] [CrossRef] [Green Version]
  28. Aashaq, S.; Batool, A.; Mir, S.A.; Beigh, M.A.; Andrabi, K.I.; Shah, Z.A. TGF-beta signaling: A recap of SMAD-independent and SMAD-dependent pathways. J. Cell Physiol. 2022, 237, 59–85. [Google Scholar] [CrossRef]
  29. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/beta-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal. Transduct. Target Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  30. Doheny, D.; Manore, S.G.; Wong, G.L.; Lo, H.W. Hedgehog Signaling and Truncated GLI1 in Cancer. Cells 2020, 9, 2114. [Google Scholar] [CrossRef]
  31. Sheng, W.; Shi, X.; Lin, Y.; Tang, J.; Jia, C.; Cao, R.; Sun, J.; Wang, G.; Zhou, L.; Dong, M. Musashi2 promotes EGF-induced EMT in pancreatic cancer via ZEB1-ERK/MAPK signaling. J. Exp. Clin. Cancer Res. 2020, 39, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Sheng, W.; Chen, C.; Dong, M.; Wang, G.; Zhou, J.; Song, H.; Li, Y.; Zhang, J.; Ding, S. Calreticulin promotes EGF-induced EMT in pancreatic cancer cells via Integrin/EGFR-ERK/MAPK signaling pathway. Cell. Death Dis. 2017, 8, e3147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Debnath, P.; Huirem, R.S.; Dutta, P.; Palchaudhuri, S. Epithelial-mesenchymal transition and its transcription factors. Biosci. Rep. 2022, 42, BSR20211754. [Google Scholar] [CrossRef] [PubMed]
  34. Ho, W.J.; Jaffee, E.M.; Zheng, L. The tumour microenvironment in pancreatic cancer—Clinical challenges and opportunities. Nat. Rev. Clin. Oncol. 2020, 17, 527–540. [Google Scholar] [CrossRef]
  35. Erin, N.; Grahovac, J.; Brozovic, A.; Efferth, T. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist. Updat. 2020, 53, 100715. [Google Scholar] [CrossRef]
  36. Chattopadhyay, I.; Ambati, R.; Gundamaraju, R. Exploring the Crosstalk between Inflammation and Epithelial-Mesenchymal Transition in Cancer. Mediat. Inflamm. 2021, 2021, 9918379. [Google Scholar] [CrossRef]
  37. Hamada, S.; Masamune, A.; Yoshida, N.; Takikawa, T.; Shimosegawa, T. IL-6/STAT3 Plays a Regulatory Role in the Interaction between Pancreatic Stellate Cells and Cancer Cells. Dig. Dis. Sci. 2016, 61, 1561–1571. [Google Scholar] [CrossRef]
  38. Ren, B.; Cui, M.; Yang, G.; Wang, H.; Feng, M.; You, L.; Zhao, Y. Tumor microenvironment participates in metastasis of pancreatic cancer. Mol. Cancer 2018, 17, 108. [Google Scholar] [CrossRef] [Green Version]
  39. Tao, J.; Yang, G.; Zhou, W.; Qiu, J.; Chen, G.; Luo, W.; Zhao, F.; You, L.; Zheng, L.; Zhang, T.; et al. Targeting hypoxic tumor microenvironment in pancreatic cancer. J. Hematol. Oncol. 2021, 14, 14. [Google Scholar] [CrossRef]
  40. Cowman, S.J.; Koh, M.Y. Revisiting the HIF switch in the tumor and its immune microenvironment. Trends Cancer 2022, 8, 28–42. [Google Scholar] [CrossRef]
  41. Cheng, Z.X.; Wang, D.W.; Liu, T.; Liu, W.X.; Xia, W.B.; Xu, J.; Zhang, Y.H.; Qu, Y.K.; Guo, L.Q.; Ding, L.; et al. Effects of the HIF-1alpha and NF-kappaB loop on epithelial-mesenchymal transition and chemoresistance induced by hypoxia in pancreatic cancer cells. Oncol. Rep. 2014, 31, 1891–1898. [Google Scholar] [CrossRef] [Green Version]
  42. Yang, J.; Zhang, X.; Zhang, Y.; Zhu, D.; Zhang, L.; Li, Y.; Zhu, Y.; Li, D.; Zhou, J. HIF-2alpha promotes epithelial-mesenchymal transition through regulating Twist2 binding to the promoter of E-cadherin in pancreatic cancer. J. Exp. Clin. Cancer Res. 2016, 35, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Wood, L.D.; Canto, M.I.; Jaffee, E.M.; Simeone, D.M. Pancreatic Cancer: Pathogenesis, Screening, Diagnosis, and Treatment. Gastroenterology 2022, 163, 386–402 e1. [Google Scholar] [CrossRef] [PubMed]
  44. Singh, A.; Greninger, P.; Rhodes, D.; Koopman, L.; Violette, S.; Bardeesy, N.; Settleman, J. A gene expression signature associated with ’’K-Ras addiction’’ reveals regulators of EMT and tumor cell survival. Cancer Cell 2021, 39, 441–442. [Google Scholar] [CrossRef] [PubMed]
  45. Shao, D.D.; Xue, W.; Krall, E.B.; Bhutkar, A.; Piccioni, F.; Wang, X.; Schinzel, A.C.; Sood, S.; Rosenbluh, J.; Kim, J.W.; et al. KRAS and YAP1 converge to regulate EMT and tumor survival. Cell 2014, 158, 171–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Zhang, Y.; Liu, J.L.; Wang, J. KRAS gene silencing inhibits the activation of PI3K-Akt-mTOR signaling pathway to regulate breast cancer cell epithelial-mesenchymal transition, proliferation and apoptosis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 3085–3096. [Google Scholar]
  47. Dardare, J.; Witz, A.; Merlin, J.L.; Gilson, P.; Harle, A. SMAD4 and the TGFbeta Pathway in Patients with Pancreatic Ductal Adenocarcinoma. Int. J. Mol. Sci. 2020, 21, 3534. [Google Scholar] [CrossRef]
  48. Racu, M.L.; Lebrun, L.; Schiavo, A.A.; Van Campenhout, C.; De Clercq, S.; Absil, L.; Minguijon Perez, E.; Maris, C.; Decaestecker, C.; Salmon, I.; et al. The Role of SMAD4 Inactivation in Epithelial-Mesenchymal Plasticity of Pancreatic Ductal Adenocarcinoma: The Missing Link? Cancers 2022, 14, 973. [Google Scholar] [CrossRef]
  49. David, C.J.; Huang, Y.H.; Chen, M.; Su, J.; Zou, Y.; Bardeesy, N.; Iacobuzio-Donahue, C.A.; Massague, J. TGF-beta Tumor Suppression through a Lethal EMT. Cell 2016, 164, 1015–1030. [Google Scholar] [CrossRef] [Green Version]
  50. Mohd Faheem, M.; Rasool, R.U.; Ahmad, S.M.; Jamwal, V.L.; Chakraborty, S.; Katoch, A.; Gandhi, S.G.; Bhagat, M.; Goswami, A. Par-4 mediated Smad4 induction in PDAC cells restores canonical TGF-beta/ Smad4 axis driving the cells towards lethal EMT. Eur. J. Cell Biol. 2020, 99, 151076. [Google Scholar] [CrossRef]
  51. Kang, Y.; Ling, J.; Suzuki, R.; Roife, D.; Chopin-Laly, X.; Truty, M.J.; Chatterjee, D.; Wang, H.; Thomas, R.M.; Katz, M.H.; et al. SMAD4 regulates cell motility through transcription of N-cadherin in human pancreatic ductal epithelium. PLoS ONE 2014, 9, e107948. [Google Scholar] [CrossRef] [PubMed]
  52. Yamada, S.; Fujii, T.; Shimoyama, Y.; Kanda, M.; Nakayama, G.; Sugimoto, H.; Koike, M.; Nomoto, S.; Fujiwara, M.; Nakao, A.; et al. SMAD4 expression predicts local spread and treatment failure in resected pancreatic cancer. Pancreas 2015, 44, 660–664. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, Z.; Li, Y.; Zhan, S.; Zhang, L.; Zhang, S.; Tang, Q.; Li, M.; Tan, Z.; Liu, S.; Xing, X. SMAD4 Y353C promotes the progression of PDAC. BMC Cancer 2019, 19, 1037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef]
  55. Nakano, M.; Kikushige, Y.; Miyawaki, K.; Kunisaki, Y.; Mizuno, S.; Takenaka, K.; Tamura, S.; Okumura, Y.; Ito, M.; Ariyama, H.; et al. Dedifferentiation process driven by TGF-beta signaling enhances stem cell properties in human colorectal cancer. Oncogene 2019, 38, 780–793. [Google Scholar] [CrossRef]
  56. Ji, C.; Yang, L.; Yi, W.; Xiang, D.; Wang, Y.; Zhou, Z.; Qian, F.; Ren, Y.; Cui, W.; Zhang, X.; et al. Capillary morphogenesis gene 2 maintains gastric cancer stem-like cell phenotype by activating a Wnt/beta-catenin pathway. Oncogene 2018, 37, 3953–3966. [Google Scholar] [CrossRef] [Green Version]
