Next Article in Journal
Efficacy of an Antibiotic Loaded Ceramic-Based Bone Graft Substitute for the Treatment of Infected Non-Unions
Next Article in Special Issue
Interplay between the m6A Epitranscriptome and Tumor Metabolism: Mechanisms and Therapeutic Implications
Previous Article in Journal
Immune Checkpoint Inhibitors and Other Immune Therapies in Breast Cancer: A New Paradigm for Prolonged Adjuvant Immunotherapy
Previous Article in Special Issue
Exploiting the Molecular Basis of Oesophageal Cancer for Targeted Therapies and Biomarkers for Drug Response: Guiding Clinical Decision-Making
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting the Hippo Pathway in Gastric Cancer and Other Malignancies in the Digestive System: From Bench to Bedside

1
State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, China
2
State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong 999077, China
3
Sir Y.K. Pao Cancer Center, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong 999077, China
4
CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518000, China
5
Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong 999077, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 25 July 2022 / Revised: 29 September 2022 / Accepted: 30 September 2022 / Published: 8 October 2022
(This article belongs to the Special Issue Genomic and Epigenomic Alterations in Gastrointestinal (GI) Cancers)

Abstract

:
The Hippo pathway is an evolutionally conserved signaling cascade that controls organ size and tissue regeneration under physiological conditions, and its aberrations have been well studied to promote tumor initiation and progression. Dysregulation of the Hippo tumor suppressor signaling frequently occurs in gastric cancer (GC) and other solid tumors and contributes to cancer development through modulating multiple aspects, including cell proliferation, survival, metastasis, and oncotherapy resistance. In the clinic, Hippo components also possess diagnostic and prognostic values for cancer patients. Considering its crucial role in driving tumorigenesis, targeting the Hippo pathway may greatly benefit developing novel cancer therapies. This review summarizes the current research progress regarding the core components and regulation of the Hippo pathway, as well as the mechanism and functional roles of their dysregulation in gastrointestinal malignancies, especially in GC, and discusses the therapeutic potential of targeting the Hippo pathway against cancers.

1. Introduction

Gastric cancer (GC) is an aggressive malignant disease that arises from the stomach and represents the fourth leading cause of cancer-associated deaths worldwide [1]. Although treatment of GC, such as adjuvant chemo- or radiotherapies, has been dramatically improved, most GC patients are diagnosed at advanced stages with a worse prognosis and limited therapeutic options. Therefore, gaining new insights into the cancer biology of GC is essential for identifying effectors that drive gastric carcinogenesis and developing new therapeutic strategies against GC.
The Hippo pathway is highly conserved signaling throughout Drosophila melanogaster to mammals and acts as a master controller of organ size and tissue regeneration [2,3,4,5]. In the past decade, increasing evidence has emerged to support the contribution of the dysfunctional Hippo pathway, especially core Hippo signaling effectors yes-associated protein 1 (YAP1) and WW domain-containing transcription regulator 1 (TAZ), in tumor initiation, progression, and recurrence. In addition, aberrant expression or activation of the Hippo pathway is frequently observed in various cancer types, including GC, and exhibits excellent clinical value in diagnosis and prognosis for monitoring disease status. Thus, understanding the underlying mechanism by which the Hippo signaling augments tumor-facilitating properties can help to develop promising approaches for patients suffering from cancers.
Accumulative research has delineated the molecular mechanisms of Hippo-YAP1/TAZ signaling in GC, and some research has focused on developing novel therapeutic strategies targeting Hippo pathway-related cross talks. It is necessary to summarize the current research progress of the Hippo pathway in GC and other gastroenterology (GI) malignancies. In this review, we will introduce the main kinase cascade of the Hippo pathway, the aberrant activation of Hippo-YAP1/TAZ signaling in gastric carcinogenesis, the recent findings of the Hippo pathway in other GI cancers; and the clinical significance of Hippo signatures in GI cancers. With the deep investigation of the Hippo pathway in tumorigenesis, kinds of therapeutic strategies were developed for targeting the upstream or downstream of this pathway. This review will also update the identification of small molecule inhibitors for explicitly targeting the oncogenic components in the Hippo pathway. Finally, we will summarize the recent findings and point out our concerns and future directions. With further investigation of the Hippo pathway by cutting-edge techniques in GI cancer, we believe we can develop more precise strategies for eliminating Hippo-deregulated cancer.

2. Kinase Cascade of the Hippo Pathway

In mammalian cells, the canonical axis of the Hippo pathway comprises core kinases, including mammalian Ste20-like kinases 1/2 (MST1/2), salvador homolog 1 (SAV1), large tumor suppressor1/2 (LATS1/2), and MOB kinase activator 1 (MOB1) [6], as well as two transcriptional co-activators, YAP1 and TAZ. As critical downstream effectors of the Hippo pathway, YAP1 and TAZ lack DNA-binding motifs and thus function through interacting with TEA domain transcription factors (TEADs) to regulate the expression of target genes responsible for tumor cell proliferation and survival.
In general, activation of the Hippo pathway involves a series of phosphorylation cascades (Figure 1). MST1/2 works in complex with SAV1 to phosphorylate and activate LATS1/2 and its regulatory partner MOB1, which further induces the phosphorylation of YAP1 and TAZ for their inactivation. Phosphorylated YAP1 and TAZ then lead to 14-3-3 protein-dependent YAP1/TAZ cytoplasmic retention or ubiquitylation-induced degradation [7,8]. By contrast, inactivated upstream Hippo kinases and their partners prevent YAP1 and TAZ from phosphorylation, facilitating their nuclear translocation and subsequent transcription [9,10,11]. Nuclear localized YAP1 and TAZ could form the YAP1/TAZ-TEAD complex by binding with transcriptional factors TEAD1-4 to modulate subsequent gene transcription [12,13]. Taken together, Hippo effectors YAP1 and TAZ are tightly controlled by upstream kinases and regulators, and they exhibit an active form when they accumulate in the nuclei, while cytoplasmic sequestration leads to an inhibitory effect. Various genes responsible for cell growth and survival have been illustrated as direct targets of YAP1 and TAZ, and many of them are also involved in tumorigenesis upon dysregulation.
In the past years, accumulating evidence has focused on the causal relationship between irregular activation of the Hippo pathway, particularly YAP1 and TAZ, and tumor occurrence in numerous cancer types. Upon dysregulation, YAP1 and TAZ have also been illustrated to possess the tumor-facilitating potentials that contribute to tumor initiation and progression [14,15,16,17,18].

3. Hippo Pathway and GC

Mounting evidence proves that the Hippo pathway plays an important role in the initiation, progression, and recurrence of GC. In GC cell lines and human GC tumors, there is a consistent downregulation of Hippo kinases, including MST1/2 and LATS1/2, and an upregulation of downstream YAP1 and TAZ. Moreover, YAP1 signatures are preferentially expressed in the microsatellite stability/epithelial-to-mesenchymal transition (MSS/EMT) subtype group of GC [16]. The expression pattern of these Hippo components was closely associated with clinicopathologic features and prognosis of GC patients and could serve as reliable diagnostic and prognostic biomarkers in the clinic.

3.1. Functional Roles of Dysregulated Hippo Signaling in GC

Numerous studies suggest that dysregulated Hippo components coordinate to promote GC malignancy for tumor initiation, progression, and recurrence. As the most common tumor features, uncontrolled cell proliferation and survival have been well studied to be attributed to aberrant Hippo signaling inside GC tumors [19,20]. GC cells with lower MST1/2 or LATS1/2, concordant with high YAP1- and TAZ-expressing ones, possessed higher cell proliferative capability [21,22]. Upstream MST1 exerted its tumor-suppressive role in GC through a mechanism by which MST1 hindered the AMPK-Sirt3 pathway to cause excessive mitochondrial fission for cell death [23]. YAP1 and TAZ sustained enhanced cell proliferation mainly through a mechanism involving the transcriptional regulation of genes associated with cell division and cell cycle progression [24]. Targeting YAP1 resulted in impaired viability and colony formation in human GC cell lines and suppressed tumor growth in carcinogen N-methyl-N′-nitro-N-nitrosoguanidine (MNNG)-induced and Helicobacter pylori (H. pylori)-associated GC models as well [25]. Except for YAP1/TAZ, the upregulation of their well-established downstream targets, such as CTGF, BIRC5, and AREG, has also been revealed to drive gastric carcinogenesis and tumor progression [26,27]. Furthermore, dysfunctional Hippo signaling contributes to the enhanced metastatic capability of GC. Ectopically expressed YAP1 in GC MKN45 cells without YAP1 expression potentiated the malignant transition to a more invasive phenotype [28], whereas TAZ knockdown attenuated the ability of GC cells to migrate and invade [29]. In diffuse-type GC, activation of the YAP1 signaling induced by KRT17-mediated cytoskeletal reorganization elevated the IL-6 level, further accelerating EMT for tumor metastasis [30].
In addition, YAP1 was responsible for the stem-like properties of GC. In peritoneal carcinomatosis cells derived from GC patients, highly expressed YAP1 was positively associated with stemness-related signatures, such as SOX9, HES1, CD133, ITGA6, and ALDH1 [31]. High YAP1-expressing peritoneal carcinomatosis cell subset was also prone to develop PDX tumors, while genetic knockout or pharmacological inhibition of YAP1 abolished its tumorigenicity [31]. Furthermore, TAZ directly interacted with TEAD4 to augment the TAZ-dependent formation of vasculogenic mimicry and EMT in GC cells [32]. However, it is worth noting that despite YAP1 and TAZ having functional overlaps, integrative transcriptome analysis on GC cells identified the more dominant role of YAP1 in cell-substrate junction [33].
Taken together, dysregulation of the Hippo pathway promotes GC by regulating sustained proliferative signaling, enhanced metastasis, resistance to cell death, cancer stemness, and tumoral vascular formation.

3.2. The Upstream Regulatory Mechanisms on Hippo Signaling in GC

In the upstream, several regulators target different Hippo components. FBXW5 inactivated the Hippo pathway by enhancing LATS1 ubiquitination and degradation, and thus facilitated the metastasis and drug resistance of GC cells [34]. WBP2 inhibited LATS2 phosphorylation through direct interaction with the N-terminus kinase domain of LATS2 [35]. It further increased YAP-TEAD transcriptional activity and subsequently enhanced cell migration in GC [35]. Tang et al. demonstrated that STRN3 recruited MST1/2 to protein phosphatase 2A (PP2A) core enzyme complex for MST1/2 dephosphorylation, resulting in YAP1 hyperactivation [36]. Leukaemia inhibitory factor (LIF) activated tumor-suppressive LATS1/2 to diminish gastric cancer stemness cells (CSCs) and enhance the cancer-killing effect of 5-FU and doxorubicin [37]. Apart from the classical Hippo regulation, YAP1 can also be modulated by a non-canonical pathway. Protease-activated receptor-1 (PAR1) stimulated YAP1 activation via Rho, which further caused GC cell migration and stem-like features [38]. IRF3 enhanced the YAP1-TEAD4 complex interaction to increase YAP1 activity, whereas the blockade of IRF3 by amlexanox abolished GC growth [39]. The interaction between AMOTL1 and YAP1 protected YAP1 from ubiquitin-mediated degradation and facilitated nuclear translocation, thereby enhancing the tumorigenic role of YAP1 in GC [40]. In contrast, tumor suppressor RUNX3 was directly associated with TEAD via the Runt domain, which covered the DNA-recognition helix of TEAD and thus blocked its binding with DNA in GC [41]. As a result, the ability of the TEAD-YAP1 complex to promote GC cell proliferation was also hindered [41]. Furthermore, MST4–YAP axis has been demonstrated to be indispensable for GC development. Tumor suppressor MST4 kinase induced YAP1 phosphorylation at Thr83 to impede its interaction with importin α, thus restricting YAP1 activation. Depleting MST4 diminished YAP1 hyperactivation and thereby promoted GC growth [42]. Netrin-1 upregulated YAP1 via its classical receptor neogenin to promote GC cell metastasis as knockdown of neogenin abolished Netrin-1-induced YAP1 phosphorylation [43].
In addition, lncRNAs (e.g., RP11-323N12.5, LINC00649, and LncRNA MYLK-AS1) and microRNAs (e.g., microRNA-15a, microRNA-141, and microRNA-506) were also involved in Hippo dysregulation in GC [44,45,46,47,48,49,50]. LncRNAs MYLK-AS1 and LATS2-AS1-001 exerted their tumor-suppressive role by binding with EZH2 to increase LATS2 activity [44,50]. RP11-323N12.5 induced YAP1 transcription by binding to the c-MYC on its promoter region, resulting in enhanced GC progression and immunosuppression [45], while LINC00649 facilitated YAP1-dependent gastric tumorigenesis via sponging miR-16-5p [46]. For microRNAs, elevated miR-424 caused by circLARP4 repressed LATS1 translation to promote GC [51]. On the contrary, microRNA-141 directly downregulated TAZ for GC tumor suppression [47].
Recently, Hippo signaling has been illustrated to control the development of H. pylori-driven GC [52,53,54,55] (Figure 2). Chronic infection of H. pylori, especially those cytotoxin-associated gene A (CagA) positive strains, is a major risk factor for GC as it commonly delivers the oncoprotein CagA into gastric epithelial cells for tumorigenesis. The gradual increase in LATS2 and nuclear YAP1 levels was observed inside infected gastric mucosa from gastritis, and intestinal metaplasia, to GC [52]. Other Hippo signature genes, such as MST1, MST1R, vestigial-like family member 4 (VGLL4), and CTGF, were also upregulated following H. pylori infection [52]. Mechanistically, Molina-Castro et al. reported that LATS2 abolished H. pylori-induced EMT and the intestinal metaplasia phenotype, thereby preventing the occurrence of gastric tumors [52]. In addition, gastric epithelial cells infected with CagA-positive H. pylori have been shown to activate YAP1 and thus promote EMT and tumor growth of GC [53]. CagA-positive H. pylori stimulated YAP1 nuclear translocation and transactivated the proinflammatory IL-1ß for GC cell proliferation [54]. Furthermore, TAZ participated in H. pylori-driven EMT and gastric carcinogenesis [55]. Increased nuclear TAZ expression was observed in the gastric mucosa of H. pylori-associated GC mouse models and patients, which further upregulated EMT marker zinc finger E-box-binding homeobox 1 (ZEB1) to potentiate the EMT and stemness induced by H. pylori infection [55]. In addition to H. pylori, viruses also induced YAP1 hyperactivation, as RNA-seq analyses revealed an enrichment of YAP1 signature genes in human GC HGC-27 cells infected with the Sendai virus strain [39].
Furthermore, posttranslational modifications also affected the Hippo activity. N6-methyladenosine modification of YAP1 by METTL3 facilitated GC growth and migration [56]. The extracellular matrix stiffness has recently been identified to determine the hypermethylation of the YAP1 promoter [57]. The activated YAP1 further upregulated DNA methylation inhibitors, including GRHL2, TET2, and KMT2A to counteract the methylation for enhanced tumorigenic properties of GC under a stiff matrix [57].
Taken together, H. pylori promote the activation of YAP1/TAZ in gastric carcinogenesis, or the aberrant inaction of upstream quenches the suppressive effects of Hippo on YAP1/TAZ, leading to the YAP1/TAZ nuclear accumulation and oncogenic transformation. Then, how does YAP1/TAZ drive gastric carcinogenesis?

