Open Access

Functional roles of E3 ubiquitin ligases in gastric cancer (Review)

  • Authors:
    • Mingliang Wang
    • Wei Dai
    • Zhangyan Ke
    • Yongxiang Li
  • View Affiliations

  • Published online on: July 16, 2020     https://doi.org/10.3892/ol.2020.11883
  • Article Number: 22
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

To date, >650 E3 ubiquitin ligases have been described in humans, including >600 really interesting new genes (RINGs), 28 homologous to E6‑associated protein C‑terminus (HECTs) and several RING‑in‑between‑RINGs. They are considered key regulators and therapeutic targets of many types of human cancers, including gastric cancer (GC). Among them, some RING and HECT E3 ligases are closely related to the proliferation, infiltration and prognosis of GC. During the past few years, abnormal expressions and functions of many E3 ligases have been identified in GC. However, the functional roles of E3 ligases in GC have not been fully elucidated. The present article focuses on the functional roles of E3 ligases related to the proteasome in GC. In this comprehensive review, the latest research progress on E3 ligases involved in GC and elaborate their structure, classification, functional roles and therapeutic value in GC was summarized. Finally, 30 E3 ligases that serve essential roles in regulating the development of GC were described. Some of these ligases may serve as oncogenes or tumor suppressors in GC, whereas the pathological mechanism of others needs further study; for example, constitutive photomorphogenic 1. In conclusion, the present review demonstrated that E3 ligases are crucial tumor regulatory factors and potential therapeutic targets in GC. Therefore, more studies should focus on the therapeutic targeting of E3 ligases in GC.

Introduction

Gastric cancer (GC) is the fourth most commonly diagnosed cancer and the third leading cause of cancer-related mortality worldwide (1). The pathogenic mechanisms and progression of GC are complex. Currently, well-known pathogenic factors include poor eating habits, chronic Helicobacter pylori infection and the misregulation of oncogenes or tumor suppressors (2). The 5-year survival rate of patients with GC is only 27.4% in China (3). The poor prognosis of GC is due to tumor invasion and metastasis (4,5), which are complex processes involving a series of cell biological regulations and requiring multi-step genetic mutations (6). The genes and their products involved in each step of tumor progression are potential prognostic markers and therapeutic targets. Among these biomarkers, E3 ligases play a crucial role in the proliferation, invasion and metastasis of GC (7).

The ubiquitin-proteasome system (UPS) is a common post-translational modification pathway involved in the regulation of cell survival and differentiation (8). The ubiquitination pathway is catalyzed by three types of key enzymes: Ubiquitin-activating enzyme (E1), ubiquitin-conjugating enzyme (E2) and ubiquitin ligases (E3) (9). E3 ligase is the most important component in the UPS owing to its specific ability to recognize target proteins and transfer ubiquitin to substrates for degradation (9,10). To date, >650 E3 ubiquitin ligases have been described in humans, and they can be subdivided into three different families: Homologous to E6-associated protein C-terminus (HECT), really interesting new gene (RING) and RING-in-between-RING (RBR) E3 ligases (11,12). However, RBR E3 ligases, an emerging group of E3 ligases that feature with characteristics of RING and HECT, have not been discovered in GC (13). Hence, RING and HECT E3 ligases are dysregulated in GC cells. Owing to the notable role of E3 ligases on the ubiquitination of tumor-associated signaling molecules, such as the AKT pathway, targeting E3 ligases could be an efficient approach in cancer treatment (14). Recently, many new types of E3 ligases have been increasingly detected in GC, such as RNF6 and RNF38 (15,16). Herein, a comprehensive review of studies is presented to summarize the latest progress and treatment prospects.

Classification of E3 ubiquitin ligases

As aforementioned, only RING- and HECT-type E3 ligases are involved in GC (Table I). The RING family comprises >600 members (17). The RING finger domain is an important component of RING-type E3 ligases and can be sub-divided into two groups: Typical and atypical. The typical conserved region RING domain harbors a RING fold structure coupled with zinc ions. Another atypical type, called the U-box domain, possesses extremely similar RING folds but lacks cysteine residues, which affects zinc ions coordination (18). Both RING and U-box E3 ligases can function as momodimers, heterodimers or multiple subunits (17,19). The cullin-RING ligase (CRL) is a kind of multi-subunit RING E3 ligase that can be further divided into the S phase kinase associated protein 1 (SKP1)/cullin 1 (CUL1)/F-box protein complex (SCF), CUL2-elongin B/C-VHL or SOCS proteins (CRL2), CUL3-BTBs, CUL4-DDB1-DCAFs (CRL4), CUL5-elonginB/C-SOCS proteins, and the CUL7/F-box/WD repeat-containing 8 (FBXW8) subfamily of E3 ligases (20,21).