  57. Chen, C.; Okita, Y.; Watanabe, Y.; Abe, F.; Fikry, M.A.; Ichikawa, Y.; Suzuki, H.; Shibuya, A.; Kato, M. Glycoprotein nmb Is Exposed on the Surface of Dormant Breast Cancer Cells and Induces Stem Cell-like Properties. Cancer Res. 2018, 78, 6424–6435. [Google Scholar] [CrossRef] [Green Version]
  58. Lonardo, E.; Hermann, P.C.; Mueller, M.T.; Huber, S.; Balic, A.; Miranda-Lorenzo, I.; Zagorac, S.; Alcala, S.; Rodriguez-Arabaolaza, I.; Ramirez, J.C.; et al. Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy. Cell Stem. Cell 2011, 9, 433–446. [Google Scholar] [CrossRef] [Green Version]
  59. Mueller, M.T.; Hermann, P.C.; Witthauer, J.; Rubio-Viqueira, B.; Leicht, S.F.; Huber, S.; Ellwart, J.W.; Mustafa, M.; Bartenstein, P.; D’Haese, J.G.; et al. Combined targeted treatment to eliminate tumorigenic cancer stem cells in human pancreatic cancer. Gastroenterology 2009, 137, 1102–1113. [Google Scholar] [CrossRef]
  60. Barman, S.; Fatima, I.; Singh, A.B.; Dhawan, P. Pancreatic Cancer and Therapy: Role and Regulation of Cancer Stem Cells. Int. J. Mol. Sci. 2021, 22, 4765. [Google Scholar] [CrossRef]
  61. Wang, F.; Ma, L.; Zhang, Z.; Liu, X.; Gao, H.; Zhuang, Y.; Yang, P.; Kornmann, M.; Tian, X.; Yang, Y. Hedgehog Signaling Regulates Epithelial-Mesenchymal Transition in Pancreatic Cancer Stem-Like Cells. J. Cancer 2016, 7, 408–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Ding, Q.; Miyazaki, Y.; Tsukasa, K.; Matsubara, S.; Yoshimitsu, M.; Takao, S. CD133 facilitates epithelial-mesenchymal transition through interaction with the ERK pathway in pancreatic cancer metastasis. Mol. Cancer 2014, 13, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Onufriev, A.V.; Schiessel, H. The nucleosome: From structure to function through physics. Curr. Opin. Struct. Biol. 2019, 56, 119–130. [Google Scholar] [CrossRef] [PubMed]
  64. Lyko, F. The DNA methyltransferase family: A versatile toolkit for epigenetic regulation. Nat. Rev. Genet. 2018, 19, 81–92. [Google Scholar] [CrossRef]
  65. Nodelman, I.M.; Bowman, G.D. Biophysics of Chromatin Remodeling. Annu. Rev. Biophys. 2021, 50, 73–93. [Google Scholar] [CrossRef]
  66. Chen, J.; Xue, Y. Emerging roles of non-coding RNAs in epigenetic regulation. Sci. China Life Sci. 2016, 59, 227–235. [Google Scholar] [CrossRef] [Green Version]
  67. Millan-Zambrano, G.; Burton, A.; Bannister, A.J.; Schneider, R. Histone post-translational modifications—Cause and consequence of genome function. Nat. Rev. Genet. 2022, 23, 563–580. [Google Scholar] [CrossRef]
  68. Chen, Y.; Ren, B.; Yang, J.; Wang, H.; Yang, G.; Xu, R.; You, L.; Zhao, Y. The role of histone methylation in the development of digestive cancers: A potential direction for cancer management. Signal. Transduct. Target Ther. 2020, 5, 143. [Google Scholar] [CrossRef]
  69. Song, Y.; Wu, F.; Wu, J. Targeting histone methylation for cancer therapy: Enzymes, inhibitors, biological activity and perspectives. J. Hematol. Oncol. 2016, 9, 49. [Google Scholar] [CrossRef] [Green Version]
  70. Bhat, K.P.; Umit Kaniskan, H.; Jin, J.; Gozani, O. Epigenetics and beyond: Targeting writers of protein lysine methylation to treat disease. Nat. Rev. Drug Discov. 2021, 20, 265–286. [Google Scholar] [CrossRef]
  71. Shvedunova, M.; Akhtar, A. Modulation of cellular processes by histone and non-histone protein acetylation. Nat. Rev. Mol. Cell Biol. 2022, 23, 329–349. [Google Scholar] [CrossRef] [PubMed]
  72. Mattiroli, F.; Penengo, L. Histone Ubiquitination: An Integrative Signaling Platform in Genome Stability. Trends Genet. 2021, 37, 566–581. [Google Scholar] [CrossRef] [PubMed]
  73. Salas-Lloret, D.; Gonzalez-Prieto, R. Insights in Post-Translational Modifications: Ubiquitin and SUMO. Int. J. Mol. Sci. 2022, 23, 3281. [Google Scholar] [CrossRef] [PubMed]
  74. Buetow, L.; Huang, D.T. Structural insights into the catalysis and regulation of E3 ubiquitin ligases. Nat. Rev. Mol. Cell Biol. 2016, 17, 626–642. [Google Scholar] [CrossRef] [Green Version]
  75. Mevissen, T.E.T.; Komander, D. Mechanisms of Deubiquitinase Specificity and Regulation. Annu. Rev. Biochem. 2017, 86, 159–192. [Google Scholar] [CrossRef] [Green Version]
  76. Giuliani, V.; Miller, M.A.; Liu, C.Y.; Hartono, S.R.; Class, C.A.; Bristow, C.A.; Suzuki, E.; Sanz, L.A.; Gao, G.; Gay, J.P.; et al. PRMT1-dependent regulation of RNA metabolism and DNA damage response sustains pancreatic ductal adenocarcinoma. Nat. Commun. 2021, 12, 4626. [Google Scholar] [CrossRef]
  77. Lin, Z.; Chen, Y.; Lin, Z.; Chen, C.; Dong, Y. Overexpressing PRMT1 Inhibits Proliferation and Invasion in Pancreatic Cancer by Inverse Correlation of ZEB1. IUBMB Life 2018, 70, 1032–1039. [Google Scholar] [CrossRef] [Green Version]
  78. Repenning, A.; Happel, D.; Bouchard, C.; Meixner, M.; Verel-Yilmaz, Y.; Raifer, H.; Holembowski, L.; Krause, E.; Kremmer, E.; Feederle, R.; et al. PRMT1 promotes the tumor suppressor function of p14(ARF) and is indicative for pancreatic cancer prognosis. EMBO J. 2021, 40, e106777. [Google Scholar] [CrossRef]
  79. Song, C.; Chen, T.; He, L.; Ma, N.; Li, J.A.; Rong, Y.F.; Fang, Y.; Liu, M.; Xie, D.; Lou, W. PRMT1 promotes pancreatic cancer growth and predicts poor prognosis. Cell Oncol. 2020, 43, 51–62. [Google Scholar] [CrossRef]
  80. Wang, L.; Jia, Z.; Xie, D.; Zhao, T.; Tan, Z.; Zhang, S.; Kong, F.; Wei, D.; Xie, K. Methylation of HSP70 Orchestrates Its Binding to and Stabilization of BCL2 mRNA and Renders Pancreatic Cancer Cells Resistant to Therapeutics. Cancer Res. 2020, 80, 4500–4513. [Google Scholar] [CrossRef]
  81. Wang, Y.; Hsu, J.M.; Kang, Y.; Wei, Y.; Lee, P.C.; Chang, S.J.; Hsu, Y.H.; Hsu, J.L.; Wang, H.L.; Chang, W.C.; et al. Oncogenic Functions of Gli1 in Pancreatic Adenocarcinoma Are Supported by Its PRMT1-Mediated Methylation. Cancer Res. 2016, 76, 7049–7058. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Zheng, N.N.; Zhou, M.; Sun, F.; Huai, M.X.; Zhang, Y.; Qu, C.Y.; Shen, F.; Xu, L.M. Combining protein arginine methyltransferase inhibitor and anti-programmed death-ligand-1 inhibits pancreatic cancer progression. World J. Gastroenterol. 2020, 26, 3737–3749. [Google Scholar] [CrossRef] [PubMed]
  83. Ge, L.; Wang, H.; Xu, X.; Zhou, Z.; He, J.; Peng, W.; Du, F.; Zhang, Y.; Gong, A.; Xu, M. PRMT5 promotes epithelial-mesenchymal transition via EGFR-beta-catenin axis in pancreatic cancer cells. J. Cell Mol. Med. 2020, 24, 1969–1979. [Google Scholar] [CrossRef] [Green Version]
  84. Qin, Y.; Hu, Q.; Xu, J.; Ji, S.; Dai, W.; Liu, W.; Xu, W.; Sun, Q.; Zhang, Z.; Ni, Q.; et al. PRMT5 enhances tumorigenicity and glycolysis in pancreatic cancer via the FBW7/cMyc axis. Cell Commun. Signal. 2019, 17, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wei, X.; Yang, J.; Adair, S.J.; Ozturk, H.; Kuscu, C.; Lee, K.Y.; Kane, W.J.; O’Hara, P.E.; Liu, D.; Demirlenk, Y.M.; et al. Targeted CRISPR screening identifies PRMT5 as synthetic lethality combinatorial target with gemcitabine in pancreatic cancer cells. Proc. Natl. Acad. Sci. USA 2020, 117, 28068–28079. [Google Scholar] [CrossRef]