3.3. Downstream Effectors of Hippo Pathway in GC and Crosstalks between Signalings

In the downstream effectors, MYC is a direct downstream effector of YAP1/TAZ that has been identified to initiate GC [16]. In a mice model with conditional LATS1/2 knockout (Lats1/2iΔLgr5), specifically activated YAP1 and TAZ in Lgr5+ epithelial stem cells induced GC initiation via transcriptional upregulation of MYC, whereas inhibition of MYC significantly attenuated the YAP1/TAZ-driven GC development both in vitro and in vivo [16]. Transactivation of SLC35B4 induced by the YAP1-TEAD complex also led to GC progression [58]. Moreover, YAP1 is bound to peroxisome proliferator-activated receptors (PPARs) for SOX9 transactivation. It further caused enhanced tumorigenic function of GC [59]. Qiao et al. demonstrated that overexpression of YAP1 potentiated GC tumor metastasis through modulating cytoskeletal arrangement [60]. YAP1 induced RhoA suppressor ARHGAP29 transcription to promote actin depolymerization by turning F-actin into G-actin, thus causing enhanced metastatic capability of GC [60]. In addition, YAP1 was reported to potentiate GC cell survival, migration, and invasion by inhibiting endoplasmic reticulum stress via ERK signaling [61].
The main regulatory mechanisms for the Hippo pathway and the direct YAP1 downstream targets in gastric carcinogenesis have been summarized in Table 1.

3.4. Therapeutic Potential of Targeting the Hippo Signaling in GC

To date, chemotherapy has acted as the standard treatment for advanced GC; however, the acquired resistance has become a major clinical challenge since it vastly limits the therapeutic efficiency [63,64,65,66]. Recent studies have reported that YAP knockdown enhanced the sensitivity of GC cells to cisplatin, indicating that targeting YAP may improve the chemotherapeutic efficacy of GC [67].
Enhanced stability and nuclear accumulation of YAP1 induced by EphA2 mediated the 5-fluorouracil (5-FU) resistance in GC both in vitro and in vivo [68]. Furthermore, annexin A6 in cancer-associated fibroblast extracellular vesicles (CAF-EV) conferred cisplatin-resistant phenotypes in GC cells by activating the FAK-YAP1 signaling via stabilizing β1 integrin at the cell surface [69]. Pharmacological inhibition of YAP1 could effectively reverse the CAF-EV-induced drug resistance [69]. Trastuzumab is the only FDA-approved drug for the treatment of HER2-positive GC. YAP1 was highly expressed within trastuzumab-resistant GC tumors, where YAP1 activated by HER4 promoted EMT and subsequent trastuzumab resistance [70]. However, the detailed mechanism behind YAP-driven resistance warrants further investigation.

4. Hippo Pathway and Other Digestive Malignancies

In addition to GC, other malignancies in the digestive system (also known as GI cancer), such as colorectal, liver, esophageal, and pancreatic cancers, also threaten millions of patients’ lives [71]. Similar to GC, dysregulated Hippo signatures are commonly found in these GI cancers and have been proved to drive their malignant features. In this section, we summarized the main mechanisms of Hippo signaling pathway and its core components in other GI malignancies, apart from GC (Table 2).
Colorectal cancer (CRC): MST1/2 and LATS1/2 were downregulated in CRC, while the overexpressed YAP1 and TAZ were predominantly localized in the nucleus and consistently elevated in human CRCs from multiple cohorts [72]. In particular, increased YAP1 expression was positively associated with CRC patients’ clinicopathological characteristics, such as nodal status and TNM stage [86]. Previous studies have revealed that overexpressed YAP1 in human CRC cells was mainly attributed to dysregulated Wnt/β-catenin signaling through a mechanism by which β-catenin and TCF4 in a complex bound onto the DNA enhancer of YAP1 to induce its transactivation [78].
LATS1 functions as a tumor suppressor to diminish CRC growth and migration via glioma-associated oncogene homolog 1 (Gli1) downregulation [87]. Phosphorylation of MST1 and LATS2 promoted by metallothionein 2A (MT2A) restrained the YAP1 nuclear localization, thereby preventing CRC from enhanced tumor growth and liver metastasis [88]. In most CRC cases, upregulated YAP/TAZ commonly exerts oncogenic functions. Knockdown YAP or TAZ inhibited the ability of CRC cells to proliferate, form colonies, migrate, and invade, while co-silencing YAP and TAZ displayed synergistic inhibitory effects [77]. Mechanistically, YAP mainly drives CRC proliferation by activating the β-catenin signaling, thereby promoting the expansion of the colon CSCs population [75]. The increase in YAP enables KRAS-dependent CRC cell survival under KRAS suppression, as evidenced by ectopic expression of YAP1 in KRAS-mutated CRC cell lines could effectively restore cell viability [89]. YAP1-mediated transcription program of EMT is also responsible for potentiating the carcinogenetic effect of KRAS [89]. In addition, the Lats2/Sav1/YAP1/HIF1A signaling modulated by miR-103a-3p also controlled the glycolysis within CRC tumors through modulating glycolytic enzymes (e.g., HK2, PFK1, and LDHA) [90]. However, it is worth noting that aberrant YAP1 and TAZ have recently been considered tumor suppressors in CRC in several studies [91,92,93,94].
Liver cancer: Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC) are the most common primary liver cancers that account for the majority of all cases [95]. The nuclear abundance of YAP1 was commonly observed in human HCC samples [7], possibly due to the amplification of YAP1 on 11q22 amplicon [96]. Zhou et al. demonstrated that since Mst1/2 are canonical YAP regulators, depletion of Mst1/2 abolished the phosphorylation of YAP and thus promoted YAP-driven liver enlargement and hepatocarcinogenesis [73]. RNA-binding motif protein 3 (RBM3) induced YAP1 upregulation and potentiated the cell proliferation of HCC [80]. Ubiquitin-specific peptidase 10 (USP10) was reported to interact with YAP1 and TAZ to suppress their ubiquitination, resulting in accelerated HCC tumor growth [97]. By contrast, the YAP1 activity suppressed by miR-375 or miR-590-5P led to impaired cell proliferation and invasion [81,82]. Furthermore, as we know, hepatitis B virus (HBV) infection is a critical driver of HCC initiation. It has been demonstrated that HBV X protein (HBx) transcriptionally upregulated YAP1 expression in a cAMP-response element binding protein (CREB)-dependent manner [98]. Consistently, hepatocytes expressing HBV preΔS2 mutation caused ER dysfunction and YAP1 activation, leading to inflammation, fibrosis, hepatomegaly, and eventually hepatocarcinogenesis in vivo [99].
In addition to HCC, ubiquitous activation of TAZ was observed in human ICC, another subtype of primary liver cancer. Using a mice model with TAZ and AKT coactivation (AKT/TAZ mice), Cigliano et al. identified that TAZS89A and AKT collaboratively contributed to ICC through a mechanism by which TAZ interacted with TEAD to induce the transdifferentiation of mature hepatocytes. At the same time, the blockade of the TAZ-TEAD binding merely led to an enrichment of lipid-rich giant cells [100]. Moreover, the knockdown of YAP1 also delayed the formation of AKT/TAZ-initiated ICC [100].
Translationally, YAP1 and TAZ are also involved in sorafenib resistance, the first-line targeted drug for advanced HCC [101,102]. YAP1/TAZ coordinated with ATF4 to transactivate SLC7A11, which further conferred HCC resistance to sorafenib [101]. Moreover, this process could also be driven by YAP1-mediated survival expression [102]. Inhibition of the Hippo-YAP1 signaling using verteporfin (VP) in combination with transcatheter arterial chemoembolization (TACE), a commonly used therapeutic approach for unresectable HCC, allows reduced tumor burden and improved survival in a transplanted HCC mice model [103].
Esophageal cancer (EC): EC cell lines and human EC samples often express constitutively active YAP1 and TAZ [104], which is significantly correlated with poor survival and adverse clinicopathological features such as histological classification and tumor stage [105]. Four YAP1 isoforms (α, ß, γ, δ) were detected in human ESCC cell lines, all of which are essential for cell proliferation [105]. Depletion of YAP1 suppressed cell proliferation, migration, and invasion but enhanced cell apoptosis in vitro and inhibited tumor growth in vivo [83,104]. YAP1 and Gli1 have been demonstrated to form a positive feedback loop to promote EC development, where Gli1 upregulated YAP1 by suppressing LATS1-mediated phosphorylation, which increased Gli1 level through direct regulation of PI3K/AKT pathway [83]. Moreover, TAZ displayed similar clinical significance and oncogenic functions as YAP1 did in EC [106,107] and was responsible for tribbles pseudokinase 3 (TRIB3)-driven cancer stemness and radioresistance of ESCC [106,108].
Pancreatic ductal adenocarcinoma (PDAC): Several studies have reported that YAP1 and TAZ are overexpressed and hyperactivated in human PDACs, facilitating the proliferation and metastasis of PC cells. Knocking down the expression of YAP1 decreased cell growth and colony formation in PC cell lines [109]. YAP1 can also regulate the expression of genes associated with secreted factors, which collectively maintain the neoplastic proliferation and tumorigenic stromal responses of the tumor microenvironment [110]. Moreover, increased YAP1 promoted the EMT in PC cells via activation of the AKT pathway and conferred resistance to gemcitabine [111]. YAP1/TAZ have been demonstrated to be downstream effectors in the malignant transformation of both the KRAS pathway and the mitogen-activated protein kinase (MAPK) pathway. KRAS mutations occur in about 90% of PDAC patients and have been well studied to promote PDAC initiation in transgenic models by stimulating YAP1/TAZ to trigger JAK-STAT3 signaling activation [85]. Activation of JAK-STAT3 then modulated the inflammatory cytokines’ response and thus promoted the progression from pancreatic intraepithelial neoplasias (PanIN) to PDAC in KRasG12D mice [85]. Overall, these studies suggest that targeting YAP1/TAZ might provide clinical benefits to patients with PC.