Table I.

Characteristics of E3 ligase associated with gastric cancer.

Table I.

Characteristics of E3 ligase associated with gastric cancer.

Author, yearGene Family/SubfamilyExpressionSubstratesPathwayRefs
Che et al, 2017; Xu et al, 2017; Zhang et al, 2009Cbl-bRING/Cbl Up/downregulatedIGF-1R; c-Src,IGF-1R(67-69)
Kashima et al, 2012CHFRRING/NADownregulatedPARP-1NA(74)
Wang et al, 2017MARCH8RING/MARCHDownregulatedDR4JWA/MARCH8/DR4(43)
Ko A et al, 2012MKRN1RING/NADownregulatedp14ARF p14ARF-associated(76)
Yang et al, 2017PIRH2RING/PIRHUpregulatedP53p53(59)
Zhang et al, 2018RNF6RING/RNFUpregulatedSHP-1SHP-1/STAT3(15)
Huang et al, 2018RNF38RING/RNFUpregulatedSHP-1SHP-1/STAT3(16)
Gao et al, 2017; Zhou et al, 2013RNF43RING/RNFDownregulatedβ-catenin; Lgr5 Wnt/β-catenin/TCF(34,38)
Qui et al, 2018RNF185RING/RNFUpregulatedJWANA(33)
Zhou et al, 2014TRIM59RING/TRIMUpregulatedP53p53(49)
Chi et al, 2009MDM2RING/MDMUpregulatedP53; RUNX3MDM2/ITGB1(80)
Liu et al, 2015CHIPRING/NADownregulatedTRAF2TRAF2/NF-κB(85)
Bai et al, 2011Cullin1RING/SCFUpregulatedp27NA(112)
Li et al, 2016FBXL2RING/SCFDownregulatedFoxM1NA(101)
Cen et al, 2014; Wu et al, 2015FBXL5RING/SCFDownregulatedCortactin; Snail1NA(102,103)
Zou et al, 2018FBXO31RING/SCFDownregulatedSnail1NA(104)
Huang et al, 2018; Kuai et al, 2019; Zhou et al, 2014FBXW7RING/SCFDownregulatedc-Myc; RhoA; Brg1; GFI1RhoA(92-94)
Gao et al, 2013; Wang et al, 2016β-TRCPRING/SCFUpregulatedPHLPP1; FOX3AKT(99,100)
Li et al, 2012COP1RING/CRL4Upregulatedp53NA(125)
Ding et al, 2018pVHLRING/CRL2DownregulatedNEK8NA(119)
Kim et al, 2008NEDD4-1HECT NEDD4UpregulatedPTENAKT(130)
Tao et al, 2017SMURF1HECT NEDD4UpregulatedDAB2IPPI3K/AKT(139)
Zhang et al, 2015WWP1HECT NEDD4UpregulatedPTENAKT(132)
Yang et al, 2016UBR5HECT/OtherUpregulatedGKN1p16/Rb(140)

[i] NA, not applicable.

HECTs, the second largest E3 ligase family in humans, comprises 28 members and can be categorized into three subfamilies: Neural precursor cell expressed developmentally downregulated protein (NEDD)4 family, HECT domain-containing protein (HERC) family and ‘other’ HECT ligases (22). Studies to date have suggested that only the NEDD4 family and a member of the Other HECT ligase families are related to GC. The NEDD4 subfamily is the most characteristic family, including nine members in humans: NEDD4-1 (also known as NEDD4), NEDD4-2, Itchy E3 ubiquitin-protein ligase, SMAD ubiquitin regulatory factor (Smurf)1, Smurf2, WW domain-containing E3 ubiquitin protein ligase (WWP)1, WWP2, NEDD4-like 1 (NEDL1) and NEDL2 (23). The Other HECT ligase family includes 13 members, and they all contain different domains in addition to the HECT domain.

The structure and functional significance of RINGs in GC will be described in the following paragraphs in the order of monomer, dimer and multiple subunit E3 ligases, and the HECTs will be described in the order of the NEDD4 family, HERC family and other HECT ligases.