  86. Orben, F.; Lankes, K.; Schneeweis, C.; Hassan, Z.; Jakubowsky, H.; Krauss, L.; Boniolo, F.; Schneider, C.; Schafer, A.; Murr, J.; et al. Epigenetic drug screening defines a PRMT5 inhibitor-sensitive pancreatic cancer subtype. JCI Insight 2022, 7, e151353. [Google Scholar] [CrossRef] [PubMed]
  87. Kato, H.; Tateishi, K.; Fujiwara, H.; Ijichi, H.; Yamamoto, K.; Nakatsuka, T.; Kakiuchi, M.; Sano, M.; Kudo, Y.; Hayakawa, Y.; et al. Deletion of Histone Methyltransferase G9a Suppresses Mutant Kras-driven Pancreatic Carcinogenesis. Cancer Genom. Proteom. 2020, 17, 695–705. [Google Scholar] [CrossRef]
  88. Pan, M.R.; Hsu, M.C.; Luo, C.W.; Chen, L.T.; Shan, Y.S.; Hung, W.C. The histone methyltransferase G9a as a therapeutic target to override gemcitabine resistance in pancreatic cancer. Oncotarget 2016, 7, 61136–61151. [Google Scholar] [CrossRef] [Green Version]
  89. Urrutia, G.; de Assuncao, T.M.; Mathison, A.J.; Salmonson, A.; Kerketta, R.; Zeighami, A.; Stodola, T.J.; Adsay, V.; Pehlivanoglu, B.; Dwinell, M.B.; et al. Inactivation of the Euchromatic Histone-Lysine N-Methyltransferase 2 Pathway in Pancreatic Epithelial Cells Antagonizes Cancer Initiation and Pancreatitis-Associated Promotion by Altering Growth and Immune Gene Expression Networks. Front. Cell Dev. Biol. 2021, 9, 681153. [Google Scholar] [CrossRef]
  90. Tian, Y.F.; Wang, H.C.; Luo, C.W.; Hung, W.C.; Lin, Y.H.; Chen, T.Y.; Li, C.F.; Lin, C.Y.; Pan, M.R. Preprogramming therapeutic response of PI3K/mTOR dual inhibitor via the regulation of EHMT2 and p27 in pancreatic cancer. Am. J. Cancer Res. 2018, 8, 1812–1822. [Google Scholar]
  91. Lu, C.; Paschall, A.V.; Shi, H.; Savage, N.; Waller, J.L.; Sabbatini, M.E.; Oberlies, N.H.; Pearce, C.; Liu, K. The MLL1-H3K4me3 Axis-Mediated PD-L1 Expression and Pancreatic Cancer Immune Evasion. J. Natl. Cancer Inst. 2017, 109, djw283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Koutsioumpa, M.; Hatziapostolou, M.; Polytarchou, C.; Tolosa, E.J.; Almada, L.L.; Mahurkar-Joshi, S.; Williams, J.; Tirado-Rodriguez, A.B.; Huerta-Yepez, S.; Karavias, D.; et al. Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming. Gut 2019, 68, 1271–1286. [Google Scholar] [CrossRef]
  93. Ettel, M.; Zhao, L.; Schechter, S.; Shi, J. Expression and prognostic value of NSD1 and SETD2 in pancreatic ductal adenocarcinoma and its precursor lesions. Pathology 2019, 51, 392–398. [Google Scholar] [CrossRef] [PubMed]
  94. Niu, N.; Lu, P.; Yang, Y.; He, R.; Zhang, L.; Shi, J.; Wu, J.; Yang, M.; Zhang, Z.G.; Wang, L.W.; et al. Loss of Setd2 promotes Kras-induced acinar-to-ductal metaplasia and epithelia-mesenchymal transition during pancreatic carcinogenesis. Gut 2020, 69, 715–726. [Google Scholar] [CrossRef] [PubMed]
  95. Reynoird, N.; Mazur, P.K.; Stellfeld, T.; Flores, N.M.; Lofgren, S.M.; Carlson, S.M.; Brambilla, E.; Hainaut, P.; Kaznowska, E.B.; Arrowsmith, C.H.; et al. Coordination of stress signals by the lysine methyltransferase SMYD2 promotes pancreatic cancer. Genes Dev. 2016, 30, 772–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Paladino, D.; Yue, P.; Furuya, H.; Acoba, J.; Rosser, C.J.; Turkson, J. A novel nuclear Src and p300 signaling axis controls migratory and invasive behavior in pancreatic cancer. Oncotarget 2016, 7, 7253–7267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Zhu, C.L.; Huang, Q. Overexpression of the SMYD3 Promotes Proliferation, Migration, and Invasion of Pancreatic Cancer. Dig. Dis. Sci. 2020, 65, 489–499. [Google Scholar] [CrossRef]
  98. Shan, L.; Hao, C.; Jun, Z.; Qinghe, C. Histone methyltransferase Dot1L inhibits pancreatic cancer cell apoptosis by promoting NUPR1 expression. J. Int. Med. Res. 2022, 50, 3000605221088431. [Google Scholar] [CrossRef]
  99. Liu, M.; Shi, Y.; Hu, Q.; Qin, Y.; Ji, S.; Liu, W.; Zhuo, Q.; Fan, G.; Ye, Z.; Song, C.; et al. SETD8 induces stemness and epithelial-mesenchymal transition of pancreatic cancer cells by regulating ROR1 expression. Acta Biochim. Biophys. Sin. 2021, 53, 1614–1624. [Google Scholar] [CrossRef]
  100. Liu, M.; Qin, Y.; Hu, Q.; Liu, W.; Ji, S.; Xu, W.; Fan, G.; Ye, Z.; Zhang, Z.; Xu, X.; et al. SETD8 potentiates constitutive ERK1/2 activation via epigenetically silencing DUSP10 expression in pancreatic cancer. Cancer Lett. 2021, 499, 265–278. [Google Scholar] [CrossRef]
  101. Viotti, M.; Wilson, C.; McCleland, M.; Koeppen, H.; Haley, B.; Jhunjhunwala, S.; Klijn, C.; Modrusan, Z.; Arnott, D.; Classon, M.; et al. SUV420H2 is an epigenetic regulator of epithelial/mesenchymal states in pancreatic cancer. J. Cell Biol. 2018, 217, 763–777. [Google Scholar] [CrossRef] [PubMed]
  102. Hasegawa, S.; Nagano, H.; Konno, M.; Eguchi, H.; Tomokuni, A.; Tomimaru, Y.; Asaoka, T.; Wada, H.; Hama, N.; Kawamoto, K.; et al. A crucial epithelial to mesenchymal transition regulator, Sox4/Ezh2 axis is closely related to the clinical outcome in pancreatic cancer patients. Int. J. Oncol. 2016, 48, 145–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Patil, S.; Steuber, B.; Kopp, W.; Kari, V.; Urbach, L.; Wang, X.; Kuffer, S.; Bohnenberger, H.; Spyropoulou, D.; Zhang, Z.; et al. EZH2 Regulates Pancreatic Cancer Subtype Identity and Tumor Progression via Transcriptional Repression of GATA6. Cancer Res. 2020, 80, 4620–4632. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, Y.; Xie, D.; Yin Li, W.; Man Cheung, C.; Yao, H.; Chan, C.Y.; Chan, C.Y.; Xu, F.P.; Liu, Y.H.; Sung, J.J.; et al. RNAi targeting EZH2 inhibits tumor growth and liver metastasis of pancreatic cancer in vivo. Cancer Lett. 2010, 297, 109–116. [Google Scholar] [CrossRef] [PubMed]
  105. Jin, X.; Yang, C.; Fan, P.; Xiao, J.; Zhang, W.; Zhan, S.; Liu, T.; Wang, D.; Wu, H. CDK5/FBW7-dependent ubiquitination and degradation of EZH2 inhibits pancreatic cancer cell migration and invasion. J. Biol. Chem. 2017, 292, 6269–6280. [Google Scholar] [CrossRef] [Green Version]
  106. Han, T.; Jiao, F.; Hu, H.; Yuan, C.; Wang, L.; Jin, Z.L.; Song, W.F.; Wang, L.W. EZH2 promotes cell migration and invasion but not alters cell proliferation by suppressing E-cadherin, partly through association with MALAT-1 in pancreatic cancer. Oncotarget 2016, 7, 11194–11207. [Google Scholar] [CrossRef] [Green Version]
  107. Qin, Y.; Zhu, W.; Xu, W.; Zhang, B.; Shi, S.; Ji, S.; Liu, J.; Long, J.; Liu, C.; Liu, L.; et al. LSD1 sustains pancreatic cancer growth via maintaining HIF1alpha-dependent glycolytic process. Cancer Lett. 2014, 347, 225–232. [Google Scholar] [CrossRef]
  108. Hou, X.; Li, Q.; Yang, L.; Yang, Z.; He, J.; Li, Q.; Li, D. KDM1A and KDM3A promote tumor growth by upregulating cell cycle-associated genes in pancreatic cancer. Exp. Biol. Med. 2021, 246, 1869–1883. [Google Scholar] [CrossRef]