5. Clinical Significance of Hippo Signatures in GI Cancers

To comprehensively characterize the genetic and epigenetic alterations of Hippo signature genes in GI cancers, we analyzed the genomics data of YAP1 and TAZ in different GI cancer types from the TCGA PanCancer database (Figure 3). For example, among the 405 GC patients, YAP1 and TAZ showed 8% and 12% of genetic alterations and mRNA upregulation, respectively. By single-cell RNA-seq analysis of data provided by Stanford Medicine [112], YAP1 was found predominantly expressed in cancer cells, fibroblasts, and endothelial cells, while TAZ expression was diffusely distributed with slightly higher expression in T cells [113] (Figure 4).
Clinically, YAP1 and TAZ can be used as diagnostic and prognostic tools values as their upregulation is not only responsible for early tumor detection but also predicts a worse prognosis in patients suffering from GI cancers [76,114]. In GC, there was a gradual increase of approximately three-fold in the ratio of YAP1-to-VGLL4 protein expression from GC grade I to grade IV, and their mRNA ratio was closely associated with aggressive tumor features [25]. For YAP1-positive GC, those with positive VGLL4 had significantly worse survival than those with negative VGLL4, suggesting that a combination of YAP1 and VGLL4 showed superior efficacy for GC surveillance [25]. TAZ alone is also capable of predicting tumor metastasis for patients diagnosed with gastric cardia adenocarcinoma [29]. In CRC, YAP1 and TAZ are independent prognostic factors associated with poor clinical outcomes [77,115]. Notably, CRC patients with high levels of YAP1 and TAZ experienced the comparatively shortest overall survival, suggesting that the combination of YAP1 and TAZ achieved the highest predictive value compared to YAP1 or TAZ alone [77]. In addition to YAP1/TAZ, serum MST1 level also aids early CRC detection, which showed high sensitivity and specificity of 92.3% and 100% when combined with carcinoembryonic antigen (CEA) and the fecal occult blood test (FOBT) [116].
In addition, YAP1 and TAZ could monitor therapy resistance, as elevated YAP1 expression was observed in 5-FU-resistant CRC and EC cell lines cells as well as in doxorubicin-resistant HCC HepG2 and Huh7 cells. The acquired resistance possibly arose from the YAP1/TAZ-induced EGFR upregulation [82,117,118,119]. In addition to chemoresistance, YAP1 and TAZ are also implicated in developing radiotherapy resistance [120,121,122]. Thus, YAP1 and TAZ may be utilized as an indicator of chemo- or radiotherapy response to select the optimal treatment for cancer patients.
In summary, the main Hippo components with aberrant expression in GI cancers can serve as prognostic or predictive biomarkers, especially indicating chemo- or radioresistance and tumor recurrence.

6. Therapeutic Strategies for Directly Targeting the Hippo Pathway

Given the crucial role of Hippo signaling in driving malignancies, many inhibitors targeting different Hippo components have been developed to provide new insights for cancer therapies.
Verteporfin (VP): Over 3300 compounds from the FDA-approved drug library underwent toxicological, pharmacokinetic, and pharmacodynamic tests in search of YAP1-specific inhibitors, where VP stood out as the initially discovered small molecule inhibitor [123,124]. VP has been demonstrated to inhibit YAP1/TAZ signaling and YAP1-potentiated oncogenic properties by disrupting the interaction between YAP1-TEAD in a transgenic murine model [124]. In GC, VP inhibited the expression of YAP1/TAZ-TEAD to suppress stem cell sphere formation and tumorigenesis [125]. Moreover, VP reduced the cyclinD1, MMP2, and Ang2 expression levels via blockade of the YAP1-TEAD complex, thus suppressing cell viability, angiogenesis, and angiogenic mimicry in PDAC [126]. In head and neck squamous carcinoma, VP downregulated the expression of EMT and CSC-associated genes through YAP1 in a light-dependent manner [127]. Strikingly, VP synergistically enhanced the antitumor effect of 5-FU in GC, as evidenced by significantly elevated cell apoptosis and reduced tumor growth in vivo [68].
CA3: Using the TEAD/YAP1 luciferase reporter assay, CA3 was recently initially identified to be a potent inhibitor of the YAP1-TEAD complex [128]. Functionally, CA3 suppressed the growth of YAP1high esophageal adenocarcinoma cells, mesothelioma cancer, and oral squamous epithelial cell carcinoma [129,130]. In addition, CA3 treatment restrained cancer stemness and tumorigenicity of cells derived from GC peritoneal metastases in a YAP1-dependent mechanism [31].
Statins: The anticancer effect of statins has recently been proposed to act by targeting YAP1/TAZ. Mechanistically, statins such as cerivastatin and simvastatin induce YAP1/TAZ phosphorylation and inhibit their nuclear translocation via blockade HMG-CoA conversion to mevalonic acid, thereby affecting the expression of their target genes [131,132,133]. The inhibition of statins on YAP1/TAZ activity could be strongly reinforced when combined with dasatinib [134]. These statins could functionally inhibit the development of pancreatic intraepithelial neoplasia [131,135].
Metformin: An AMP-activated protein kinase (AMPK) activator that has recently been proved to target the YAP1-TEAD complex in solid tumors. Metformin induced YAP1 phosphorylation to abolish the YAP1-dependent stem-like properties in glioma [136]. In addition, metformin can serve as the sensitizer of 5-FU in HCC through modulating YAP1 [137].
TEAD inhibitors: VGLL4 is a transcriptional co-factor that could bind onto TEAD via its Tondu (TDU) domains [138,139]. Recent studies demonstrated that VGLL4 exhibited tumor-suppressive roles in many cancer types, a process mainly depends on its competition with YAP1 for binding to TEAD [140]. Moreover, a peptide mimicking VGLL4 (also known as Super-TDU) has been shown to decrease GC growth by targeting the interaction of YAP1-TEAD [25].
Since auto-palmitoylation of TEADs at a conserved cysteine is essential for maintaining their stabilization and functional roles [141,142], researchers have found that MGH-CP1 could directly target the auto-palmitoylation of TEAD to inhibit YAP1/TAZ-mediated transcriptional activation. Compounds 1 functions by binding to TEAD lipid pockets and damaging S-palmitoylation [143]. By evaluating the binding domain of YAP1 and TEADs, Pobbati et al. indicated that the YAP1-TEAD complex showed a significantly high affinity to a nonsteroidal anti-inflammatory drug, fluphenazine. TED-347 and its derivatives disrupt the protein interaction between TEAD-YAP1, thus inhibiting TEAD transcriptional activity [143,144,145,146,147]. A high-throughput drug screening identified VT101 and VT102 as selective TEAD palmitoylation inhibitors in NF2-deficient mesothelioma [148]. One recent study verified that K-975, a novel TEAD inhibitor, showed potent and high-selective inhibitory effects on the YAP1/TAZ-TEAD interaction in malignant pleural mesothelioma [149]. All these small molecules could inhibit YAP1-driven malignant properties by disrupting TEAD activity, suggesting their potential as candidates for promising cancer therapies.
Other compounds potentially target the Hippo pathway for GC treatment: Tyrosine kinase inhibitors, such as dasatinib and pazopanib, have been shown to elicit phosphorylation and degradation of YAP1/TAZ for their cytoplasmic sequestration [150]. The therapeutic efficacy of these tyrosine kinase inhibitors also relies on the expression and activity of YAP1/TAZ. In GC SNU-1 and SNU-484 cells, 3,3′-Diindolylmethane exerted the antitumor effect by promoting the binding of RASSF1 with the MST1/2-LATS1-Mob1 complex, which further led to Hippo activation and YAP1 phosphorylation [151].
As indicated in Table 3, we summarized some current clinical studies of selected Hippo pathway inhibitors in GI cancers, including GC (https://clinicaltrials.gov, accessed on 6 September 2022).

7. Conclusions

Investigations on the Hippo signaling pathway field have rapidly expanded since Kieran F. Harvey first discovered it nearly 20 years ago. With the evidence available now, the Hippo signaling is not only dysregulated in GC and other GI cancers but also confers multiple pro-tumorigenic traits such as sustaining cell proliferation, survival, and metastasis for tumor progression. YAP1/TAZ acts as a determining effector of the Hippo pathway to drive tumorigenesis. From the diagnosis and prognosis aspects, the decreased expression of Hippo kinases and increased YAP1 and TAZ within tumor tissues serve as indicators for clinical outcomes of GI cancer patients. In addition, tumoral Hippo components could provide intriguing tools for early detection and therapeutic strategies of cancers.

8. Discussion

The past research has focused on the molecular mechanisms of the Hippo pathway, especially on the mRNA/protein upregulation or signal transduction. Meanwhile, as prognostic or predictive biomarkers, the main Hippo components have been highlighted in GI cancer. However, the current knowledge is far from presenting the whole image of the Hippo pathway in GI cancers as more mechanisms and hallmarks have emerged, such as the microbiota in shaping the Hippo pathway, the Hippo pathway in regulating the tumor microenvironment, etc. Meanwhile, with the development of high technologies and concepts, such as single-cell RNA sequencing and tumor evolution, it is urgent to differentiate the aberrant activation of Hippo components in different cell types and how they communicate with each other to drive carcinogenesis. For small molecule inhibitor development, endeavors are required to identify more potent inhibitors with fewer side effects, as so far, no small molecule has been successfully approved for clinical use.

9. Future Directions

To successfully translate the current knowledge from basic studies into clinical practice, we need to address several questions: First, in addition to what is known, it is still unclear which transcription factors predominantly activate YAP1/TAZ transcriptionally to drive GC progression in the H. pylori-positive cases. Meanwhile, the downstream effectors of the Hippo pathway that facilitate GC cancer initiation and progression should be further identified and investigated. Second, characterizing the specific mechanism behind Hippo-mediated therapy resistance, with the aim of improving the efficacy of chemo- and radiotherapies, is urgently needed. In addition, the potential role of Hippo signaling in modulating other tumor-facilitating properties (e.g., metabolic reprogramming, immune escape, and maintaining CSC properties) of GC remains unclear. Third but not least, YAP1 and TAZ have been shown to exert opposite functions in specific cancer types pending further clarification. For example, in CRC, many studies have reported that YAP1 and TAZ function as oncoproteins, while growing evidence suggests overexpressed YAP1 and TAZ could prevent CRC development [91,92,93,94].
Taken together, the dysregulated Hippo pathway plays a pivotal role in GI cancer progression; serves as a diagnostic, predictive, and surveillance biomarker; and provides a potential therapeutic target for GI tumor eradication. A deep investigation of the Hippo pathway in GI malignancies will not only broaden our knowledge about tumor progression but also provide novel therapeutic strategies for eliminating cancer.

Author Contributions

Conceptualization, K.F.T. and W.K.; writing—original draft preparation, X.L. and Y.W.; writing—review and editing, A.H.K.C., J.Y., W.K. and X.L.; visualization, B.C., W.N.C., C.W.M., J.Z. and K.Y.W.; project administration, K.F.T.; funding acquisition, W.K. All authors have read and agreed to the published version of the manuscript.

Funding

This study is supported by National Natural Science Foundation of China (NSFC) (2022, No. 82272990), the Research Grants Council of the Hong Kong Special Administrative Region (GRF Project No.: CUHK 14100019), Health and Medical Research Fund from Hong Kong Food and Health Bureau (HMRF Project No.: 09203216), and CUHK Direct Grant for Research (2020.001) from The Chinese University of Hong Kong.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are available within the manuscript.