Structure and function of RING-type E3 ligases in GC

Monomeric RING domain E3 ligase
RING finger (RNF) domain subfamily

The RNF protein family contains a large number of members that are associated with several types of digestive system tumors, such as colorectal cancer and hepatocellular carcinoma (24,25). RNFs also play a vital role in the occurrence of GC. RNF6 encodes a 685-amino-acid protein with a coiled-coil domain at the N-terminus and a RING-H2 finger at the C-terminus (26). RNF38 shares a similar structure with RNF6 (27), and previous studies have shown that RNF6 and RNF38 are overexpressed in GC and regulate GC cell growth. Both RNF6 and RNF38 induce polyubiquitination of tyrosine-protein phosphatase non-receptor type 6 and subsequently enhance STAT3 signaling, which promotes the proliferation of GC cells (Fig. 1) (15,16).

RNF26, which is located in the endoplasmic reticulum, is a polypeptide of 433 amino acids with an N-terminal leucine zipper domain and a C-terminal RING finger domain (28,29). The expression level of RNF26 is upregulated in several types of human cancer cell lines, including HL-60, HeLa S3, SW480 and MKN7 cells (28). As the substrate protein of RNF26, the mediator of interferon regulatory factor 3 can be ubiquitinated and regulate the innate antiviral response (30). However, the functional mechanism of RNF26 in GC has not been fully elucidated.

Similar to RNF6, RNF26 and RNF38, RNF185 also acts as an oncogene in GC, but with distinct subcellular localization and a distinct mechanism of action. RNF185 localizes to the mitochondria, contains a C3HC4-type RING domain and two transmembrane domains (31). PRA1 family protein 3 (JWA) is a multifunctional cytoskeleton-binding protein induced by all-trans retinoic acid. The function of JWA involves enhancing intracellular defenses against H2O2-induced oxidative stress and reducing cell apoptosis (32). RN F185 can downregulate the expression of JWA and promote GC cell migration (Fig. 1) (33). Generally, higher expression of RNF185 is associated with a worse prognosis of GC.

RNF43 and zinc/RING finger 3 (ZNRF3) are homologous proteins that have antagonistic effects in combination with leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5) (34). The Lgr5 protein, a member of the G-protein coupled receptor family of proteins, was identified as a novel gastrointestinal stem cell marker (35). Moreover, Lgr5-positive gastric stem cells are cancer-initiating cells able to drive GC cell self-renewal, which contributes to malignant progression (3537). RNF43 negatively regulates the Wnt/β-catenin pathway by recognizing Lgr5 and markedly downregulating the expression of Lgr5 protein (34). A previous study demonstrated that ZNRF3 acts as a tumor suppressor by downregulating the expression levels of β-catenin and transcription factor 4 protein (38). RNF43/ZNRF3 mediates the ubiquitylation of seven transmembrane domains of frizzled receptors and subsequently inhibits the proliferation of GC cells (39). A few large-scale genomic analyses have reported RNF43 mutations in different cancer types, including GC (40). Whole-genome sequencing revealed that RNF43 is mutated in 4.8% of microsatellite-stable and 54.6% of microsatellite-unstable tumors (41). The mutational landscape of RNF43 may provide a new approach to facilitate genome-guided personalized therapy in GC. Overall, RNF43/ZNRF3 is a tumor suppressor and a potential therapeutic target for GC. Therefore, five members of the RNF subfamily participate in regulating the development of GC, where RNF6, RNF26, RNF38 and RNF185 function as carcinogenic factors, while RNF43/ZNRF3 acts as a tumor suppressor.

Membrane-associated RING-CH (MARCH)8

MARCH8 is a member of the MARCH subfamily. A recent study identified 11 E3 ligases that contained the RING-CH domain and were named the MARCH1-11 subfamily (42). The structure of MARCH8 contains the RING-CH domain and transmembrane domains. JWA is a downstream protein of RNF185 ligase (33); it promotes the ubiquitination of death receptor 4 by increasing the expression of MARCH8 in GC cells, thereby reducing tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis (Fig. 1) (43). Moreover, MARCH8 induces the apoptosis of GC cell lines by inactivating the PI3K and β-catenin/STAT3 signaling pathways (44). In summary, MARCH8 is a tumor suppressor in GC.