  109. Wanna-Udom, S.; Terashima, M.; Suphakhong, K.; Ishimura, A.; Takino, T.; Suzuki, T. KDM2B is involved in the epigenetic regulation of TGF-beta-induced epithelial-mesenchymal transition in lung and pancreatic cancer cell lines. J. Biol. Chem. 2021, 296, 100213. [Google Scholar] [CrossRef]
  110. Tzatsos, A.; Paskaleva, P.; Ferrari, F.; Deshpande, V.; Stoykova, S.; Contino, G.; Wong, K.K.; Lan, F.; Trojer, P.; Park, P.J.; et al. KDM2B promotes pancreatic cancer via Polycomb-dependent and -independent transcriptional programs. J. Clin. Investig. 2013, 123, 727–739. [Google Scholar] [CrossRef] [Green Version]
  111. Pu, N.; Gao, S.; Yin, H.; Li, J.A.; Wu, W.; Fang, Y.; Zhang, L.; Rong, Y.; Xu, X.; Wang, D.; et al. Cell-intrinsic PD-1 promotes proliferation in pancreatic cancer by targeting CYR61/CTGF via the hippo pathway. Cancer Lett. 2019, 460, 42–53. [Google Scholar] [CrossRef] [PubMed]
  112. Quan, M.; Chen, Z.; Jiao, F.; Xiao, X.; Xia, Q.; Chen, J.; Chao, Q.; Li, Y.; Gao, Y.; Yang, H.; et al. Lysine demethylase 2 (KDM2B) regulates hippo pathway via MOB1 to promote pancreatic ductal adenocarcinoma (PDAC) progression. J. Exp. Clin. Cancer Res. 2020, 39, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Dandawate, P.; Ghosh, C.; Palaniyandi, K.; Paul, S.; Rawal, S.; Pradhan, R.; Sayed, A.A.A.; Choudhury, S.; Standing, D.; Subramaniam, D.; et al. The Histone Demethylase KDM3A, Increased in Human Pancreatic Tumors, Regulates Expression of DCLK1 and Promotes Tumorigenesis in Mice. Gastroenterology 2019, 157, 1646–1659 e11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Li, J.; Yuan, S.; Norgard, R.J.; Yan, F.; Sun, Y.H.; Kim, I.K.; Merrell, A.J.; Sela, Y.; Jiang, Y.; Bhanu, N.V.; et al. Epigenetic and Transcriptional Control of the Epidermal Growth Factor Receptor Regulates the Tumor Immune Microenvironment in Pancreatic Cancer. Cancer Discov. 2021, 11, 736–753. [Google Scholar] [CrossRef] [PubMed]
  115. Ding, G.; Xu, X.; Li, D.; Chen, Y.; Wang, W.; Ping, D.; Jia, S.; Cao, L. Fisetin inhibits proliferation of pancreatic adenocarcinoma by inducing DNA damage via RFXAP/KDM4A-dependent histone H3K36 demethylation. Cell Death Dis. 2020, 11, 893. [Google Scholar] [CrossRef] [PubMed]
  116. Li, S.; Wu, L.; Wang, Q.; Li, Y.; Wang, X. KDM4B promotes epithelial-mesenchymal transition through up-regulation of ZEB1 in pancreatic cancer. Acta Biochim. Biophys. Sin. 2015, 47, 997–1004. [Google Scholar] [CrossRef] [Green Version]
  117. Li, H.; Peng, C.; Zhu, C.; Nie, S.; Qian, X.; Shi, Z.; Shi, M.; Liang, Y.; Ding, X.; Zhang, S.; et al. Hypoxia promotes the metastasis of pancreatic cancer through regulating NOX4/KDM5A-mediated histone methylation modification changes in a HIF1A-independent manner. Clin. Epigenet. 2021, 13, 18. [Google Scholar] [CrossRef] [PubMed]
  118. Batie, M.; Frost, J.; Frost, M.; Wilson, J.W.; Schofield, P.; Rocha, S. Hypoxia induces rapid changes to histone methylation and reprograms chromatin. Science 2019, 363, 1222–1226. [Google Scholar] [CrossRef] [PubMed]
  119. Cui, J.; Quan, M.; Xie, D.; Gao, Y.; Guha, S.; Fallon, M.B.; Chen, J.; Xie, K. A novel KDM5A/MPC-1 signaling pathway promotes pancreatic cancer progression via redirecting mitochondrial pyruvate metabolism. Oncogene 2020, 39, 1140–1151. [Google Scholar] [CrossRef] [PubMed]
  120. Yi, Z.; Wei, S.; Jin, L.; Jeyarajan, S.; Yang, J.; Gu, Y.; Kim, H.S.; Schechter, S.; Lu, S.; Paulsen, M.T.; et al. KDM6A Regulates Cell Plasticity and Pancreatic Cancer Progression by Noncanonical Activin Pathway. Cell Mol. Gastroenterol. Hepatol. 2022, 13, 643–667. [Google Scholar] [CrossRef] [PubMed]
  121. Watanabe, S.; Shimada, S.; Akiyama, Y.; Ishikawa, Y.; Ogura, T.; Ogawa, K.; Ono, H.; Mitsunori, Y.; Ban, D.; Kudo, A.; et al. Loss of KDM6A characterizes a poor prognostic subtype of human pancreatic cancer and potentiates HDAC inhibitor lethality. Int. J. Cancer 2019, 145, 192–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Kalisz, M.; Bernardo, E.; Beucher, A.; Maestro, M.A.; Del Pozo, N.; Millan, I.; Haeberle, L.; Schlensog, M.; Safi, S.A.; Knoefel, W.T.; et al. HNF1A recruits KDM6A to activate differentiated acinar cell programs that suppress pancreatic cancer. EMBO J. 2020, 39, e102808. [Google Scholar] [CrossRef] [PubMed]
  123. Barthel, S.; Schneider, G.; Saur, D. Blocking the road to de-differentiation: HNF1A/KDM6A complex safeguards epithelial integrity in pancreatic cancer. EMBO J. 2020, 39, e104759. [Google Scholar] [CrossRef] [PubMed]
  124. Gayther, S.A.; Batley, S.J.; Linger, L.; Bannister, A.; Thorpe, K.; Chin, S.F.; Daigo, Y.; Russell, P.; Wilson, A.; Sowter, H.M.; et al. Mutations truncating the EP300 acetylase in human cancers. Nat. Genet. 2000, 24, 300–303. [Google Scholar] [CrossRef]
  125. Sun, Y.; Ren, D.; Zhou, Y.; Shen, J.; Wu, H.; Jin, X. Histone acetyltransferase 1 promotes gemcitabine resistance by regulating the PVT1/EZH2 complex in pancreatic cancer. Cell Death Dis. 2021, 12, 878. [Google Scholar] [CrossRef]
  126. Fan, P.; Zhao, J.; Meng, Z.; Wu, H.; Wang, B.; Wu, H.; Jin, X. Overexpressed histone acetyltransferase 1 regulates cancer immunity by increasing programmed death-ligand 1 expression in pancreatic cancer. J. Exp. Clin. Cancer Res. 2019, 38, 47. [Google Scholar] [CrossRef] [Green Version]
  127. von Burstin, J.; Eser, S.; Paul, M.C.; Seidler, B.; Brandl, M.; Messer, M.; von Werder, A.; Schmidt, A.; Mages, J.; Pagel, P.; et al. E-cadherin regulates metastasis of pancreatic cancer in vivo and is suppressed by a SNAIL/HDAC1/HDAC2 repressor complex. Gastroenterology 2009, 137, 361–371. [Google Scholar] [CrossRef]
  128. Aghdassi, A.; Sendler, M.; Guenther, A.; Mayerle, J.; Behn, C.O.; Heidecke, C.D.; Friess, H.; Buchler, M.; Evert, M.; Lerch, M.M.; et al. Recruitment of histone deacetylases HDAC1 and HDAC2 by the transcriptional repressor ZEB1 downregulates E-cadherin expression in pancreatic cancer. Gut 2012, 61, 439–448. [Google Scholar] [CrossRef]
  129. Shinke, G.; Yamada, D.; Eguchi, H.; Iwagami, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K.; Gotoh, K.; Kobayashi, S.; et al. Role of histone deacetylase 1 in distant metastasis of pancreatic ductal cancer. Cancer Sci. 2018, 109, 2520–2531. [Google Scholar] [CrossRef] [Green Version]
  130. Fritsche, P.; Seidler, B.; Schuler, S.; Schnieke, A.; Gottlicher, M.; Schmid, R.M.; Saur, D.; Schneider, G. HDAC2 mediates therapeutic resistance of pancreatic cancer cells via the BH3-only protein NOXA. Gut 2009, 58, 1399–1409. [Google Scholar] [CrossRef] [Green Version]
  131. Schuler, S.; Fritsche, P.; Diersch, S.; Arlt, A.; Schmid, R.M.; Saur, D.; Schneider, G. HDAC2 attenuates TRAIL-induced apoptosis of pancreatic cancer cells. Mol. Cancer 2010, 9, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Zhou, Y.; Jin, X.; Yu, H.; Qin, G.; Pan, P.; Zhao, J.; Chen, T.; Liang, X.; Sun, Y.; Wang, B.; et al. HDAC5 modulates PD-L1 expression and cancer immunity via p65 deacetylation in pancreatic cancer. Theranostics 2022, 12, 2080–2094. [Google Scholar] [CrossRef] [PubMed]