Acknowledgments

We acknowledge the BioRender (https://biorender.com/, accessed on 6 May 2022) and TCGA Research Network (http://cancergenome.nih.gov/, accessed on 6 June 2022) for providing the datasets and analysis. We also acknowledge the technical support from Core Utilities of Cancer Genomics and Pathobiology of the Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-FU, 5-fluorouracil; AMPK, AMP-activated protein kinase; CAF-EV, cancer-associated fibroblast extracellular vesicles; CagA, cytotoxin-associated gene A; CEA, carcinoembryonic antigen; CRC, colorectal cancer; CREB, cAMP-response element binding protein; CSCs, cancer stemness cells; EC, esophageal cancer; EMT, epithelial-to-mesenchymal transition; FOBT, fecal occult blood test; GC, gastric cancer; Gli1, glioma-associated oncogene homolog 1; HBV, hepatitis B virus; HBx, HBV X protein; H. pylori, Helicobacter pylori; HCC, hepatocellular carcinoma; HMG-CoA, hydroxy-3-methylglutaryl coenzyme A; ICC, intrahepatic cholangiocarcinoma; LATS1/2, large tumor suppressor1/2; LIF, leukaemia inhibitory factor; MAPK, mitogen-activated protein kinase; MNNG, N-methyl-N′-nitro-N-nitrosoguanidine; MOB1, MOB kinase activator 1; MSS, microsatellite stability; MST1/2, mammalian Ste20-like kinases 1/2; MT2A, metallothionein 2A; PanIN, pancreatic intraepithelial neoplasias; PAR1, protease-activated receptor 1; PDAC, pancreatic ductal adenocarcinoma; PP2A, protein phosphatase 2A; PPARs, peroxisome proliferator-activated receptors; RBM3, RNA binding motif protein 3; SAV1, salvador homolog 1; TACE, transcatheter arterial chemoembolization; TAZ, WW domain-containing transcription regulator 1; TDU, Tondu; TEADs, TEA domain transcription factors; TRIB3, tribbles pseudokinase 3; USP10, ubiquitin-specific peptidase 10; VGLL4, vestigial-like family member 4; VP, verteporfin; YAP1, yes-associated protein 1; ZEB1, zinc finger E-box-binding homeobox 1.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [Green Version]
  2. Harvey, K.F.; Pfleger, C.M.; Hariharan, I.K. The Drosophila Mst ortholog, hippo, restricts growth and cell proliferation and promotes apoptosis. Cell 2003, 114, 457–467. [Google Scholar] [CrossRef] [Green Version]
  3. Staley, B.K.; Irvine, K.D. Hippo signaling in Drosophila: Recent advances and insights. Dev. Dyn. 2012, 241, 3–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yu, F.X.; Zhao, B.; Guan, K.L. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell 2015, 163, 811–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Miranda, M.M.; Lowry, W.E. Hip to the Game: YAP/TAZ is required for nonmelanoma skin cancers. EMBO J. 2018, 37, e99921. [Google Scholar] [CrossRef] [PubMed]
  6. Yu, F.X.; Guan, K.L. The Hippo pathway: Regulators and regulations. Genes Dev. 2013, 27, 355–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Zhao, B.; Wei, X.; Li, W.; Udan, R.S.; Yang, Q.; Kim, J.; Xie, J.; Ikenoue, T.; Yu, J.; Li, L.; et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 2007, 21, 2747–2761. [Google Scholar] [CrossRef] [Green Version]
  8. Zhao, B.; Li, L.; Tumaneng, K.; Wang, C.Y.; Guan, K.L. A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCF (beta-TRCP). Genes Dev. 2010, 24, 72–85. [Google Scholar] [CrossRef] [Green Version]
  9. Meng, Z.; Moroishi, T.; Guan, K.L. Mechanisms of Hippo pathway regulation. Genes Dev. 2016, 30, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Hong, A.W.; Meng, Z.; Guan, K.L. The Hippo pathway in intestinal regeneration and disease. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 324–337. [Google Scholar] [CrossRef] [PubMed]
  11. Fu, V.; Plouffe, S.W.; Guan, K.L. The Hippo pathway in organ development, homeostasis, and regeneration. Curr. Opin. Cell Biol. 2017, 49, 99–107. [Google Scholar] [CrossRef]
  12. Kim, M.K.; Jang, J.W.; Bae, S.C. DNA binding partners of YAP/TAZ. BMB Rep. 2018, 51, 126–133. [Google Scholar] [CrossRef] [Green Version]
  13. Hansen, C.G.; Moroishi, T.; Guan, K.L. YAP and TAZ: A nexus for Hippo signaling and beyond. Trends Cell Biol. 2015, 25, 499–513. [Google Scholar] [CrossRef] [Green Version]
  14. Overholtzer, M.; Zhang, J.; Smolen, G.A.; Muir, B.; Li, W.; Sgroi, D.C.; Deng, C.X.; Brugge, J.S.; Haber, D.A. Transforming properties of YAP, a candidate oncogene on the chromosome 11q22 amplicon. Proc. Natl. Acad. Sci. USA 2006, 103, 12405–12410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Zanconato, F.; Cordenonsi, M.; Piccolo, S. YAP/TAZ at the Roots of Cancer. Cancer Cell 2016, 29, 783–803. [Google Scholar] [CrossRef] [Green Version]
  16. Choi, W.; Kim, J.; Park, J.; Lee, D.H.; Hwang, D.; Kim, J.H.; Ashktorab, H.; Smoot, D.; Kim, S.Y.; Choi, C.; et al. YAP/TAZ Initiates Gastric Tumorigenesis via Upregulation of MYC. Cancer Res. 2018, 78, 3306–3320. [Google Scholar] [CrossRef] [Green Version]
  17. Zhang, X.; Tang, J.Z.; Vergara, I.A.; Zhang, Y.; Szeto, P.; Yang, L.; Mintoff, C.; Colebatch, A.; McIntosh, L.; Mitchell, K.A.; et al. Somatic Hypermutation of the YAP Oncogene in a Human Cutaneous Melanoma. Mol. Cancer Res. MCR 2019, 17, 1435–1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Moroishi, T.; Hansen, C.G.; Guan, K.L. The emerging roles of YAP and TAZ in cancer. Nat. Rev. Cancer 2015, 15, 73–79. [Google Scholar] [CrossRef] [PubMed]
  19. Kapoor, A.; Yao, W.; Ying, H.; Hua, S.; Liewen, A.; Wang, Q.; Zhong, Y.; Wu, C.J.; Sadanandam, A.; Hu, B.; et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell 2014, 158, 185–197. [Google Scholar] [CrossRef] [Green Version]
  20. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  21. Zhang, J.; Xu, Z.P.; Yang, Y.C.; Zhu, J.S.; Zhou, Z.; Chen, W.X. Expression of Yes-associated protein in gastric adenocarcinoma and inhibitory effects of its knockdown on gastric cancer cell proliferation and metastasis. Int. J. Immunopathol. Pharm. 2012, 25, 583–590. [Google Scholar] [CrossRef]
  22. Zhou, Z.; Zhu, J.S.; Xu, Z.P.; Zhang, Q. Lentiviral vector-mediated siRNA knockdown of the YAP gene inhibits growth and induces apoptosis in the SGC7901 gastric cancer cell line. Mol. Med. Rep. 2011, 4, 1075–1082. [Google Scholar] [CrossRef] [PubMed]
  23. Yao, S.; Yan, W. Overexpression of Mst1 reduces gastric cancer cell viability by repressing the AMPK-Sirt3 pathway and activating mitochondrial fission. OncoTargets Ther. 2018, 11, 8465–8479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Camargo, F.D.; Gokhale, S.; Johnnidis, J.B.; Fu, D.; Bell, G.W.; Jaenisch, R.; Brummelkamp, T.R. YAP1 increases organ size and expands undifferentiated progenitor cells. Curr. Biol. 2007, 17, 2054–2060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Jiao, S.; Wang, H.; Shi, Z.; Dong, A.; Zhang, W.; Song, X.; He, F.; Wang, Y.; Zhang, Z.; Wang, W.; et al. A peptide mimicking VGLL4 function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell 2014, 25, 166–180. [Google Scholar] [CrossRef] [Green Version]
  26. Jiang, C.-G.; Lv, L.; Liu, F.-R.; Wang, Z.-N.; Liu, F.-N.; Li, Y.-S.; Wang, C.-Y.; Zhang, H.-Y.; Sun, Z.; Xu, H.-M. Downregulation of connective tissue growth factor inhibits the growth and invasion of gastric cancer cells and attenuates peritoneal dissemination. Mol. Cancer 2011, 10, 122. [Google Scholar] [CrossRef] [Green Version]
  27. Tu, S.P.; Jiang, X.H.; Lin, M.C.; Cui, J.T.; Yang, Y.; Lum, C.T.; Zou, B.; Zhu, Y.B.; Jiang, S.H.; Wong, W.M. Suppression of survivin expression inhibits in vivo tumorigenicity and angiogenesis in gastric cancer. Cancer Res. 2003, 63, 7724–7732. [Google Scholar]
  28. Kang, W.; Tong, J.H.; Chan, A.W.; Lee, T.L.; Lung, R.W.; Leung, P.P.; So, K.K.; Wu, K.; Fan, D.; Yu, J.; et al. Yes-associated protein 1 exhibits oncogenic property in gastric cancer and its nuclear accumulation associates with poor prognosis. Clin. Cancer Res. 2011, 17, 2130–2139. [Google Scholar] [CrossRef] [Green Version]
  29. Wei, J.; Wang, L.; Zhu, J.; Sun, A.; Yu, G.; Chen, M.; Huang, P.; Liu, H.; Shao, G.; Yang, W.; et al. The Hippo signaling effector WWTR1 is a metastatic biomarker of gastric cardia adenocarcinoma. Cancer Cell Int. 2019, 19, 74. [Google Scholar] [CrossRef]
  30. Li, M.; Rao, X.; Cui, Y.; Zhang, L.; Li, X.; Wang, B.; Zheng, Y.; Teng, L.; Zhou, T.; Zhuo, W. The keratin 17/YAP/IL6 axis contributes to E-cadherin loss and aggressiveness of diffuse gastric cancer. Oncogene 2022, 41, 770–781. [Google Scholar] [CrossRef]
  31. Ajani, J.A.; Xu, Y.; Huo, L.; Wang, R.; Li, Y.; Wang, Y.; Pizzi, M.P.; Scott, A.; Harada, K.; Ma, L.; et al. YAP1 mediates gastric adenocarcinoma peritoneal metastases that are attenuated by YAP1 inhibition. Gut 2021, 70, 55–66. [Google Scholar] [CrossRef]
  32. Zhang, Y.; Bai, J.; Cheng, R.; Zhang, D.; Qiu, Z.; Liu, T.; Che, N.; Dong, X.; Zhao, N.; Lin, X.; et al. TAZ promotes vasculogenic mimicry in gastric cancer through the upregulation of TEAD4. J. Gastroenterol. Hepatol. 2022, 37, 714–726. [Google Scholar] [CrossRef] [PubMed]
  33. Lee, Y.; Finch-Edmondson, M.; Cognart, H.; Zhu, B.; Song, H.; Low, B.C.; Sudol, M. Common and Unique Transcription Signatures of YAP and TAZ in Gastric Cancer Cells. Cancers 2020, 12, 3667. [Google Scholar] [CrossRef] [PubMed]
  34. Yao, Y.; Liu, Z.; Huang, S.; Huang, C.; Cao, Y.; Li, L.; Guo, H.; Liu, F.; Huang, S.; Liao, Q.; et al. The E3 ubiquitin ligase, FBXW5, promotes the migration and invasion of gastric cancer through the dysregulation of the Hippo pathway. Cell Death Discov. 2022, 8, 79. [Google Scholar] [CrossRef]
  35. Hum, M.; Tan, H.J.; Yang, Y.; Srivastava, S.; Teh, M.M.; Lim, Y.P. WBP2 promotes gastric cancer cell migration via novel targeting of LATS2 kinase in the Hippo tumor suppressor pathway. FASEB J. 2021, 35, e21290. [Google Scholar] [CrossRef]
  36. Tang, Y.; Fang, G.; Guo, F.; Zhang, H.; Chen, X.; An, L.; Chen, M.; Zhou, L.; Wang, W.; Ye, T.; et al. Selective Inhibition of STRN3-Containing PP2A Phosphatase Restores Hippo Tumor-Suppressor Activity in Gastric Cancer. Cancer Cell 2020, 38, 115–128.e9. [Google Scholar] [CrossRef]
  37. Seeneevassen, L.; Giraud, J.; Molina-Castro, S.; Sifre, E.; Tiffon, C.; Beauvoit, C.; Staedel, C.; Megraud, F.; Lehours, P.; Martin, O.C.B.; et al. Leukaemia Inhibitory Factor (LIF) Inhibits Cancer Stem Cells Tumorigenic Properties through Hippo Kinases Activation in Gastric Cancer. Cancers 2020, 12, 2011. [Google Scholar] [CrossRef]
  38. Fujimoto, D.; Ueda, Y.; Hirono, Y.; Goi, T.; Yamaguchi, A. PAR1 participates in the ability of multidrug resistance and tumorigenesis by controlling Hippo-YAP pathway. Oncotarget 2015, 6, 34788–34799. [Google Scholar] [CrossRef] [PubMed]
  39. Jiao, S.; Guan, J.; Chen, M.; Wang, W.; Li, C.; Wang, Y.; Cheng, Y.; Zhou, Z. Targeting IRF3 as a YAP agonist therapy against gastric cancer. J. Exp. Med. 2018, 215, 699–718. [Google Scholar] [CrossRef]