Tripartite motif-containing (TRIM) subfamily

TRIM proteins form a subfamily that regulates multiple cellular processes. TRIMs share a common N-terminal tripartite domain, a RING domain, one or two B-boxes and coiled-coil domains (45). To date, three E3 ligases have been shown to be involved in the development and metastasis of GC. They are TRIM28 (also known as KAP1 or TIF1-β), TRIM29 and TRIM59 (4649). TRIM28 is a universal transcriptional corepressor (50). The overexpression of TRIM28 causes peritoneal carcinomatosis and poor prognosis in GC (47). Similar to TRIM28, the expression of TRIM29 is also upregulated in GC; the high expression of TRIM29 mRNA may be an independent predictor of lymph node metastasis and depth of invasion (48). TRIM59 has been reported in several human tumors and acts on diverse signaling pathways, such as the focal adhesion kinase/AKT/matrix metalloproteinases pathway in epithelial ovarian cancer (51), the PI3K/AKT/mTOR pathway in human cholangiocarcinoma (52), the Wnt/β-catenin signaling pathway in neuroblastoma (53), the NF-κB pathway in non-small cell lung cancer (54) and the p53 signaling pathway in GC (49). The p53 signaling pathway can be negatively regulated by the E3 ligase TRIM59, which enhances the ubiquitination and subsequent degradation of p53 (Fig. 2). TRIM59 might promote gastric carcinogenesis through this mechanism (53). In summary, TRIM28, TRIM29 and TRIM59 play oncogenic roles in gastric tumorigenesis, but only the regulatory mechanism of TRIM59 has been elucidated.

RING finger and CHY zinc-finger domain containing 1 (PIRH2)

PIRH2 E3 ligases are crucial negative regulators of p53 (Fig. 2). In addition to p53, many other proteins, such as p63, p73, c-Myc, p27, DNA polymerase Eta and checkpoint kinase 2, can be ubiquitylated by PIRH2 (55). PIRH2 plays an important role in the regulation of many types of tumors, such as glioma, lung cancer and breast cancer (5658). In GC, PIRH2 is a key E3 ligase of p53 ubiquitination, and silencing of PIRH2 causes p53 protein accumulation (59). This study also demonstrates that a microRNA (miR)-100-RNF144B-PIRH2-p53-dependent pathway might be a novel mechanism of ubiquitin-mediated p53 degradation in GC cells (59).

CBL subfamily

CBL proteins contain two highly conserved domains: The N-terminal tyrosine kinase-binding domain and the C3HC4 RING finger domain (60). The mammalian CBL family consists of Cbl, Cbl-b and c-Cbl ligases (61). The three members have similar functions, perhaps due to the specific structure. The c-Cbl protein was the first CBL family protein discovered, followed by Cbl-b and Cbl (62). Previous studies have confirmed that all three CBL proteins are closely associated with GC. In 2000, a study reported that the c-Cbl protein was frequently tyrosine phosphorylated in a tumor-specific manner in human GC tissues (63). Subsequently, c-Cbl was found to be involved in stomach carcinogenesis by connecting with the EGFR system (64).

Cbl-b has a significant impact on the prognosis and drug sensitivity of GC, and a previous study showed that Cbl-b is an oncogene (64). However, subsequent studies provide different perspectives, in which Cbl-b is found to enhance the sensitivity to 5-fluorouracil and cetuximab in GC cells through the ubiquitination pathway (65,66). Other studies have also indicated that Cbl-b inhibits tumor metastasis and growth in multiple drug-resistant (MDR) gastric and breast cancer cells, as well as increasing the sensitivity of MDR cells to anticancer drugs (6769). These findings provide a new research direction for the chemotherapy and targeted therapy of GC.

Cbl, in conjunction with the EGFR system, might be related to gastric carcinogenesis and metastasis (70). In summary, Cbl and c-Cbl are oncogenes in GC, whereas Cbl-b can act as an oncogene or tumor suppressor, and only its regulatory mechanism has been well clarified among the three members.

Checkpoint with forkhead and RING finger domains protein (CHFR)

Previous studies have reported that the aberrant methylation of CHFR promotes the development of GC (71,72). As an E3 ligase, CHFR contains an N-terminal forkhead-associated domain, a central RING domain and a C-terminal cysteine-rich region (73). PARP-1 may be a substrate by CHFR for ubiquitination and degradation in GC; this process leads to cell cycle arrest before entering mitosis and inhibits the proliferation of GC cells (74). Thus, CHFR functions as a tumor suppressor in GC.