  133. Wauters, E.; Sanchez-Arevalo Lobo, V.J.; Pinho, A.V.; Mawson, A.; Herranz, D.; Wu, J.; Cowley, M.J.; Colvin, E.K.; Njicop, E.N.; Sutherland, R.L.; et al. Sirtuin-1 regulates acinar-to-ductal metaplasia and supports cancer cell viability in pancreatic cancer. Cancer Res. 2013, 73, 2357–2367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Zhang, J.G.; Hong, D.F.; Zhang, C.W.; Sun, X.D.; Wang, Z.F.; Shi, Y.; Liu, J.W.; Shen, G.L.; Zhang, Y.B.; Cheng, J.; et al. Sirtuin 1 facilitates chemoresistance of pancreatic cancer cells by regulating adaptive response to chemotherapy-induced stress. Cancer Sci. 2014, 105, 445–454. [Google Scholar] [CrossRef] [PubMed]
  135. Chen, I.C.; Chiang, W.F.; Huang, H.H.; Chen, P.F.; Shen, Y.Y.; Chiang, H.C. Role of SIRT1 in regulation of epithelial-to-mesenchymal transition in oral squamous cell carcinoma metastasis. Mol. Cancer 2014, 13, 254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Kugel, S.; Sebastian, C.; Fitamant, J.; Ross, K.N.; Saha, S.K.; Jain, E.; Gladden, A.; Arora, K.S.; Kato, Y.; Rivera, M.N.; et al. SIRT6 Suppresses Pancreatic Cancer through Control of Lin28b. Cell 2016, 165, 1401–1415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Proctor, E.; Waghray, M.; Lee, C.J.; Heidt, D.G.; Yalamanchili, M.; Li, C.; Bednar, F.; Simeone, D.M. Bmi1 enhances tumorigenicity and cancer stem cell function in pancreatic adenocarcinoma. PLoS ONE 2013, 8, e55820. [Google Scholar] [CrossRef] [Green Version]
  138. Guo, S.; Xu, X.; Tang, Y.; Zhang, C.; Li, J.; Ouyang, Y.; Ju, J.; Bie, P.; Wang, H. miR-15a inhibits cell proliferation and epithelial to mesenchymal transition in pancreatic ductal adenocarcinoma by down-regulating Bmi-1 expression. Cancer Lett. 2014, 344, 40–46. [Google Scholar] [CrossRef]
  139. Liang, J.X.; Ning, Z.; Gao, W.; Ling, J.; Wang, A.M.; Luo, H.F.; Liang, Y.; Yan, Q.; Wang, Z.Y. Ubiquitin-specific protease 22-induced autophagy is correlated with poor prognosis of pancreatic cancer. Oncol. Rep. 2014, 32, 2726–2734. [Google Scholar] [CrossRef] [Green Version]
  140. Li, Z.; Huang, X.; Hu, W.; Lu, H. Down-regulation of USP22 reduces cell stemness and enhances the sensitivity of pancreatic cancer cells to cisplatin by inactivating the Wnt/beta-catenin pathway. Tissue Cell 2022, 77, 101787. [Google Scholar] [CrossRef]
  141. Huang, L.; Hu, C.; Cao, H.; Wu, X.; Wang, R.; Lu, H.; Li, H.; Chen, H. MicroRNA-29c Increases the Chemosensitivity of Pancreatic Cancer Cells by Inhibiting USP22 Mediated Autophagy. Cell Physiol. Biochem. 2018, 47, 747–758. [Google Scholar] [CrossRef] [PubMed]
  142. Bai, Z.; Du, Y.; Cong, L.; Cheng, Y. The USP22 promotes the growth of cancer cells through the DYRK1A in pancreatic ductal adenocarcinoma. Gene 2020, 758, 144960. [Google Scholar] [CrossRef] [PubMed]
  143. Ning, Z.; Wang, A.; Liang, J.; Xie, Y.; Liu, J.; Yan, Q.; Wang, Z. USP22 promotes epithelial-mesenchymal transition via the FAK pathway in pancreatic cancer cells. Oncol. Rep. 2014, 32, 1451–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Li, J.; Yuan, S.; Norgard, R.J.; Yan, F.; Yamazoe, T.; Blanco, A.; Stanger, B.Z. Tumor Cell-Intrinsic USP22 Suppresses Antitumor Immunity in Pancreatic Cancer. Cancer Immunol. Res. 2020, 8, 282–291. [Google Scholar] [CrossRef]
  145. Ning, Z.; Wang, A.; Liang, J.; Xie, Y.; Liu, J.; Feng, L.; Yan, Q.; Wang, Z. USP22 promotes the G1/S phase transition by upregulating FoxM1 expression via beta-catenin nuclear localization and is associated with poor prognosis in stage II pancreatic ductal adenocarcinoma. Int. J. Oncol. 2014, 45, 1594–1608. [Google Scholar] [CrossRef] [Green Version]
  146. Chen, L.; Xu, Z.; Li, Q.; Feng, Q.; Zheng, C.; Du, Y.; Yuan, R.; Peng, X. USP28 facilitates pancreatic cancer progression through activation of Wnt/beta-catenin pathway via stabilising FOXM1. Cell Death Dis. 2021, 12, 887. [Google Scholar] [CrossRef] [PubMed]
  147. Wu, Q.; Schapira, M.; Arrowsmith, C.H.; Barsyte-Lovejoy, D. Protein arginine methylation: From enigmatic functions to therapeutic targeting. Nat. Rev. Drug Discov. 2021, 20, 509–530. [Google Scholar] [CrossRef]
  148. Zhou, C.; Yi, C.; Yi, Y.; Qin, W.; Yan, Y.; Dong, X.; Zhang, X.; Huang, Y.; Zhang, R.; Wei, J.; et al. LncRNA PVT1 promotes gemcitabine resistance of pancreatic cancer via activating Wnt/beta-catenin and autophagy pathway through modulating the miR-619-5p/Pygo2 and miR-619-5p/ATG14 axes. Mol. Cancer 2020, 19, 118. [Google Scholar] [CrossRef]
  149. Cai, K.; Deng, L.; Zheng, D.; Li, L.; He, Z.; Yu, C. MICAL1 facilitates pancreatic cancer proliferation, migration, and invasion by activating WNT/beta-catenin pathway. J. Transl. Med. 2022, 20, 528. [Google Scholar] [CrossRef]
  150. Hua, Y.Q.; Zhang, K.; Sheng, J.; Ning, Z.Y.; Li, Y.; Shi, W.D.; Liu, L.M. Fam83D promotes tumorigenesis and gemcitabine resistance of pancreatic adenocarcinoma through the Wnt/beta-catenin pathway. Life Sci. 2021, 287, 119205. [Google Scholar] [CrossRef]
  151. Wang, Z.; Li, Z.; Ji, H. Direct targeting of beta-catenin in the Wnt signaling pathway: Current progress and perspectives. Med. Res. Rev. 2021, 41, 2109–2129. [Google Scholar] [CrossRef] [PubMed]
  152. Chen, Y.; Chen, Z.; Tang, Y.; Xiao, Q. The involvement of noncanonical Wnt signaling in cancers. Biomed. Pharmacother. 2021, 133, 110946. [Google Scholar] [CrossRef]
  153. Quatannens, D.; Verhoeven, Y.; Van Dam, P.; Lardon, F.; Prenen, H.; Roeyen, G.; Peeters, M.; Smits, E.L.J.; Van Audenaerde, J. Targeting hedgehog signaling in pancreatic ductal adenocarcinoma. Pharmacol. Ther. 2022, 236, 108107. [Google Scholar] [CrossRef] [PubMed]
  154. Pal, S.; Vishwanath, S.N.; Erdjument-Bromage, H.; Tempst, P.; Sif, S. Human SWI/SNF-associated PRMT5 methylates histone H3 arginine 8 and negatively regulates expression of ST7 and NM23 tumor suppressor genes. Mol. Cell. Biol. 2004, 24, 9630–9645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Levantini, E.; Maroni, G.; Del Re, M.; Tenen, D.G. EGFR signaling pathway as therapeutic target in human cancers. Semin. Cancer Biol. 2022, 85, 253–275. [Google Scholar] [CrossRef]
  156. Molenaar, T.M.; van Leeuwen, F. SETD2: From chromatin modifier to multipronged regulator of the genome and beyond. Cell Mol. Life Sci. 2022, 79, 346. [Google Scholar] [CrossRef]
  157. Hyun, K.; Jeon, J.; Park, K.; Kim, J. Writing, erasing and reading histone lysine methylations. Exp. Mol. Med. 2017, 49, e324. [Google Scholar] [CrossRef] [Green Version]
  158. Borcherding, N.; Kusner, D.; Liu, G.H.; Zhang, W. ROR1, an embryonic protein with an emerging role in cancer biology. Protein Cell 2014, 5, 496–502. [Google Scholar] [CrossRef] [Green Version]