  40. Zhou, Y.; Zhang, J.; Li, H.; Huang, T.; Wong, C.C.; Wu, F.; Wu, M.; Weng, N.; Liu, L.; Cheng, A.S.L.; et al. AMOTL1 enhances YAP1 stability and promotes YAP1-driven gastric oncogenesis. Oncogene 2020, 39, 4375–4389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Qiao, Y.; Lin, S.J.; Chen, Y.; Voon, D.C.; Zhu, F.; Chuang, L.S.; Wang, T.; Tan, P.; Lee, S.C.; Yeoh, K.G.; et al. RUNX3 is a novel negative regulator of oncogenic TEAD-YAP complex in gastric cancer. Oncogene 2016, 35, 2664–2674. [Google Scholar] [CrossRef]
  42. An, L.; Nie, P.; Chen, M.; Tang, Y.; Zhang, H.; Guan, J.; Cao, Z.; Hou, C.; Wang, W.; Zhao, Y.; et al. MST4 kinase suppresses gastric tumorigenesis by limiting YAP activation via a non-canonical pathway. J. Exp. Med. 2020, 217, e20191817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yin, K.; Dang, S.; Cui, L.; Fan, X.; Wang, L.; Xie, R.; Qu, J.; Shang, M.; Chen, J.; Xu, Z. Netrin-1 promotes metastasis of gastric cancer by regulating YAP activity. Biochem. Biophys Res. Commun. 2018, 496, 76–82. [Google Scholar] [CrossRef] [PubMed]
  44. Luo, J.; Xiang, H. LncRNA MYLK-AS1 acts as an oncogene by epigenetically silencing large tumor suppressor 2 (LATS2) in gastric cancer. Bioengineered 2021, 12, 3101–3112. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, J.; Huang, F.; Shi, Y.; Zhang, Q.; Xu, S.; Yao, Y.; Jiang, R. RP11-323N12.5 promotes the malignancy and immunosuppression of human gastric cancer by increasing YAP1 transcription. Gastric Cancer 2021, 24, 85–102. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, H.; Di, X.; Bi, Y.; Sun, S.; Wang, T. Long non-coding RNA LINC00649 regulates YES-associated protein 1 (YAP1)/Hippo pathway to accelerate gastric cancer (GC) progression via sequestering miR-16-5p. Bioengineered 2021, 12, 1791–1802. [Google Scholar] [CrossRef] [PubMed]
  47. Zuo, Q.F.; Zhang, R.; Li, B.S.; Zhao, Y.L.; Zhuang, Y.; Yu, T.; Gong, L.; Li, S.; Xiao, B.; Zou, Q.M. MicroRNA-141 inhibits tumor growth and metastasis in gastric cancer by directly targeting transcriptional co-activator with PDZ-binding motif, TAZ. Cell Death Dis. 2015, 6, e1623. [Google Scholar] [CrossRef] [PubMed]
  48. Kang, W.; Tong, J.H.; Lung, R.W.; Dong, Y.; Zhao, J.; Liang, Q.; Zhang, L.; Pan, Y.; Yang, W.; Pang, J.C.; et al. Targeting of YAP1 by microRNA-15a and microRNA-16-1 exerts tumor suppressor function in gastric adenocarcinoma. Mol. Cancer 2015, 14, 52. [Google Scholar] [CrossRef] [Green Version]
  49. Deng, J.; Lei, W.; Xiang, X.; Zhang, L.; Yu, F.; Chen, J.; Feng, M.; Xiong, J. MicroRNA-506 inhibits gastric cancer proliferation and invasion by directly targeting Yap1. Tumor Biol. 2015, 36, 6823–6831. [Google Scholar] [CrossRef] [PubMed]
  50. Sun, D.; Wang, Y.; Wang, H.; Xin, Y. The novel long non-coding RNA LATS2-AS1-001 inhibits gastric cancer progression by regulating the LATS2/YAP1 signaling pathway via binding to EZH2. Cancer Cell Int. 2020, 20, 204. [Google Scholar] [CrossRef]
  51. Zhang, J.; Liu, H.; Hou, L.; Wang, G.; Zhang, R.; Huang, Y.; Chen, X.; Zhu, J. Circular RNA_LARP4 inhibits cell proliferation and invasion of gastric cancer by sponging miR-424-5p and regulating LATS1 expression. Mol. Cancer 2017, 16, 151. [Google Scholar] [CrossRef] [Green Version]
  52. Molina-Castro, S.E.; Tiffon, C.; Giraud, J.; Boeuf, H.; Sifre, E.; Giese, A.; Belleannee, G.; Lehours, P.; Bessede, E.; Megraud, F.; et al. The Hippo Kinase LATS2 Controls Helicobacter pylori-Induced Epithelial-Mesenchymal Transition and Intestinal Metaplasia in Gastric Mucosa. Cell Mol. Gastroenterol. Hepatol. 2020, 9, 257–276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Li, N.; Feng, Y.; Hu, Y.; He, C.; Xie, C.; Ouyang, Y.; Artim, S.C.; Huang, D.; Zhu, Y.; Luo, Z.; et al. Helicobacter pylori CagA promotes epithelial mesenchymal transition in gastric carcinogenesis via triggering oncogenic YAP pathway. J. Exp. Clin. Cancer Res. 2018, 37, 280. [Google Scholar] [CrossRef] [Green Version]
  54. Wu, Y.; Shen, L.; Liang, X.; Li, S.; Ma, L.; Zheng, L.; Li, T.; Yu, H.; Chan, H.; Chen, C.; et al. Helicobacter pylori-induced YAP1 nuclear translocation promotes gastric carcinogenesis by enhancing IL-1beta expression. Cancer Med. 2019, 8, 3965–3980. [Google Scholar] [CrossRef] [Green Version]
  55. Tiffon, C.; Giraud, J.; Molina-Castro, S.E.; Peru, S.; Seeneevassen, L.; Sifre, E.; Staedel, C.; Bessede, E.; Dubus, P.; Megraud, F.; et al. TAZ Controls Helicobacter pylori-Induced Epithelial-Mesenchymal Transition and Cancer Stem Cell-Like Invasive and Tumorigenic Properties. Cells 2020, 9, 1462. [Google Scholar] [CrossRef]
  56. Zhou, W.; Xian, Q.; Wang, Q.; Wu, C.; Yan, H.; Li, X.; Lu, L.; Wu, C.; Zhu, D.; Xu, X.; et al. m6A Methyltransferase 3 Promotes the Proliferation and Migration of Gastric Cancer Cells through the m6A Modification of YAP1. J. Oncol. 2021, 2021, 8875424. [Google Scholar] [CrossRef]
  57. Jang, M.; An, J.; Oh, S.W.; Lim, J.Y.; Kim, J.; Choi, J.K.; Cheong, J.H.; Kim, P. Matrix stiffness epigenetically regulates the oncogenic activation of the Yes-associated protein in gastric cancer. Nat. Biomed. Eng. 2021, 5, 114–123. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, J.; Zhao, X.; Wang, K.; Zhang, X.; Yu, Y.; Lv, Y.; Zhang, S.; Zhang, L.; Guo, Y.; Li, Y.; et al. A novel YAP1/SLC35B4 regulatory axis contributes to proliferation and progression of gastric carcinoma. Cell Death Dis. 2019, 10, 452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Song, S.; Wang, Z.; Li, Y.; Ma, L.; Jin, J.; Scott, A.W.; Xu, Y.; Estrella, J.S.; Song, Y.; Liu, B.; et al. PPARdelta Interacts with the Hippo Coactivator YAP1 to Promote SOX9 Expression and Gastric Cancer Progression. Mol. Cancer Res. 2020, 18, 390–402. [Google Scholar] [CrossRef] [PubMed]
  60. Qiao, Y.; Chen, J.; Lim, Y.B.; Finch-Edmondson, M.L.; Seshachalam, V.P.; Qin, L.; Jiang, T.; Low, B.C.; Singh, H.; Lim, C.T.; et al. YAP Regulates Actin Dynamics through ARHGAP29 and Promotes Metastasis. Cell Rep. 2017, 19, 1495–1502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Liu, H.; Mei, D.; Xu, P.; Wang, H.; Wang, Y. YAP promotes gastric cancer cell survival and migration/invasion via the ERK/endoplasmic reticulum stress pathway. Oncol. Lett. 2019, 18, 6752–6758. [Google Scholar] [CrossRef]
  62. Guo, H.; Zou, J.; Zhou, L.; Zhong, M.; He, Y.; Huang, S.; Chen, J.; Li, J.; Xiong, J.; Fang, Z.; et al. NUSAP1 Promotes Gastric Cancer Tumorigenesis and Progression by Stabilizing the YAP1 Protein. Front. Oncol. 2020, 10, 591698. [Google Scholar] [CrossRef]
  63. Zeng, J.F.; Ma, X.Q.; Wang, L.P.; Wang, W. MicroRNA-145 exerts tumor-suppressive and chemo-resistance lowering effects by targeting CD44 in gastric cancer. World J. Gastroenterol. 2017, 23, 2337–2345. [Google Scholar] [CrossRef]
  64. Zhang, P.F.; Sheng, L.L.; Wang, G.; Tian, M.; Zhu, L.Y.; Zhang, R.; Zhang, J.; Zhu, J.S. miR-363 promotes proliferation and chemo-resistance of human gastric cancer via targeting of FBW7 ubiquitin ligase expression. Oncotarget 2016, 7, 35284–35292. [Google Scholar] [CrossRef]
  65. Cunningham, D.; Starling, N.; Rao, S.; Iveson, T.; Nicolson, M.; Coxon, F.; Middleton, G.; Daniel, F.; Oates, J.; Norman, A.R. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N. Engl. J. Med. 2008, 358, 36–46. [Google Scholar] [CrossRef] [Green Version]
  66. Russi, S.; Verma, H.K.; Laurino, S.; Mazzone, P.; Storto, G.; Nardelli, A.; Zoppoli, P.; Calice, G.; La Rocca, F.; Sgambato, A.; et al. Adapting and Surviving: Intra and Extra-Cellular Remodeling in Drug-Resistant Gastric Cancer Cells. Int. J. Mol. Sci. 2019, 20, 3736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lu, T.; Sun, L.; Zhu, X. Yes-associated protein enhances proliferation and attenuates sensitivity to cisplatin in human gastric cancer cells. Biomed. Pharmacother. Biomed. Pharmacother. 2018, 105, 1269–1275. [Google Scholar] [CrossRef]
  68. Huang, C.; Yuan, W.; Lai, C.; Zhong, S.; Yang, C.; Wang, R.; Mao, L.; Chen, Z.; Chen, Z. EphA2-to-YAP pathway drives gastric cancer growth and therapy resistance. Int. J. Cancer 2020, 146, 1937–1949. [Google Scholar] [CrossRef] [PubMed]
  69. Uchihara, T.; Miyake, K.; Yonemura, A.; Komohara, Y.; Itoyama, R.; Koiwa, M.; Yasuda, T.; Arima, K.; Harada, K.; Eto, K.; et al. Extracellular Vesicles from Cancer-Associated Fibroblasts Containing Annexin A6 Induces FAK-YAP Activation by Stabilizing beta1 Integrin, Enhancing Drug Resistance. Cancer Res. 2020, 80, 3222–3235. [Google Scholar] [CrossRef] [PubMed]
  70. Shi, J.; Li, F.; Yao, X.; Mou, T.; Xu, Z.; Han, Z.; Chen, S.; Li, W.; Yu, J.; Qi, X.; et al. The HER4-YAP1 axis promotes trastuzumab resistance in HER2-positive gastric cancer by inducing epithelial and mesenchymal transition. Oncogene 2018, 37, 3022–3038. [Google Scholar] [CrossRef] [Green Version]
  71. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  72. Liang, K.; Zhou, G.; Zhang, Q.; Li, J.; Zhang, C. Expression of hippo pathway in colorectal cancer. Saudi J. Gastroenterol. Off. J. Saudi Gastroenterol. Assoc. 2014, 20, 188–194. [Google Scholar] [CrossRef]
  73. Zhou, D.; Conrad, C.; Xia, F.; Park, J.S.; Payer, B.; Yin, Y.; Lauwers, G.Y.; Thasler, W.; Lee, J.T.; Avruch, J.; et al. Mst1 and Mst2 maintain hepatocyte quiescence and suppress hepatocellular carcinoma development through inactivation of the Yap1 oncogene. Cancer Cell 2009, 16, 425–438. [Google Scholar] [CrossRef] [Green Version]
  74. Yi, J.; Lu, L.; Yanger, K.; Wang, W.; Sohn, B.H.; Stanger, B.Z.; Zhang, M.; Martin, J.F.; Ajani, J.A.; Chen, J.J.H. Large tumor suppressor homologs 1 and 2 regulate mouse liver progenitor cell proliferation and maturation through antagonism of the coactivators YAP and TAZ. Hepatology 2016, 64, 1757–1772. [Google Scholar] [CrossRef] [Green Version]
  75. Avruch, J.; Zhou, D.; Bardeesy, N. YAP oncogene overexpression supercharges colon cancer proliferation. Cell Cycle 2012, 11, 1090–1096. [Google Scholar] [CrossRef] [Green Version]
  76. Yuen, H.-F.; McCrudden, C.M.; Huang, Y.-H.; Tham, J.M.; Zhang, X.; Zeng, Q.; Zhang, S.-D.; Hong, W.J. TAZ expression as a prognostic indicator in colorectal cancer. PLoS ONE 2013, 8, e54211. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, L.; Shi, S.; Guo, Z.; Zhang, X.; Han, S.; Yang, A.; Wen, W.; Zhu, Q. Overexpression of YAP and TAZ is an independent predictor of prognosis in colorectal cancer and related to the proliferation and metastasis of colon cancer cells. PLoS ONE 2013, 8, e65539. [Google Scholar] [CrossRef] [Green Version]