Makorin-1 (MKRN1) and FGF-induced in gastric cancer (FIGC)

These two E3 ligases are rarely mentioned in GC. MKRN1 was first identified owing to its interaction with human telomerase reverse transcriptase (TERT) and modulation of telomere length homeostasis (75). The MKRN1-mediated ubiquitination of tumor suppressor ARF (p14ARF) was described in 2011 (76). MKRN1 induces the ubiquitination and degradation of p14ARF and downregulated p14ARF expression (Fig. 2) (76). Furthermore, MKRN1 overexpression was associated with well-differentiated gastric carcinoma, whereas p14ARF overexpression was associated with poorly differentiated gastric carcinoma. FIGC, a novel FGF-induced ubiquitin-protein ligase, consists of an N-terminal RING finger module and proline-rich region at the C-terminus. Only one study has shown that FIGC probably functions as an E3 ligase and is implicated in carcinogenesis through the dysregulation of fibroblast growth modulator (77). In brief, further research is needed to confirm the mechanism of these two E3 ligases in GC.

Dimeric RING domain E3 ligases
MDM2

Dimeric RING domain E3 ligases can be classified into homodimers and heterodimers. MDM2, a heterodimeric RING ligase, was originally identified as a ubiquitin ligase E3 that promotes the degradation of tumor suppressor p53 (Fig. 2) (78). Subsequent studies suggest that MDM2 can ubiquitinate and degrade multiple signaling molecules in GC, including p53, forkhead box protein O3A (FOXO3A) and runt-related transcription factor 3 (RUNX3) (7881). Human hTERT can interact with MDM2 and dramatically increase the ubiquitination of FOXO3A, resulting in the invasion of GC cells (79). RUNX3 is known as a tumor suppressor (82). MDM2 ligases can recognize Lys94 and Lys148 of RUNX3 and decrease the expression levels of RUNX3 (80). Overall, MDM2 acts as a carcinogenic factor in GC by affecting different signaling proteins.

C-terminus of Hsp70-interacting protein (CHIP)

CHIP includes a C-terminal U-box domain and an N-terminal tetratricopeptide repeat domain, which have E3 ubiquitin ligase activity and interact with the molecular chaperones Hsc70-Hsp70 and Hsp90, respectively (83). CHIP is characterized as a homologous dimeric RING ligase and antioncogene in human cancer (15,84). The U-box domain of CHIP facilitates TNF receptor-associated factor 2 ubiquitination for degradation and then inactivates NF-κB signaling (Fig. 1). A previous study also showed that CHIP expression prevents the angiogenesis and metastasis of GC (85). Above all, CHIP overexpression is correlated with good prognosis in GC patients, and targeting CHIP may be a new approach in GC therapy.

Multi-subunit RING domain E3 ligases
SCF subfamily

The CRL1 complex comprises SKP1, CUL1, RING box1 (RBX1) and a member of the F-box protein family (86). The F-box protein family can be further categorized into three subclasses: i) FBXW; ii) F-box and leucine-rich repeat (FBXL); and iii) F-boxes containing other domain motifs (FBXO) proteins. Each subunit of SCF has unique features as follows: i) CUL1 serves as a rigid molecular scaffold protein; ii) RBX1 contains a RING finger domain for the recruitment of E2 enzyme; iii) SKP1 functions as an adaptor; iv) F-box proteins act as a substrate-determining component (86,87), many of them function as E3 ligase and will be discussed in detail below.

FBXW proteins

The WD repeat domain comprises a 44–60 residue sequence that typically contains the GH dipeptide 11–24 residues from its N-terminus and the WD dipeptide at the C-terminus (88). This class of E3 ligases mainly recognizes proteins involved in cell cycle regulation and tumorigenesis, thereby regulating cancerous growth. FBXW7 and β-transducin repeat-containing protein (β-TRCP) are directly correlated with the progression of GC in the form of E3 ligases (89,90).

FBXW7 is a well-characterized SCF in GC and facilitates the destruction of oncogenic proteins, such as c-Myc, transforming protein RhoA (RhoA), transcription activator BRG1 (Brg1) and zinc-finger protein GFI-1 (GFI1) (9093). These targeted proteins all govern gastric tumorigenesis; for example, Brg1 promotes the metastasis of GC (92), RhoA has been implicated in gastric tumorigenesis (Fig. 1) (94), and GFI1 promotes GC cell proliferation as an oncoprotein (93). FBXW7 is also regulated by several upstream signaling molecules. Previous studies indicated that microRNAs and long noncoding RNAs are involved in the occurrence and development of GC by altering the expression of FBXW7 (95,96). Therefore, FBXW7 is a complex tumor suppressor in GC because of its involvement in numerous upstream and downstream signals.