  159. Hou, P.; Zhao, Y.; Li, Z.; Yao, R.; Ma, M.; Gao, Y.; Zhao, L.; Zhang, Y.; Huang, B.; Lu, J. LincRNA-ROR induces epithelial-to-mesenchymal transition and contributes to breast cancer tumorigenesis and metastasis. Cell Death Dis. 2014, 5, e1287. [Google Scholar] [CrossRef] [Green Version]
  160. Duan, R.; Du, W.; Guo, W. EZH2: A novel target for cancer treatment. J. Hematol. Oncol. 2020, 13, 104. [Google Scholar] [CrossRef]
  161. Moreno, C.S. SOX4: The unappreciated oncogene. Semin. Cancer Biol. 2020, 67, 57–64. [Google Scholar] [CrossRef] [PubMed]
  162. Tiwari, N.; Tiwari, V.K.; Waldmeier, L.; Balwierz, P.J.; Arnold, P.; Pachkov, M.; Meyer-Schaller, N.; Schubeler, D.; van Nimwegen, E.; Christofori, G. Sox4 is a master regulator of epithelial-mesenchymal transition by controlling Ezh2 expression and epigenetic reprogramming. Cancer Cell 2013, 23, 768–783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Jiao, F.; Hu, H.; Yuan, C.; Wang, L.; Jiang, W.; Jin, Z.; Guo, Z.; Wang, L. Elevated expression level of long noncoding RNA MALAT-1 facilitates cell growth, migration and invasion in pancreatic cancer. Oncol. Rep. 2014, 32, 2485–2492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. He, J.; Kallin, E.M.; Tsukada, Y.; Zhang, Y. The H3K36 demethylase Jhdm1b/Kdm2b regulates cell proliferation and senescence through p15(Ink4b). Nat. Struct. Mol. Biol. 2008, 15, 1169–1175. [Google Scholar] [CrossRef]
  165. Nishio, M.; Sugimachi, K.; Goto, H.; Wang, J.; Morikawa, T.; Miyachi, Y.; Takano, Y.; Hikasa, H.; Itoh, T.; Suzuki, S.O.; et al. Dysregulated YAP1/TAZ and TGF-beta signaling mediate hepatocarcinogenesis in Mob1a/1b-deficient mice. Proc. Natl. Acad. Sci. USA 2016, 113, E71–E80. [Google Scholar] [CrossRef] [Green Version]
  166. Yamane, K.; Toumazou, C.; Tsukada, Y.; Erdjument-Bromage, H.; Tempst, P.; Wong, J.; Zhang, Y. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell 2006, 125, 483–495. [Google Scholar] [CrossRef] [Green Version]
  167. Ge, Y.; Liu, H.; Zhang, Y.; Liu, J.; Yan, R.; Xiao, Z.; Fan, X.; Huang, X.; An, G. Inhibition of DCLK1 kinase reverses epithelial-mesenchymal transition and restores T-cell activity in pancreatic ductal adenocarcinoma. Transl. Oncol. 2022, 17, 101317. [Google Scholar] [CrossRef]
  168. Sureban, S.M.; May, R.; Lightfoot, S.A.; Hoskins, A.B.; Lerner, M.; Brackett, D.J.; Postier, R.G.; Ramanujam, R.; Mohammed, A.; Rao, C.V.; et al. DCAMKL-1 regulates epithelial-mesenchymal transition in human pancreatic cells through a miR-200a-dependent mechanism. Cancer Res. 2011, 71, 2328–2338. [Google Scholar] [CrossRef] [Green Version]
  169. Manni, W.; Jianxin, X.; Weiqi, H.; Siyuan, C.; Huashan, S. JMJD family proteins in cancer and inflammation. Signal Transduct. Target. Ther. 2022, 7, 304. [Google Scholar] [CrossRef]
  170. Kooistra, S.M.; Helin, K. Molecular mechanisms and potential functions of histone demethylases. Nat. Rev. Mol. Cell Biol. 2012, 13, 297–311. [Google Scholar] [CrossRef]
  171. Yang, G.J.; Zhu, M.H.; Lu, X.J.; Liu, Y.J.; Lu, J.F.; Leung, C.H.; Ma, D.L.; Chen, J. The emerging role of KDM5A in human cancer. J. Hematol. Oncol. 2021, 14, 30. [Google Scholar] [CrossRef] [PubMed]
  172. Vermot, A.; Petit-Hartlein, I.; Smith, S.M.E.; Fieschi, F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants 2021, 10, 890. [Google Scholar] [CrossRef] [PubMed]
  173. Lee, M.G.; Villa, R.; Trojer, P.; Norman, J.; Yan, K.P.; Reinberg, D.; Di Croce, L.; Shiekhattar, R. Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science 2007, 318, 447–450. [Google Scholar] [CrossRef] [PubMed]
  174. Abel, E.V.; Goto, M.; Magnuson, B.; Abraham, S.; Ramanathan, N.; Hotaling, E.; Alaniz, A.A.; Kumar-Sinha, C.; Dziubinski, M.L.; Urs, S.; et al. HNF1A is a novel oncogene that regulates human pancreatic cancer stem cell properties. Elife 2018, 7, 33947. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, Y.I.; Chang, C.C.; Hsu, M.F.; Jeng, Y.M.; Tien, Y.W.; Chang, M.C.; Chang, Y.T.; Hu, C.M.; Lee, W.H. Homophilic ATP1A1 binding induces activin A secretion to promote EMT of tumor cells and myofibroblast activation. Nat. Commun. 2022, 13, 2945. [Google Scholar] [CrossRef]
  176. Basu, M.; Bhattacharya, R.; Ray, U.; Mukhopadhyay, S.; Chatterjee, U.; Roy, S.S. Invasion of ovarian cancer cells is induced byPITX2-mediated activation of TGF-beta and Activin-A. Mol. Cancer 2015, 14, 162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Chen, Q.; Yang, B.; Liu, X.; Zhang, X.D.; Zhang, L.; Liu, T. Histone acetyltransferases CBP/p300 in tumorigenesis and CBP/p300 inhibitors as promising novel anticancer agents. Theranostics 2022, 12, 4935–4948. [Google Scholar] [CrossRef]
  178. Ono, H.; Basson, M.D.; Ito, H. P300 inhibition enhances gemcitabine-induced apoptosis of pancreatic cancer. Oncotarget 2016, 7, 51301–51310. [Google Scholar] [CrossRef] [Green Version]
  179. Zhong, Z.; Harmston, N.; Wood, K.C.; Madan, B.; Virshup, D.M. A p300/GATA6 axis determines differentiation and Wnt dependency in pancreatic cancer models. J. Clin. Investig. 2022, 132, e156305. [Google Scholar] [CrossRef]
  180. Martinelli, P.; Carrillo-de Santa Pau, E.; Cox, T.; Sainz, B., Jr.; Dusetti, N.; Greenhalf, W.; Rinaldi, L.; Costello, E.; Ghaneh, P.; Malats, N.; et al. GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer. Gut 2017, 66, 1665–1676. [Google Scholar] [CrossRef] [Green Version]
  181. Koutelou, E.; Farria, A.T.; Dent, S.Y.R. Complex functions of Gcn5 and Pcaf in development and disease. Biochim. Biophys. Acta Gene Regul. Mech. 2021, 1864, 194609. [Google Scholar] [CrossRef] [PubMed]
  182. Mizuguchi, Y.; Specht, S.; Lunz, J.G., 3rd; Isse, K.; Corbitt, N.; Takizawa, T.; Demetris, A.J. Cooperation of p300 and PCAF in the control of microRNA 200c/141 transcription and epithelial characteristics. PLoS ONE 2012, 7, e32449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Diaz-Riascos, Z.V.; Ginesta, M.M.; Fabregat, J.; Serrano, T.; Busquets, J.; Buscail, L.; Cordelier, P.; Capella, G. Expression and Role of MicroRNAs from the miR-200 Family in the Tumor Formation and Metastatic Propensity of Pancreatic Cancer. Mol. Ther. Nucleic Acids 2019, 17, 491–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Li, Y.H.; Li, Y.X.; Li, M.; Song, S.W.; Ge, Y.; Jin, J.Y.; Li, X.Y.; Tan, X.D.; Ye, J. The Ras-ERK1/2 signaling pathway regulates H3K9ac through PCAF to promote the development of pancreatic cancer. Life Sci. 2020, 256, 117936. [Google Scholar] [CrossRef] [PubMed]
  185. Giaginis, C.; Damaskos, C.; Koutsounas, I.; Zizi-Serbetzoglou, A.; Tsoukalas, N.; Patsouris, E.; Kouraklis, G.; Theocharis, S. Histone deacetylase (HDAC)-1, -2, -4 and -6 expression in human pancreatic adenocarcinoma: Associations with clinicopathological parameters, tumor proliferative capacity and patients’ survival. BMC Gastroenterol. 2015, 15, 148. [Google Scholar] [CrossRef] [Green Version]