  78. Konsavage, W.M.; Kyler, S.L.; Rennoll, S.A.; Jin, G.; Yochum, G.S. Wnt/β-catenin signaling regulates Yes-associated protein (YAP) gene expression in colorectal carcinoma cells. JBC 2012, 287, 11730–11739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Park, Y.Y.; Sohn, B.H.; Johnson, R.L.; Kang, M.H.; Kim, S.B.; Shim, J.J.; Mangala, L.S.; Kim, J.H.; Yoo, J.E.; Rodriguez-Aguayo, C.; et al. Yes-associated protein 1 and transcriptional coactivator with PDZ-binding motif activate the mammalian target of rapamycin complex 1 pathway by regulating amino acid transporters in hepatocellular carcinoma. Hepatology 2016, 63, 159–172. [Google Scholar] [CrossRef] [Green Version]
  80. Miao, X.; Zhang, N. Role of RBM3 in the regulation of cell proliferation in hepatocellular carcinoma. Exp. Mol. Pathol. 2020, 117, 104546. [Google Scholar] [CrossRef]
  81. Liu, A.M.; Poon, R.T.P.; Luk, J.M. MicroRNA-375 targets Hippo-signaling effector YAP in liver cancer and inhibits tumor properties. Biochem. Biophys. Res. Commun. 2010, 394, 623–627. [Google Scholar] [CrossRef]
  82. Chen, M.; Wu, L.; Tu, J.; Zhao, Z.; Fan, X.; Mao, J.; Weng, Q.; Wu, X.; Huang, L.; Xu, M.; et al. miR-590-5p suppresses hepatocellular carcinoma chemoresistance by targeting YAP1 expression. EBioMedicine 2018, 35, 142–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Wang, C.; Cheng, L.; Song, S.; Wu, S.; Sun, G. Gli1 interacts with YAP1 to promote tumorigenesis in esophageal squamous cell carcinoma. J. Cell. Physiol. 2020, 235, 8224–8235. [Google Scholar] [CrossRef]
  84. Zhou, Q.; Bauden, M.; Andersson, R.; Hu, D.; Marko-Varga, G.; Xu, J.; Sasor, A.; Dai, H.; Pawłowski, K.; Said Hilmersson, K.; et al. YAP1 is an independent prognostic marker in pancreatic cancer and associated with extracellular matrix remodeling. J. Transl. Med. 2020, 18, 77. [Google Scholar] [CrossRef]
  85. Gruber, R.; Panayiotou, R.; Nye, E.; Spencer-Dene, B.; Stamp, G.; Behrens, A. YAP1 and TAZ Control Pancreatic Cancer Initiation in Mice by Direct Up-regulation of JAK–STAT3 Signaling. Gastroenterology 2016, 151, 526–539. [Google Scholar] [CrossRef] [Green Version]
  86. Wang, Y.; Xie, C.; Li, Q.; Xu, K.; Wang, E. Clinical and prognostic significance of Yes-associated protein in colorectal cancer. Tumor Biol. 2013, 34, 2169–2174. [Google Scholar] [CrossRef]
  87. Shen, Z.; Pan, Y.; Chen, P.; Jiang, B.; Fang, X.; Jiang, Y. LATS1 exerts tumor suppressor functions via targeting Gli1 in colorectal cancer. J. Cancer 2021, 12, 7311–7319. [Google Scholar] [CrossRef]
  88. Liu, X.; Quan, J.; Shen, Z.; Zhang, Z.; Chen, Z.; Li, L.; Li, X.; Hu, G.; Deng, X. Metallothionein 2A (MT2A) controls cell proliferation and liver metastasis by controlling the MST1/LATS2/YAP1 signaling pathway in colorectal cancer. Cancer Cell Int 2022, 22, 205. [Google Scholar] [CrossRef]
  89. 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]
  90. Sun, Z.; Zhang, Q.; Yuan, W.; Li, X.; Chen, C.; Guo, Y.; Shao, B.; Dang, Q.; Zhou, Q.; Wang, Q.; et al. MiR-103a-3p promotes tumour glycolysis in colorectal cancer via hippo/YAP1/HIF1A axis. J. Exp. Clin. Cancer Res. 2020, 39, 250. [Google Scholar] [CrossRef]
  91. Cheung, P.; Xiol, J.; Dill, M.T.; Yuan, W.C.; Panero, R.; Roper, J.; Osorio, F.G.; Maglic, D.; Li, Q.; Gurung, B.; et al. Regenerative Reprogramming of the Intestinal Stem Cell State via Hippo Signaling Suppresses Metastatic Colorectal Cancer. Cell Stem Cell 2020, 27, 590–604.e9. [Google Scholar] [CrossRef]
  92. Zhang, S.; Wei, Q.; Yang, Y.Z.; Qin, H.L.; Li, X.X.; Cai, S.J.; Ma, Y.L. Loss of Yes-associated Protein Represents an Aggressive Subtype of Colorectal Cancer. J. Cancer 2019, 10, 689–696. [Google Scholar] [CrossRef]
  93. Strano, S.; Monti, O.; Pediconi, N.; Baccarini, A.; Fontemaggi, G.; Lapi, E.; Mantovani, F.; Damalas, A.; Citro, G.; Sacchi, A.; et al. The transcriptional coactivator Yes-associated protein drives p73 gene-target specificity in response to DNA Damage. Mol. Cell 2005, 18, 447–459. [Google Scholar] [CrossRef]
  94. Lapi, E.; Di Agostino, S.; Donzelli, S.; Gal, H.; Domany, E.; Rechavi, G.; Pandolfi, P.P.; Givol, D.; Strano, S.; Lu, X.; et al. PML, YAP, and p73 Are Components of a Proapoptotic Autoregulatory Feedback Loop. Mol. Cell 2008, 32, 803–814. [Google Scholar] [CrossRef]
  95. 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]
  96. Zender, L.; Spector, M.S.; Xue, W.; Flemming, P.; Cordon-Cardo, C.; Silke, J.; Fan, S.T.; Luk, J.M.; Wigler, M.; Hannon, G.J.; et al. Identification and validation of oncogenes in liver cancer using an integrative oncogenomic approach. Cell 2006, 125, 1253–1267. [Google Scholar] [CrossRef] [Green Version]
  97. Zhu, H.; Yan, F.; Yuan, T.; Qian, M.; Zhou, T.; Dai, X.; Cao, J.; Ying, M.; Dong, X.; He, Q.; et al. USP10 Promotes Proliferation of Hepatocellular Carcinoma by Deubiquitinating and Stabilizing YAP/TAZ. Cancer Res. 2020, 80, 2204. [Google Scholar] [CrossRef] [Green Version]
  98. Zhang, T.; Zhang, J.; You, X.; Liu, Q.; Du, Y.; Gao, Y.; Shan, C.; Kong, G.; Wang, Y.; Yang, X.; et al. Hepatitis B virus X protein modulates oncogene Yes-associated protein by CREB to promote growth of hepatoma cells. Hepatology 2012, 56, 2051–2059. [Google Scholar] [CrossRef] [PubMed]
  99. Teng, Y.C.; Neo, J.C.; Wu, J.C.; Chen, Y.F.; Kao, C.H.; Tsai, T.F. Expression of a hepatitis B virus pre-S2 deletion mutant in the liver results in hepatomegaly and hepatocellular carcinoma in mice. J. Pathol. 2017, 241, 463–474. [Google Scholar] [CrossRef]
  100. Cigliano, A.; Zhang, S.; Ribback, S.; Steinmann, S.; Sini, M.; Ament, C.E.; Utpatel, K.; Song, X.; Wang, J.; Pilo, M.G.; et al. The Hippo pathway effector TAZ induces intrahepatic cholangiocarcinoma in mice and is ubiquitously activated in the human disease. J. Exp. Clin. Cancer Res. 2022, 41, 192. [Google Scholar] [CrossRef]
  101. Gao, R.; Kalathur, R.K.R.; Coto-Llerena, M.; Ercan, C.; Buechel, D.; Shuang, S.; Piscuoglio, S.; Dill, M.T.; Camargo, F.D.; Christofori, G.; et al. YAP/TAZ and ATF4 drive resistance to Sorafenib in hepatocellular carcinoma by preventing ferroptosis. EMBO Mol. Med. 2021, 13, e14351. [Google Scholar] [CrossRef] [PubMed]
  102. Sun, T.; Mao, W.; Peng, H.; Wang, Q.; Jiao, L. YAP promotes sorafenib resistance in hepatocellular carcinoma by upregulating survivin. Cell Oncol. 2021, 44, 689–699. [Google Scholar] [CrossRef] [PubMed]
  103. Quan, Y.; Li, Z.; Zhu, K.; Liang, J. Transcatheter arterial chemoembolization combined with Hippo/YAP inhibition significantly improve the survival of rats with transplanted hepatocellular carcinoma. Lipids Health Dis. 2021, 20, 74. [Google Scholar] [CrossRef]
  104. Zhao, J.; Li, X.; Yang, Y.; Zhu, D.; Zhang, C.; Liu, D.; Wu, K.; Zhao, S. Effect of YAP1 silencing on esophageal cancer. Onco. Targets Ther. 2016, 9, 3137–3146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Muramatsu, T.; Imoto, I.; Matsui, T.; Kozaki, K.-i.; Haruki, S.; Sudol, M.; Shimada, Y.; Tsuda, H.; Kawano, T.; Inazawa, J. YAP is a candidate oncogene for esophageal squamous cell carcinoma. Carcinogenesis 2010, 32, 389–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Zhou, W.; Zhang, L.; Chen, P.; Li, S.; Cheng, Y. Thymine DNA glycosylase-regulated TAZ promotes radioresistance by targeting nonhomologous end joining and tumor progression in esophageal cancer. Cancer Sci. 2020, 111, 3613–3625. [Google Scholar] [CrossRef]
  107. Liu, L.; Lu, Z.; Hu, X.; Su, T.; Su, L.; Pu, H. Clinical significance of YAP1 and TAZ in esophageal squamous cell carcinoma. Medicine 2021, 100, e26597. [Google Scholar] [CrossRef]
  108. Zhou, S.; Liu, S.; Lin, C.; Li, Y.; Ye, L.; Wu, X.; Jian, Y.; Dai, Y.; Ouyang, Y.; Zhao, L.; et al. TRIB3 confers radiotherapy resistance in esophageal squamous cell carcinoma by stabilizing TAZ. Oncogene 2020, 39, 3710–3725. [Google Scholar] [CrossRef]
  109. Diep, C.H.; Zucker, K.M.; Hostetter, G.; Watanabe, A.; Hu, C.; Munoz, R.M.; Von Hoff, D.D.; Han, H. Down-Regulation of Yes Associated Protein 1 Expression Reduces Cell Proliferation and Clonogenicity of Pancreatic Cancer Cells. PLoS ONE 2012, 7, e32783. [Google Scholar] [CrossRef] [Green Version]
  110. Zhang, W.; Nandakumar, N.; Shi, Y.; Manzano, M.; Smith, A.; Graham, G.; Gupta, S.; Vietsch, E.E.; Laughlin, S.Z.; Wadhwa, M.; et al. Downstream of mutant KRAS, the transcription regulator YAP is essential for neoplastic progression to pancreatic ductal adenocarcinoma. Sci. Signal. 2014, 7, ra42. [Google Scholar] [CrossRef] [Green Version]
  111. Yuan, Y.; Li, D.; Li, H.; Wang, L.; Tian, G.; Dong, Y. YAP overexpression promotes the epithelial-mesenchymal transition and chemoresistance in pancreatic cancer cells. Mol. Med. Rep. 2016, 13, 237–242. [Google Scholar] [CrossRef]
  112. Sathe, A.; Grimes, S.M.; Lau, B.T.; Chen, J.; Suarez, C.; Huang, R.J.; Poultsides, G.; Ji, H.P. Single-Cell Genomic Characterization Reveals the Cellular Reprogramming of the Gastric Tumor Microenvironment. Clin. Cancer Res. 2020, 26, 2640–2653. [Google Scholar] [CrossRef] [Green Version]
  113. Butler, A.; Hoffman, P.; Smibert, P.; Papalexi, E.; Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 2018, 36, 411–420. [Google Scholar] [CrossRef]
  114. Van Haele, M.; Moya, I.M.; Karaman, R.; Rens, G.; Snoeck, J.; Govaere, O.; Nevens, F.; Verslype, C.; Topal, B.; Monbaliu, D.; et al. YAP and TAZ Heterogeneity in Primary Liver Cancer: An Analysis of Its Prognostic and Diagnostic Role. Int. J. Mol. Sci. 2019, 20, 638. [Google Scholar] [CrossRef] [Green Version]
  115. Zhou, D.; Zhang, Y.; Wu, H.; Barry, E.; Yin, Y.; Lawrence, E.; Dawson, D.; Willis, J.E.; Markowitz, S.D.; Camargo, F.D.; et al. Mst1 and Mst2 protein kinases restrain intestinal stem cell proliferation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Proc. Natl. Acad. Sci. USA 2011, 108, E1312–E1320. [Google Scholar] [CrossRef] [Green Version]
  116. Yu, J.; Zhai, X.; Li, X.; Zhong, C.; Guo, C.; Yang, F.; Yuan, Y.; Zheng, S. Identification of MST1 as a potential early detection biomarker for colorectal cancer through a proteomic approach. Sci. Rep. 2017, 7, 14265. [Google Scholar] [CrossRef] [Green Version]
  117. Touil, Y.; Igoudjil, W.; Corvaisier, M.; Dessein, A.-F.; Vandomme, J.; Monté, D.; Stechly, L.; Skrypek, N.; Langlois, C.; Grard, G.; et al. Colon Cancer Cells Escape 5FU Chemotherapy-Induced Cell Death by Entering Stemness and Quiescence Associated with the c-Yes/YAP Axis. Clin. Cancer Res. 2014, 20, 837. [Google Scholar] [CrossRef] [Green Version]
  118. Song, S.; Honjo, S.; Jin, J.; Chang, S.-S.; Scott, A.W.; Chen, Q.; Kalhor, N.; Correa, A.M.; Hofstetter, W.L.; Albarracin, C.T.; et al. The Hippo Coactivator YAP1 Mediates EGFR Overexpression and Confers Chemoresistance in Esophageal Cancer. Clin. Cancer Res. 2015, 21, 2580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Huang, C.; Chen, Z.; Yang, C.; Chen, L.; Lai, C.; Zhang, Y.; Yuan, W.; Jeong, J.H. Combinational inhibition of EGFR and YAP reverses 5-Fu resistance in colorectal cancer. J. Cancer 2020, 11, 5432–5439. [Google Scholar] [CrossRef] [PubMed]