β-TRCP has two distinct isoforms, β-TRCP1 and β-TRCP2, which share similar biochemical properties (97). It was reported that β-TRCP1 and β-TRCP2 were predominantly expressed in the stomach and the small intestine, respectively. A previous study showed that β-TRCP2 might promote gastric carcinogenesis through the activation of the Wnt signaling pathway (98). Moreover, β-TRCPs participate in the regulation of the AKT signaling pathway (Fig. 3) and epidermal growth factor receptor/glycogen synthase kinase-3β/FOXP3 axis through the ubiquitination of PH domain leucine-rich repeat-containing protein phosphatase 1 and FOX3, respectively (99,100). One previous study has reported that β-TRCP can serve as a tumor suppressor or oncoprotein in the etiology of a variety of cancers depending on the type of tumor tissue (99). Nevertheless, β-TRCP might serve as an oncoprotein in GC.

FBXL and FBXO proteins

FBXL proteins contain leucine-rich repeat sequences. FBXL2 and FBXL5 exhibit similar characteristics in GC (101,102). FBXL2 promotes the ubiquitination and degradation of FOXM1, which then inhibits GC proliferation (101). Similarly, FBXL5 can also suppress GC cell migration by the ubiquitination-mediated destruction of Cortactin and Snail1 (102,103). FBXO31, a member of the third class of the F-box protein family, can also target Snail1 for its ubiquitylation and degradation (104). Hence, FBXL2, FBXL5 and FBXO31 exert tumor inhibitory roles in GC.

CUL1

CUL1 is a member of the CUL family and acts as a scaffold protein of the SCF ubiquitin E3 ligase (105). CUL1 is modified by the ubiquitin-like protein NEDD8 and enhances the activity of SCF ligases to p27 (106). Several studies have shown that diverse types of human malignant tumors are related to CUL1. CUL1 can facilitate cell proliferation in osteosarcoma, breast cancer, prostate cancer and lung cancer in vitro and in vivo (107110). In addition, the co-expression of CUL1 and CUL2 induces the initiation of carcinogenesis in colorectal cancer by arresting p53-positive colon cells in the G1 phase of the cell cycle (111). Before these findings, immunohistochemistry results suggested that high expression levels of CUL1 were detected in 60% of all GC tissues, in a study of 792 patients (112). Further in vitro studies showed that increased CUL1 expression was correlated with poor patient survival by decreasing p27 expression (112) (Fig. 2). Therefore, CUL1, an oncoprotein, can be regarded as a prognostic biomarker of GC.

Von Hippel-Lindau disease tumor suppressor (pVHL)

Von Hippel-Lindau disease was first considered to be a heritable cancer syndrome characterized by retinal and neuronal hemangioblastoma owing to a mutation in the VHL gene (113). Although the pVHL protein itself displays no enzymatic activity, pVHL functions as a substrate recognition subunit in CRL2 E3 ligases after binding with the elongin and CUL proteins (114). pVHL is a tumor suppressor in renal cell carcinoma (RCC) (115,116). Serine/threonine-protein kinase NEK8 is a serine/threonine-specific protein kinase family member that serves a role in the progression of mitosis and carcinogenesis (117). NEK8 is reported to be a novel target of pVHL in the regulation of RCC and GC (118,119); pVHL promotes NEK8 protein degradation via the proteasome-ubiquitin pathway in GC cells (119). In summary, the E3 ligase pVHL targets NEK8 and inhibits the proliferation, colony formation and migration of GC (119).

COP1

COP1 protein structure comprises an N-terminal RING finger region, a coiled-coil domain and seven WD40 repeats at its C-terminus (120). COP1 is well studied in plants, but it has been little studied in humans. COP1 is known as a central repressor in the light signaling pathway and forms a complex with suppressor of PHYA-1 (COP1-SPA1) (121). This complex interacts with the CUL4-DDB1 ligase, which belongs to the CRL4 family, to form CUL4-DDB1COP1-SPA1 ligases in plants (122). However, the catalytic mechanism of COP1 has not been fully elucidated in humans. Previous studies revealed that the ubiquitin ligase COP1 promoted the progression of multiple cancer types in vitro, including GC, by downregulating the expression of p53 (Fig. 2) (123125). The role of COP1 in GC is controversial (123126). One previous study indicates that low COP1 expression resulted in poorer prognoses in patients with GCs (126); however, more studies have suggested that COP1 functions as an oncogene (123125).