  186. Miyake, K.; Yoshizumi, T.; Imura, S.; Sugimoto, K.; Batmunkh, E.; Kanemura, H.; Morine, Y.; Shimada, M. Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: Correlation with poor prognosis with possible regulation. Pancreas 2008, 36, e1–e9. [Google Scholar] [CrossRef]
  187. Chen, X.; Huan, H.; Liu, C.; Luo, Y.; Shen, J.; Zhuo, Y.; Zhang, Z.; Qian, C. Deacetylation of beta-catenin by SIRT1 regulates self-renewal and oncogenesis of liver cancer stem cells. Cancer Lett. 2019, 463, 1–10. [Google Scholar] [CrossRef]
  188. Blackledge, N.P.; Klose, R.J. The molecular principles of gene regulation by Polycomb repressive complexes. Nat. Rev. Mol. Cell Biol. 2021, 22, 815–833. [Google Scholar] [CrossRef]
  189. Chen, J.; Xu, H.; Zou, X.; Wang, J.; Zhu, Y.; Chen, H.; Shen, B.; Deng, X.; Zhou, A.; Chin, Y.E.; et al. Snail recruits Ring1B to mediate transcriptional repression and cell migration in pancreatic cancer cells. Cancer Res. 2014, 74, 4353–4363. [Google Scholar] [CrossRef] [Green Version]
  190. Song, W.; Tao, K.; Li, H.; Jin, C.; Song, Z.; Li, J.; Shi, H.; Li, X.; Dang, Z.; Dou, K. Bmi-1 is related to proliferation, survival and poor prognosis in pancreatic cancer. Cancer Sci. 2010, 101, 1754–1760. [Google Scholar] [CrossRef]
  191. Young, M.J.; Hsu, K.C.; Lin, T.E.; Chang, W.C.; Hung, J.J. The role of ubiquitin-specific peptidases in cancer progression. J. Biomed. Sci. 2019, 26, 42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Feng, T.; Ling, S.; Xu, C.; Ying, L.; Su, D.; Xu, X. Ubiquitin-specific peptidase 22 in cancer. Cancer Lett. 2021, 514, 30–37. [Google Scholar] [CrossRef]
  193. Zhao, J.; Guan, J.L. Signal transduction by focal adhesion kinase in cancer. Cancer Metastasis Rev. 2009, 28, 35–49. [Google Scholar] [CrossRef] [PubMed]
  194. Li, F.; Han, H.; Sun, Q.; Liu, K.; Lin, N.; Xu, C.; Zhao, Z.; Zhao, W. USP28 regulates deubiquitination of histone H2A and cell proliferation. Exp. Cell Res. 2019, 379, 11–18. [Google Scholar] [CrossRef] [PubMed]
  195. Cao, J.; Li, J.; Sun, L.; Qin, T.; Xiao, Y.; Chen, K.; Qian, W.; Duan, W.; Lei, J.; Ma, J.; et al. Hypoxia-driven paracrine osteopontin/integrin alphavbeta3 signaling promotes pancreatic cancer cell epithelial-mesenchymal transition and cancer stem cell-like properties by modulating forkhead box protein M1. Mol. Oncol. 2019, 13, 228–245. [Google Scholar] [CrossRef] [Green Version]
  196. Quan, M.; Cui, J.; Xia, T.; Jia, Z.; Xie, D.; Wei, D.; Huang, S.; Huang, Q.; Zheng, S.; Xie, K. Merlin/NF2 Suppresses Pancreatic Tumor Growth and Metastasis by Attenuating the FOXM1-Mediated Wnt/beta-Catenin Signaling. Cancer Res. 2015, 75, 4778–4789. [Google Scholar] [CrossRef] [Green Version]
  197. Bates, S.E. Epigenetic Therapies for Cancer. N. Engl. J. Med. 2020, 383, 650–663. [Google Scholar] [CrossRef]
  198. Eich, M.L.; Athar, M.; Ferguson, J.E., 3rd; Varambally, S. EZH2-Targeted Therapies in Cancer: Hype or a Reality. Cancer Res. 2020, 80, 5449–5458. [Google Scholar] [CrossRef]
  199. Avan, A.; Crea, F.; Paolicchi, E.; Funel, N.; Galvani, E.; Marquez, V.E.; Honeywell, R.J.; Danesi, R.; Peters, G.J.; Giovannetti, E. Molecular mechanisms involved in the synergistic interaction of the EZH2 inhibitor 3-deazaneplanocin A with gemcitabine in pancreatic cancer cells. Mol. Cancer Ther. 2012, 11, 1735–1746. [Google Scholar] [CrossRef] [Green Version]
  200. Sun, Y.; Fan, J.; Wang, B.; Meng, Z.; Ren, D.; Zhao, J.; Liu, Z.; Li, D.; Jin, X.; Wu, H. The aberrant expression of ADAR1 promotes resistance to BET inhibitors in pancreatic cancer by stabilizing c-Myc. Am. J. Cancer Res. 2020, 10, 148–163. [Google Scholar]
  201. Feng, W.; Zhang, B.; Cai, D.; Zou, X. Therapeutic potential of histone deacetylase inhibitors in pancreatic cancer. Cancer Lett. 2014, 347, 183–190. [Google Scholar] [CrossRef] [PubMed]
  202. Roca, M.S.; Moccia, T.; Iannelli, F.; Testa, C.; Vitagliano, C.; Minopoli, M.; Camerlingo, R.; De Riso, G.; De Cecio, R.; Bruzzese, F.; et al. HDAC class I inhibitor domatinostat sensitizes pancreatic cancer to chemotherapy by targeting cancer stem cell compartment via FOXM1 modulation. J. Exp. Clin. Cancer Res. 2022, 41, 83. [Google Scholar] [CrossRef] [PubMed]
  203. Chao, M.W.; Chang, L.H.; Tu, H.J.; Chang, C.D.; Lai, M.J.; Chen, Y.Y.; Liou, J.P.; Teng, C.M.; Pan, S.L. Combination treatment strategy for pancreatic cancer involving the novel HDAC inhibitor MPT0E028 with a MEK inhibitor beyond K-Ras status. Clin. Epigenetics 2019, 11, 85. [Google Scholar] [CrossRef]
  204. Moertl, S.; Payer, S.; Kell, R.; Winkler, K.; Anastasov, N.; Atkinson, M.J. Comparison of Radiosensitization by HDAC Inhibitors CUDC-101 and SAHA in Pancreatic Cancer Cells. Int. J. Mol. Sci. 2019, 20, 3259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Liu, S.; Zhao, S.; Dong, Y.; Wang, T.; Niu, X.; Zhao, L.; Wang, G. Antitumor activity and mechanism of resistance of the novel HDAC and PI3K dual inhibitor CUDC-907 in pancreatic cancer. Cancer Chemother. Pharmacol. 2021, 87, 415–423. [Google Scholar] [CrossRef]
  206. Edderkaoui, M.; Chheda, C.; Soufi, B.; Zayou, F.; Hu, R.W.; Ramanujan, V.K.; Pan, X.; Boros, L.G.; Tajbakhsh, J.; Madhav, A.; et al. An Inhibitor of GSK3B and HDACs Kills Pancreatic Cancer Cells and Slows Pancreatic Tumor Growth and Metastasis in Mice. Gastroenterology 2018, 155, 1985–1998 e5. [Google Scholar] [CrossRef]
  207. He, S.; Dong, G.; Li, Y.; Wu, S.; Wang, W.; Sheng, C. Potent Dual BET/HDAC Inhibitors for Efficient Treatment of Pancreatic Cancer. Angew. Chem. Int. Ed. Engl. 2020, 59, 3028–3032. [Google Scholar] [CrossRef]
  208. Principe, D.R.; Xiong, R.; Li, Y.; Pham, T.N.D.; Kamath, S.D.; Dubrovskyi, O.; Ratia, K.; Huang, F.; Zhao, J.; Shen, Z.; et al. XP-524 is a dual-BET/EP300 inhibitor that represses oncogenic KRAS and potentiates immune checkpoint inhibition in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2022, 119, e2116764119. [Google Scholar] [CrossRef]
Figure 1. The signaling pathways associated with EMT in pancreatic cancer. TGF-β induces EMT through SMAD-mediated and non-SMAD signaling. TGF-β signals through a tetrameric complex of TβRI and TβRII to activate SMAD2 and SMAD3, which then combine with SMAD4. The trimeric SMAD complex regulates EMT-related transcriptional regulators. In addition, TGF-β can activate the ERK MAPK, Rho GTPase, and PI3K-AKT pathways. WNT signaling inhibits GSK3β to stabilize β-catenin, which translocates to the nucleus to promote the EMT-related gene expression program. In Hh signaling, Hh ligands bind to the PTCH1 to inhibit the SMO, activating the GLI transcription factors that induce the expression of EMT-related genes. Tyrosine kinase receptors promote EMT through the regulation of the PI3K/AKT and ERK/MAPK signaling pathways. The cell microenvironment also regulates EMT. During inflammation, IL-6 can promote EMT through the STAT3-mediated signaling pathway. Hypoxia in the tumor environment can promote EMT through HIF-1α, which interacts with the NF-κB transcriptional complex.