  120. Ciamporcero, E.; Shen, H.; Ramakrishnan, S.; Yu Ku, S.; Chintala, S.; Shen, L.; Adelaiye, R.; Miles, K.M.; Ullio, C.; Pizzimenti, S.; et al. YAP activation protects urothelial cell carcinoma from treatment-induced DNA damage. Oncogene 2016, 35, 1541–1553. [Google Scholar] [CrossRef] [Green Version]
  121. Fernandez-L, A.; Squatrito, M.; Northcott, P.; Awan, A.; Holland, E.C.; Taylor, M.D.; Nahlé, Z.; Kenney, A.M. Oncogenic YAP promotes radioresistance and genomic instability in medulloblastoma through IGF2-mediated Akt activation. Oncogene 2012, 31, 1923–1937. [Google Scholar] [CrossRef]
  122. Akervall, J.; Nandalur, S.; Zhang, J.; Qian, C.-N.; Goldstein, N.; Gyllerup, P.; Gardinger, Y.; Alm, J.; Lorenc, K.; Nilsson, K.; et al. A novel panel of biomarkers predicts radioresistance in patients with squamous cell carcinoma of the head and neck. Eur. J. Cancer 2014, 50, 570–581. [Google Scholar] [CrossRef]
  123. Johnson, R.; Halder, G. The two faces of Hippo: Targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat. Rev. Drug Discov. 2014, 13, 63–79. [Google Scholar] [CrossRef] [Green Version]
  124. Liu-Chittenden, Y.; Huang, B.; Shim, J.S.; Chen, Q.; Lee, S.J.; Anders, R.A.; Liu, J.O.; Pan, D. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev. 2012, 26, 1300–1305. [Google Scholar] [CrossRef] [Green Version]
  125. Giraud, J.; Molina-Castro, S.; Seeneevassen, L.; Sifré, E.; Izotte, J.; Tiffon, C.; Staedel, C.; Boeuf, H.; Fernandez, S.; Barthelemy, P.; et al. Verteporfin targeting YAP1/TAZ-TEAD transcriptional activity inhibits the tumorigenic properties of gastric cancer stem cells. Mol. Cancer Biol. 2020, 146, 2255–2267. [Google Scholar] [CrossRef]
  126. Wei, H.; Wang, F.; Wang, Y.; Li, T.; Xiu, P.; Zhong, J.; Sun, X.; Li, J. Verteporfin suppresses cell survival, angiogenesis and vasculogenic mimicry of pancreatic ductal adenocarcinoma via disrupting the YAP-TEAD complex. Cancer Sci. 2017, 108, 478–487. [Google Scholar] [CrossRef] [Green Version]
  127. Liu, K.; Du, S.; Gao, P.; Zheng, J. Verteporfin suppresses the proliferation, epithelial-mesenchymal transition and stemness of head and neck squamous carcinoma cells via inhibiting YAP1. J. Cancer 2019, 10, 4196–4207. [Google Scholar] [CrossRef] [Green Version]
  128. Song, S.; Xie, M.; Scott, A.W.; Jin, J.; Ma, L.; Dong, X.; Skinner, H.D.; Johnson, R.L.; Ding, S.; Ajani, J.A. A Novel YAP1 Inhibitor Targets CSC-Enriched Radiation-Resistant Cells and Exerts Strong Antitumor Activity in Esophageal Adenocarcinoma. Mol. Cancer Ther. 2018, 17, 443–454. [Google Scholar] [CrossRef] [Green Version]
  129. Kandasamy, S.; Adhikary, G.; Rorke, E.A.; Friedberg, J.S.; Mickle, M.B.; Alexander, H.R.; Eckert, R.L. The YAP1 Signaling Inhibitors, Verteporfin and CA3, Suppress the Mesothelioma Cancer Stem Cell Phenotype. Mol. Cancer Res. MCR 2020, 18, 343–351. [Google Scholar] [CrossRef]
  130. Qi, L.; Shi, C.; Li, J.; Xu, S.; Han, Y.; Li, J.; Zhang, L. Yes-associated protein promotes cell migration via activating Wiskott-Aldrich syndrome protein family member 1 in oral squamous cell carcinoma. J. Oral Pathol. Med. Off. Publ. Int. Assoc. Oral Pathol. Am. Acad. Oral Pathol. 2019, 48, 290–298. [Google Scholar] [CrossRef]
  131. Sorrentino, G.; Ruggeri, N.; Specchia, V.; Cordenonsi, M.; Mano, M.; Dupont, S.; Manfrin, A.; Ingallina, E.; Sommaggio, R.; Piazza, S.; et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat. Cell Biol. 2014, 16, 357–366. [Google Scholar] [CrossRef] [PubMed]
  132. Gronich, N.; Rennert, G. Beyond aspirin—Cancer prevention with statins, metformin and bisphosphonates. Nat. Rev. Clin. Oncol. 2013, 10, 625–642. [Google Scholar] [CrossRef]
  133. Wang, Z.; Wu, Y.; Wang, H.; Zhang, Y.; Mei, L.; Fang, X.; Zhang, X.; Zhang, F.; Chen, H.; Liu, Y.; et al. Interplay of mevalonate and Hippo pathways regulates RHAMM transcription via YAP to modulate breast cancer cell motility. Proc. Natl. Acad. Sci. USA 2014, 111, E89. [Google Scholar] [CrossRef] [Green Version]
  134. Taccioli, C.; Sorrentino, G.; Zannini, A.; Caroli, J.; Beneventano, D.; Anderlucci, L.; Lolli, M.; Bicciato, S.; Del Sal, G. MDP, a database linking drug response data to genomic information, identifies dasatinib and statins as a combinatorial strategy to inhibit YAP/TAZ in cancer cells. Oncotarget 2015, 6, 38854–38865. [Google Scholar] [CrossRef] [Green Version]
  135. Hao, F.; Xu, Q.; Wang, J.; Yu, S.; Chang, H.H.; Sinnett-Smith, J.; Eibl, G.; Rozengurt, E. Lipophilic statins inhibit YAP nuclear localization, co-activator activity and colony formation in pancreatic cancer cells and prevent the initial stages of pancreatic ductal adenocarcinoma in KrasG12D mice. PLoS ONE 2019, 14, e0216603. [Google Scholar] [CrossRef] [Green Version]
  136. Yuan, X.; Wei, W.; Bao, Q.; Chen, H.; Jin, P.; Jiang, W. Metformin inhibits glioma cells stemness and epithelial-mesenchymal transition via regulating YAP activity. Biomed. Pharmacother. 2018, 102, 263–270. [Google Scholar] [CrossRef]
  137. Tian, Y.; Tang, B.; Wang, C.; Sun, D.; Zhang, R.; Luo, N.; Han, Z.; Liang, R.; Gao, Z.; Wang, L. Metformin mediates resensitivity to 5-fluorouracil in hepatocellular carcinoma via the suppression of YAP. Oncotarget 2016, 7, 46230–46241. [Google Scholar] [CrossRef] [Green Version]
  138. Vaudin, P.; Delanoue, R.; Davidson, I.; Silber, J.; Zider, A. TONDU (TDU), a novel human protein related to the product of vestigial (vg) gene of Drosophila melanogaster interacts with vertebrate TEF factors and substitutes for Vg function in wing formation. Development 1999, 126, 4807–4816. [Google Scholar] [CrossRef]
  139. Zhang, W.; Gao, Y.; Li, P.; Shi, Z.; Guo, T.; Li, F.; Han, X.; Feng, Y.; Zheng, C.; Wang, Z.; et al. VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex. Cell Res. 2014, 24, 331–343. [Google Scholar] [CrossRef] [Green Version]
  140. Mickle, M.; Adhikary, G.; Shrestha, S.; Xu, W.; Eckert, R.L. VGLL4 inhibits YAP1/TEAD signaling to suppress the epidermal squamous cell carcinoma cancer phenotype. Mol. Carcinog. 2021, 60, 497–507. [Google Scholar] [CrossRef]
  141. Noland, C.L.; Gierke, S.; Schnier, P.D.; Murray, J.; Sandoval, W.N.; Sagolla, M.; Dey, A.; Hannoush, R.N.; Fairbrother, W.J.; Cunningham, C.N.J.S. Palmitoylation of TEAD transcription factors is required for their stability and function in Hippo pathway signaling. Structure 2016, 24, 179–186. [Google Scholar] [CrossRef]
  142. Chan, P.; Han, X.; Zheng, B.; DeRan, M.; Yu, J.; Jarugumilli, G.K.; Deng, H.; Pan, D.; Luo, X.; Wu, X. Autopalmitoylation of TEAD proteins regulates transcriptional output of the Hippo pathway. Nat. Chem. Biol. 2016, 12, 282–289. [Google Scholar] [CrossRef] [Green Version]
  143. Li, Q.; Sun, Y.; Jarugumilli, G.K.; Liu, S.; Dang, K.; Cotton, J.L.; Xiol, J.; Chan, P.Y.; DeRan, M.; Ma, L.; et al. Lats1/2 Sustain Intestinal Stem Cells and Wnt Activation through TEAD-Dependent and Independent Transcription. Cell Stem Cell 2020, 26, 675–692.e8. [Google Scholar] [CrossRef]
  144. Pobbati, A.V.; Han, X.; Hung, A.W.; Weiguang, S.; Huda, N.; Chen, G.-Y.; Kang, C.; Chia, C.S.B.; Luo, X.; Hong, W.; et al. Targeting the Central Pocket in Human Transcription Factor TEAD as a Potential Cancer Therapeutic Strategy. Structure 2015, 23, 2076–2086. [Google Scholar] [CrossRef] [Green Version]
  145. Holden, J.K.; Crawford, J.J.; Noland, C.L.; Schmidt, S.; Zbieg, J.R.; Lacap, J.A.; Zang, R.; Miller, G.M.; Zhang, Y.; Beroza, P.; et al. Small Molecule Dysregulation of TEAD Lipidation Induces a Dominant-Negative Inhibition of Hippo Pathway Signaling. Cell Rep. 2020, 31, 107809. [Google Scholar] [CrossRef]
  146. Lu, W.; Wang, J.; Li, Y.; Tao, H.; Xiong, H.; Lian, F.; Gao, J.; Ma, H.; Lu, T.; Zhang, D. Discovery and biological evaluation of vinylsulfonamide derivatives as highly potent, covalent TEAD autopalmitoylation inhibitors. Eur. J. Med. Chem. 2019, 184, 111767. [Google Scholar] [CrossRef]
  147. Bum-Erdene, K.; Zhou, D.; Gonzalez-Gutierrez, G.; Ghozayel, M.K.; Si, Y.; Xu, D.; Shannon, H.E.; Bailey, B.J.; Corson, T.W.; Pollok, K.E.; et al. Small-Molecule Covalent Modification of Conserved Cysteine Leads to Allosteric Inhibition of the TEAD⋅Yap Protein-Protein Interaction. Cell Chem. Biol. 2019, 26, 378–389.e13. [Google Scholar] [CrossRef]
  148. Tang, T.T.; Konradi, A.W.; Feng, Y.; Peng, X.; Ma, M.; Li, J.; Yu, F.-X.; Guan, K.-L.; Post, L. Small Molecule Inhibitors of TEAD Auto-palmitoylation Selectively Inhibit Proliferation and Tumor Growth of NF2-deficient Mesothelioma. Mol. Cancer Ther. 2021, 20, 986. [Google Scholar] [CrossRef]
  149. Kaneda, A.; Seike, T.; Danjo, T.; Nakajima, T.; Otsubo, N.; Yamaguchi, D.; Tsuji, Y.; Hamaguchi, K.; Yasunaga, M.; Nishiya, Y.; et al. The novel potent TEAD inhibitor, K-975, inhibits YAP1/TAZ-TEAD protein-protein interactions and exerts an anti-tumor effect on malignant pleural mesothelioma. Am. J. Cancer Res. 2020, 10, 4399–4415. [Google Scholar] [PubMed]
  150. Oku, Y.; Nishiya, N.; Shito, T.; Yamamoto, R.; Yamamoto, Y.; Oyama, C.; Uehara, Y. Small molecules inhibiting the nuclear localization of YAP/TAZ for chemotherapeutics and chemosensitizers against breast cancers. FEBS Open Bio. 2015, 5, 542–549. [Google Scholar] [CrossRef] [Green Version]
  151. Li, X.J.; Park, E.S.; Park, M.H.; Kim, S.M. 3,3′-Diindolylmethane suppresses the growth of gastric cancer cells via activation of the Hippo signaling pathway. Oncol. Rep. 2013, 30, 2419–2426. [Google Scholar] [CrossRef]
Figure 1. Core components of the Hippo pathway in human cells. When the Hippo pathway is ON, MST1/2 and SAV1 in complex phosphorylate LATS1/2 and MOB1/2. Activated LATS1/2 then phosphorylate YAP1 and TAZ, leading to YAP1/TAZ cytoplasmic retention or ubiquitin-mediated proteasomal degradation. On the other hand, when the Hippo pathway is OFF, dephosphorylated YAP1/TAZ translocate into the nucleus, where they bind with TEADs to induce the transcription of downstream target genes involved in cell growth and survival.
Figure 1. Core components of the Hippo pathway in human cells. When the Hippo pathway is ON, MST1/2 and SAV1 in complex phosphorylate LATS1/2 and MOB1/2. Activated LATS1/2 then phosphorylate YAP1 and TAZ, leading to YAP1/TAZ cytoplasmic retention or ubiquitin-mediated proteasomal degradation. On the other hand, when the Hippo pathway is OFF, dephosphorylated YAP1/TAZ translocate into the nucleus, where they bind with TEADs to induce the transcription of downstream target genes involved in cell growth and survival.
Biomedicines 10 02512 g001