In summary, there are eight multi-subunit RING domain E3 ligases associated with GC, including six SCFs, one CRL2 ligase and one CRL4 ligase. Among these eight CRLs, β-TRCP, CUL1 and COP1 are oncoproteins, whereas FBXW7, FBXL2, FBXL5, FBXO31 and pVHL act as tumor suppressors.

Structure and function of HECT-type E3 ligases in GC

NEDD4 subfamily

The E3 ligases of the NEDD4 subfamily are characterized by an N-terminal C2 domain responsible for subcellular localization, between two and four WW domains that recruit substrates and a HECT domain at the C-terminus (127). Three NEDD4 subfamily members might be related to GC, including NEDD4-1, WWP1 and Smurf1, and they will be elaborated below.

NEDD4-1

NEDD4-1 contains four WW domains and is an ancestral member of the NEDD4 family (23). Previous studies have indicated that NEDD4-1 is frequently overexpressed in several types of human cancers, including hepatocellular carcinoma, lung cancer and gastrointestinal cancer (128130). A range of tumor suppressors can be ubiquitinated by NEDD4-1, including PTEN, c-Myc and large tumor suppressor kinase 1 (128,131). Immunohistochemical analysis conducted by Kim et al (130) showed that NEDD4-1 is overexpressed in colorectal and gastric carcinomas. NEDD4-1 was also found to promote GC cell migration and invasion (129). As a carcinogenic factor, the targets of NEDD4-1 remain unclear in human GC, and further studies are needed to explore this research topic.

WW domain-containing 1 (WWP1)

WWP1 is another GC-related member of the NEDD4 subfamily that also has four WW domains. Similar to NEDD4-1, WWP1 has been revealed as a versatile E3 ligase with a large repertoire of substrates (23). In GC cell lines, WWP1 is overexpressed, and is closely associated with worse survival by regulating the PTEN-AKT signaling pathway in patients with GC (Fig. 3) (132). The overall survival rate of patients who were positive for WWP1 protein was 25.9%, whereas it was 66.0% in patients who without WWP1 protein in China in 2015 (132). Subsequent studies further confirmed that WWP1 might play a role as an oncogene in GC. miR-584-5p, miR-129-5p and miR-129-3p were found to suppress WWP1 protein expression and inhibit the proliferation of GC in vivo and in vitro (133,134). These findings suggested that WWP1 might be a valuable prognostic marker and potential target in the treatment of GC.

Smurf1

Smurf1 was first recognized in selective interactions with receptor-regulated SMADs, which led to its initial naming (135). Smurf1 contains two WW domains and negatively regulates the transforming growth factor-β/bone morphogenetic protein signaling (136). DAB2-interacting protein (DAB2IP), a tumor suppressor, is known to be downregulated in GC (137); Smurf1 significantly promotes the ubiquitination-dependent degradation of DAB2IP (Fig. 3) (138). In addition, a subsequent study concluded that the Smurf1/DAB2IP signaling axis has an important impact in GC (139). Overall, Smurf1 might act an oncoprotein in GC.

Other HECT ligases
UBR5

UBR5 is the only member of Other HECT ligase family that serves a role in regulating GC cell growth (140). Structurally, UBR5 is composed of an N-terminal ubiquitin-associated domain, two nuclear localization signals, a ubiquitin recognin box domain, a C-terminal poly(A)-binding domain and a HECT domain at its far C-terminus (141). A previous study showed that UBR5 gene mutations occur in 27.8% of GC and 23.3% of colorectal cancer (142). Gastrokine 1, a gastric tumor suppressor, can be downregulated by UBR5 E3 ligase (Fig. 1), and the overexpression of UBR5 is associated with poor overall and disease-free survival (140). Thus, UBR5 may serve as a carcinogenic agent and a prognostic factor in GC.