Figure 1. The signaling pathways associated with EMT in pancreatic cancer. TGF-β induces EMT through SMAD-mediated and non-SMAD signaling. TGF-β signals through a tetrameric complex of TβRI and TβRII to activate SMAD2 and SMAD3, which then combine with SMAD4. The trimeric SMAD complex regulates EMT-related transcriptional regulators. In addition, TGF-β can activate the ERK MAPK, Rho GTPase, and PI3K-AKT pathways. WNT signaling inhibits GSK3β to stabilize β-catenin, which translocates to the nucleus to promote the EMT-related gene expression program. In Hh signaling, Hh ligands bind to the PTCH1 to inhibit the SMO, activating the GLI transcription factors that induce the expression of EMT-related genes. Tyrosine kinase receptors promote EMT through the regulation of the PI3K/AKT and ERK/MAPK signaling pathways. The cell microenvironment also regulates EMT. During inflammation, IL-6 can promote EMT through the STAT3-mediated signaling pathway. Hypoxia in the tumor environment can promote EMT through HIF-1α, which interacts with the NF-κB transcriptional complex.
Ijms 24 04820 g001
Figure 2. Epigenetic changes include DNA methylation, histone modification, chromatin remodeling, and changes to noncoding RNA profiles. DNA methylation involves the transfer a methyl group to the C5 position of cytosine in CpG dinucleotides to form 5-methylcytosine (5mC). Histone core protein possesses a characteristic tail that is enriched for lysine and arginine residues, which are subject to post-translational modifications. Acetylation, methylation, ribosylation, or phosphorylation of histone proteins can influence gene expression. The ncRNAs include circular RNA (circRNA), long non-coding RNA (lncRNA), and microRNA (miRNA). Some specific lncRNAs are associated with chromatin remodeling and modification complexes. Chromatin remodeling involves the ATP-dependent SWItch/sucrose non-fermentable (SWI/SNF) complex that functions in the mobilization, ejection, and exchange of histones.
Figure 2. Epigenetic changes include DNA methylation, histone modification, chromatin remodeling, and changes to noncoding RNA profiles. DNA methylation involves the transfer a methyl group to the C5 position of cytosine in CpG dinucleotides to form 5-methylcytosine (5mC). Histone core protein possesses a characteristic tail that is enriched for lysine and arginine residues, which are subject to post-translational modifications. Acetylation, methylation, ribosylation, or phosphorylation of histone proteins can influence gene expression. The ncRNAs include circular RNA (circRNA), long non-coding RNA (lncRNA), and microRNA (miRNA). Some specific lncRNAs are associated with chromatin remodeling and modification complexes. Chromatin remodeling involves the ATP-dependent SWItch/sucrose non-fermentable (SWI/SNF) complex that functions in the mobilization, ejection, and exchange of histones.
Ijms 24 04820 g002
Table 1. Histone-modifying enzymes in pancreatic cancer.
Table 1. Histone-modifying enzymes in pancreatic cancer.
Epigenetic MechanismFamilyNameSynonymsSiteFunction
Histone
methylation
PRMTsPRMT1HRMT1L2, HMT2, ANM1H4R3Promotes tumor initiation, proliferation, migration, and invasion; inhibition of PRMT1 reduces the resistance to gemcitabine and anti-PD-L1 checkpoint inhibitors [76,77,78,79,80,81,82]
PRMT5HRMT1L5,
SKB1,
HSL7
H3R2Promotes tumorigenesis and aerobic glycolysis; induces EMT and promotes tumor migration and invasion; as a potential therapeutic target, has synergistic cytotoxicity with gemcitabine [83,84,85,86]
KMTsKMT1CG9a,
EHMT2
H3K9Promotes tumor initiation and proliferation; inhibition of G9a reduces the resistance to gemcitabine [87,88,89,90]
KMT2AMLL1H3K4Upgrades the expression of programmed death-ligand 1 (PD-L1) in pancreatic cancer cells [91]
KMT2DMLL4H3K4Regulates tumor proliferation and metabolism [92]
KMT3ASETD2H3K36Promotes EMT and tumor migration [93,94]
KMT3C/ESMYD2/3H4K5Promotes tumor proliferation, migration, and invasion [95,96,97]
KMT4DOT1LH3K79Inhibits tumor apoptosis [98]
KMT5ASETD8H4K20Promotes tumor proliferation; enhances cancer cell stemness and induces EMT [99,100]
KMT5CSUV420H2H4K20Activates EMT, reduces cell stemness, and increases drug sensitivity when downregulated [101]
KMT6EZH2H3K27Promotes tumor proliferation, migration, and invasion; depletion of EZH2 sensitizes cancer cells to doxorubicin and gemcitabine [102,103,104,105,106]
KDMKDM1ALSD1, AOF2H3K4Regulates tumor metabolism and promotes tumor proliferation [107,108]
KDM2BFBXL10, JHDM1BH3K4, H3K36Induces EMT and promotes tumor proliferation, migration, and invasion [109,110,111,112]
KDM3AJMJD1A, JHDM2AH3K9Regulates the immunological environment; promotes tumorigenesis and tumor metastasis and progression [108,113,114]
KDM4AJMJD2AH3K36, H3K9Impairs homologous recombination (HR), affecting DNA repair [115]
KDM4BJMJD2BH3K9Regulates EMT and promotes tumor metastasis and progression [116]
KDM5AJARID1A, RBP2H3K4Regulates tumor metabolism and EMT, promotes tumor migration and invasion [117,118,119]
KDM6AUTXH3K27Activates EMT and promotes tumor metastasis and progression [120,121,122,123]
Histone
acetylation
KATp300/CBP H3K18, H3K27Regulates EMT; inhibition of p300 enhances the cytotoxicity of gemcitabine [124]
HAT1 H4K5, H4K12Promotes tumor progression; enhances the expression of PD-L1 in pancreatic cancer cells; sensitizes cancer cells to gemcitabine [125,126]
KDACHDAC 1 Promotes EMT and tumor metastasis and invasion [127,128,129].
HDAC 2 Inhibition of EZH2 sensitizes cancer cells to apoptosis [130,131]
HDAC5 Inhibits the expression of PD-L1 in pancreatic cancer cells [132]
SIRT1 H3K9, H3K14, H3K56, H4K16, H1K26Promotes tumor initiation and EMT; inhibition of SIRT1 reduces the resistance to gemcitabine [133,134,135]
SIRT6 H3K9, H3K56Inactivation of SIRT6 promotes tumor metastasis and progression [136]
Histone
ubiquitylation
Ring1A, Ring1B, Bmi1 H2AK119Regulates EMT and cell cycle and promotes tumor invasion [137,138]
USPUSP22 H2A,
H2B
Regulates EMT and promotes tumor proliferation; downregulation of USP22 inhibits autophagy, promotes apoptosis, and regulates the immune microenvironment, which reduces the resistance to chemotherapy and immunotherapy [139,140,141,142,143,144,145]
USP28 H2APromotes EMT and accelerates cancer cell growth [146]
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

Xu, Y.; Zhu, Q. Histone Modifications Represent a Key Epigenetic Feature of Epithelial-to-Mesenchyme Transition in Pancreatic Cancer. Int. J. Mol. Sci. 2023, 24, 4820. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054820

AMA Style

Xu Y, Zhu Q. Histone Modifications Represent a Key Epigenetic Feature of Epithelial-to-Mesenchyme Transition in Pancreatic Cancer. International Journal of Molecular Sciences. 2023; 24(5):4820. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054820

Chicago/Turabian Style

Xu, Ying, and Qing Zhu. 2023. "Histone Modifications Represent a Key Epigenetic Feature of Epithelial-to-Mesenchyme Transition in Pancreatic Cancer" International Journal of Molecular Sciences 24, no. 5: 4820. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24054820

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