Figure 2. The role of H. pylori in Hippo pathway remodeling. CagA-positive H. pylori infection activates YAP1 and TAZ. CagA promoted the nuclear YAP1 and TAZ levels inside gastric mucosa from gastritis, and intestinal metaplasia, to GC. The Hippo signature genes, such as CTGF, Cyr61, IL-1β (Interleukin-1β), LATS2, etc., are upregulated following H. pylori infection. Moreover, TAZ participates in H. pylori-driven EMT and gastric carcinogenesis. In the gastric mucosa of H. pylori-associated GC mouse models and patients, nuclear TAZ accumulation activated ZEB1 expression, potentiating the EMT and stemness.
Figure 2. The role of H. pylori in Hippo pathway remodeling. CagA-positive H. pylori infection activates YAP1 and TAZ. CagA promoted the nuclear YAP1 and TAZ levels inside gastric mucosa from gastritis, and intestinal metaplasia, to GC. The Hippo signature genes, such as CTGF, Cyr61, IL-1β (Interleukin-1β), LATS2, etc., are upregulated following H. pylori infection. Moreover, TAZ participates in H. pylori-driven EMT and gastric carcinogenesis. In the gastric mucosa of H. pylori-associated GC mouse models and patients, nuclear TAZ accumulation activated ZEB1 expression, potentiating the EMT and stemness.
Biomedicines 10 02512 g002
Figure 3. Genetic aberrations and mRNA upregulation of YAP1 and TAZ in gastrointestinal cancers. The TCGA PanCancer dataset illustrated the proportion of YAP1/TAZ alterations (including gene amplification, deep deletion, inframe and missense mutations, structural variants, and mRNA changes) in primary digestive cancers. Only positive cases with alterations were demonstrated in this Figure. COAD, colon adenocarcinoma; ESCA, esophageal carcinoma; LIHC, liver hepatocellular carcinoma; PAAD, pancreatic adenocarcinoma; STAD stomach adenocarcinoma.
Figure 3. Genetic aberrations and mRNA upregulation of YAP1 and TAZ in gastrointestinal cancers. The TCGA PanCancer dataset illustrated the proportion of YAP1/TAZ alterations (including gene amplification, deep deletion, inframe and missense mutations, structural variants, and mRNA changes) in primary digestive cancers. Only positive cases with alterations were demonstrated in this Figure. COAD, colon adenocarcinoma; ESCA, esophageal carcinoma; LIHC, liver hepatocellular carcinoma; PAAD, pancreatic adenocarcinoma; STAD stomach adenocarcinoma.
Biomedicines 10 02512 g003
Figure 4. The expression of YAP1 and TAZ in the GC microenvironment. By scRNA-seq analysis, YAP1 was predominantly expressed in cancer cells, cancer-associated fibroblasts, and endothelial cells, while the immune cells (T and B cells) demonstrated less YAP1/TAZ expression. TAZ expression was diffusely distributed in the different cell types, with slightly higher expression in T cells.
Figure 4. The expression of YAP1 and TAZ in the GC microenvironment. By scRNA-seq analysis, YAP1 was predominantly expressed in cancer cells, cancer-associated fibroblasts, and endothelial cells, while the immune cells (T and B cells) demonstrated less YAP1/TAZ expression. TAZ expression was diffusely distributed in the different cell types, with slightly higher expression in T cells.
Biomedicines 10 02512 g004
Table 1. Summary of upstream regulators and downstream targets of the Hippo pathway in GC.
Table 1. Summary of upstream regulators and downstream targets of the Hippo pathway in GC.
RegulatorsTargetsRegulatory MechanismsReferences
FBXW5LATS1FBXW5 inactivates the Hippo pathway by enhancing LATS1 ubiquitination and degradation, which promotes GC cell invasion, metastasis, and drug resistance.[34]
circLARP4circLARP4 increases miR-424 level, which in turn inhibits LATS1 translation.[51]
WBP2LATS2WBP2 directly interacts with LATS2 promoter to inhibit LATS2 and activate the YAP1-TEAD transcriptional activity.[35]
lncRNAsBoth MYLK-AS1 and LATS2-AS1-001 bind with EZH2 to induce LATS2 activation.[44,50]
LIFLATS1/2Activates LATS1/2 to suppress the malignant properties of CSCs.[37]
PAR1YAP1PAR1 activates YAP1 in a Rho-dependent manner.[38]
IRF3IRF3 strengthens the interaction between YAP1 and TEAD4 for YAP1 activation.[39]
AMOTL1AMOTL1 associates with YAP1 to prevent the ubiquitin-mediated YAP1 degradation, thus promoting its nuclear translocation and subsequent tumorigenic roles.[40]
MST4MST4 phosphorylates YAP1 at Thr83 to impede its interaction with importin α for YAP1 inactivation.[42]
Netrin-1Netrin-1 phosphorylates YAP1 via its receptor neogenin.[43]
RP11-323N12.5LncRNA RP11-323N12.5 binds with c-MYC to transactivate YAP1.[45]
METTL3METTL3 regulates the m6A modification of YAP1 for its activation.[56]
NUSAP1NUSAP1 stabilizes YAP1 protein through direct binding.[62]
KRT17KRT17 loss stimulates RhoA-dependent F-actin polymerization and induces YAP1 signaling activation and nuclear localization.[30]
microRNA-141TAZmicroRNA-141 directly downregulates TAZ.[47]
RUNX3TEADRUNX3 interacts with TEAD to abrogate the DNA binding ability of TEAD, thus limiting the oncogenic property of the TEAD-YAP1 complex in GC.[41]
H. PyloriHippo componentsControls the expression of MST1, LATS2, YAP1, and TAZ to promote EMT and other tumorigenic properties.-
Downstream Targets of YAP1 in GCRegulatory MechanismsReferences
YAP1MYCUpon YAP1 activation, activated YAP1 transcriptionally modulates MYC by binding to MYC enhancer and sequesters p72 to hinder miRNAs-mediated MYC suppression.[16]
IL-1ßYAP1-TEAD complex binds onto IL-1ß promoter to transcriptionally upregulate IL-1ß production, thus resulting in GC cell proliferation.[54]
SLC35B4YAP1-TEAD complex induces SLC35B4 transactivation via binding to the promoter region of SLC35B4.[58]
SOX9YAP1 binds to peroxisome proliferator-activated receptors (PPARs) to induce transcriptional regulation of SOX9, which further leads to enhanced cancer stemness and tumorigenicity.[31,59]
ARHGAP29YAP1 transcriptionally upregulates ARHGAP29 to suppress the RhoA-LIMK-cofilin pathway for F-actin destabilization. The YAP1-ARHGAP29 signaling is essential for GC metastasis.[60]
Table 2. Summary of the main Hippo signaling pathway core components in GI malignancies.
Table 2. Summary of the main Hippo signaling pathway core components in GI malignancies.
MembersCancer TypeExpression FunctionMechanismsReference
MST1/2CRCDownregulatedTumor suppressorDepletion of Mst1/2 driven CRC.[72]
HCCMst1/2 kinases are canonical YAP/TAZ regulators.[73]
LATS1/2CRCDownregulatedTumor suppressorLATS1/2 can inhibit CRC growth and metastasis via downregulating Gli1 expression and MT2A-mediated absence of YAP1 nuclear localization.[72,74,75]
HCCDepletion of LAST1/2 lead to hyperactivation of YAP/TAZ.[74]
YAP1/YAZCRCUpregulatedOncogeneThe expression of YAP1 is driven via aberrant Wnt/β-catenin signaling in human CRC cells.[75,76,77,78]
HCCYAP1/TAZ activates mTORC1 in HCC cells. The interaction of RBM3 -YAP1 is important for HCC proliferation, and USP10 was reported to inhibit YAP1/TAZ ubiquitinating. Overexpression of miR-375 and miR-590-5P inhibited cell proliferation and invasion by downregulating YAP1 activity.[79,80,81,82]
ECGli1 upregulated YAP1 with LATS1 independent mode. Simultaneously, YAP1 increased Gli1 expression by direct regulation PI3K/AKT pathway.[83]
PDACYAP1/TAZ acts as an essential downstream in the oncogenic transition of both KRAS and MAPK pathways. KRAS-mutation promotes PDAC initiation in transgenic models by activating YAP1/TAZ to upregulate JAK-STAT3 signaling activation.[84,85]
Table 3. Clinical trials on Hippo inhibitors-based therapy in GI cancers.
Table 3. Clinical trials on Hippo inhibitors-based therapy in GI cancers.
ClinicalTrials.gov IdentifierOfficial TitleCondition or DiseaseIntervention/TreatmentPhaseStatus
NCT00944463Trial of Simvastatin and Gemcitabine in pancreatic cancer patientsPDACDrug: Gemcitabine + simvastatin
Drug: Gemcitabine+Placebo
2Completed
NCT01813994Role of Statin on gastric inflammation in patients at high risk of gastric cancerEarly GC or gastric adenomaDrug: Arm1: Statin
Drug: Arm2: Placebo
Not applicableCompleted
NCT02968810Simvastatin in preventing liver cancer in patients with liver cirrhosisCirrhosisOther: Placebo Administration
Other: Questionnaire Administration
Drug: Simvastatin
2Active, not recruiting
NCT04947020dataBase for analysis of rectal cancer oncological results (BARO)Rectal cancerDrug: MetforminNot applicableRecruiting
NCT01099085Trial of XP (Capecitabine/CDDP) Simvastatin in advanced gastric cancer patientsGCDrug: Simvastatin
Drug: Placebo
3Completed
NCT02569645Rosuvastatin in the treatment of rectal cancerRectal cancerDrug: Rosuvastatin2Completed
NCT01075555Sorafenib Tosylate with or without pravastatin in treating patients with liver cancer and cirrhosisLiver cancerDrug: Pravastatin sodium
Drug: Sorafenib tosylate
3Completed
NCT02026583A Single Arm, Phase II study of Simvastatin Plus XELOX and Bevacizumab as first-line chemotherapy in metastatic CRC Patients CRCDrug: Simvastatin2Completed
NCT01281761Simvastatin + Cetuximab/Irinotecan in K-ras mutated CRCMetastatic CRCDrug: Cetuximab/irinotecan/simvastatin2Completed
NCT00313859Phase II Study of Simvastatin Plus Irinotecan, Fluorouracil, and Leucovorin (FOLFIRI) for metastatic CRCMetastasis CRC Drug: Simvastatin2Completed
NCT03889795Phase IB Metformin, Digoxin, Simvastatin in solid tumorsAdvanced PDACDrug: Metformin
Drug: Simvastatin
Drug: Digoxin
1Recruiting
NCT05368805Fruquintinib DDI study with P-gp and BCRP substratesMetastatic CRCDrug: Fruquintinib
Drug: Dabigatran Etexilate
Drug: Rosuvastatin
1Active, not recruiting
NCT04767984Testing Atorvastatin to lower colon cancer Risk in longstanding ulcerative colitisCRC and ulcerative colitisDrug: Atorvastatin Calcium2Recruiting
NCT03033225Ultrasound-Guided Verteporfin photodynamic therapy for the treatment of unresectable solid pancreatic tumors or advanced pancreatic cancer, VERTPAC-02 StudyAdvanced PDAC
Locally advanced PDAC
Metastatic PDAC
Drug: Photodynamic Therapy
Drug: Verteporfin
2Recruiting
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Liu, X.; Wang, Y.; Chen, B.; Chan, W.N.; Mui, C.W.; Cheung, A.H.K.; Zhang, J.; Wong, K.Y.; Yu, J.; Kang, W.; et al. Targeting the Hippo Pathway in Gastric Cancer and Other Malignancies in the Digestive System: From Bench to Bedside. Biomedicines 2022, 10, 2512. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10102512

AMA Style

Liu X, Wang Y, Chen B, Chan WN, Mui CW, Cheung AHK, Zhang J, Wong KY, Yu J, Kang W, et al. Targeting the Hippo Pathway in Gastric Cancer and Other Malignancies in the Digestive System: From Bench to Bedside. Biomedicines. 2022; 10(10):2512. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10102512

Chicago/Turabian Style

Liu, Xiaoli, Yifei Wang, Bonan Chen, Wai Nok Chan, Chun Wai Mui, Alvin H.K. Cheung, Jinglin Zhang, Kit Yee Wong, Jun Yu, Wei Kang, and et al. 2022. "Targeting the Hippo Pathway in Gastric Cancer and Other Malignancies in the Digestive System: From Bench to Bedside" Biomedicines 10, no. 10: 2512. https://0-doi-org.brum.beds.ac.uk/10.3390/biomedicines10102512

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