Therapeutics targeting E3 ligases in GC

E3 ligases have potent effects on the origin, progression and prognosis of GC through a series of signaling pathways. E3 ligase-targeting molecules and drugs may provide a new approach to GC treatment. Bortezomib was the first proteasome inhibitor approved by the US Food and Drug Administration in multiple myeloma (143). In vitro, bortezomib has a significant negative effect on the growth of GC cells (144). It is possible that bortezomib may become a common adjuvant therapeutic target in GC because it has a significant negative effect on the proliferation of GC cells (144). However, only a few E3 ligase-targeting molecules have the ability to suppress the progression of GC.

APG115 has been identified as a novel inhibitor of MDM2 ligase, and its potential for treating GC has been shown in vitro and in vivo (145). In vitro, APG115 inhibited the proliferation of GC cell lines that harbored MDM2 expression by downregulating the mRNA expression of MDM2. In a xenograft mouse model, APG115 contributed to a smaller GC tumor size and enhanced the effect of radiotherapy. As previously mentioned, MDM2 downregulates several tumor suppressors in GC. Consequently, the MDM2 inhibitor APG115 may be applied for GC treatment in the future.

Nutlin proteins were identified in 2004 as the first selective small molecules of MDM2, which could antagonize p53-MDM2 binding (146). Among the nutlins, only nutlin-3 represents a promising therapeutic candidate for drug development in human cancer (147). Over the past decade, it has been confirmed that nutlin-3 can induce cell growth arrest and apoptosis in a number of cancer cell types (148,149). In p53-defective cancer cells, there is a synergistic effect between nutlin-3 and bortezomib; cotreatment with bortezomib and nutlin-3 significantly induce paraptosis and cell death (150). Nutlin-3 has not been used in the treatment of GC; however, the antitumor activity of nutlin-3 against GC cells has been demonstrated in vivo and in vitro (151). It has been reported that nutlin-3 induces G1 arrest in MKN-45 and SNU-1 gastric adenocarcinoma cell lines in vitro, and the activation of p53 by nutlin-3 effectively increased the incidence of apoptosis in wild-type p53 GC cells. In vivo, the combined treatment of nutlin-3 and fluorouracil led to a more potent inhibitory effect on the tumor growth of experimental animals compared with treatment with each agent alone. Overall, nutlin-3 is a broad-spectrum antitumor agent and has the potential to be used in the treatment of GC by targeting the E3 ligase MDM2.

Triptolide is a compound purified from tripterygium wilfordii that exhibits antitumor effects in GC (152155). In 1991, Chinese scholars found that triptolide had antitumor activity in a variety of cancer cell lines, including GC (152). A subsequent study demonstrated that triptolide treatment of GC cells containing wild-type p53 gene resulted in a significant inhibitory effect on cell growth, whereas GC cells with mutant p53 did not exhibit this effect (153). Another study indicated that this p53-dependent antitumor activity was achieved by inhibiting the overexpression of MDM2 (154). Moutan cortex is another Traditional Chinese Medicine that can also induce apoptosis through the MDM2-p53-dependent pathway in GC cells (155). Therefore, Chinese herbs, such as triptolide and moutan cortex, might be potential anticancer agents for GC.

MLN4924, a neddylation inhibitor, is an indirect inhibitor of CRL E3 ligases. MLN4924 acts as a promoter of apoptosis and is a potential anticancer drug in diverse types of human cancers, including GC (156). It has been reported that MLN4924 downregulates the expression of CRLs and then suppresses the growth of GC cells. There are some other small molecules that can also target E3 ligase. miR-223 can target FBXW7 and downregulate its expression, so drugs that promote degradation of miR-223 may be useful in patients with GC (157). As aforementioned, WWP1 is an oncoprotein in GC, and miR-584-5p, miR-129-5p and miR-129-3p suppress WWP1 protein expression and inhibit the proliferation of GC (133,134). Thus, miRNAs may also be a research direction for E3-targeted therapy for GC. In summary, there are still no drugs targeting E3 used in the clinical treatment of GC, and the effects of the compounds mentioned above are still in the research stage.

Conclusions and perspectives

Over the past few years, an increasing number of E3 ligases have been described as tumor regulators in GC. The present review summarizes approximately thirty types of E3 ligases that play essential roles in regulating the development of GC, including RING and HECT ligases. The function and significance of E3 ligases in GC has been well examined, but several E3 ligases, such as COP1, need further studies to elucidate their mechanisms. It has been shown that many synthetic and natural compounds targeting E3 ligases could regulate the level of various signaling proteins through UPS-mediated degradation in human cancers (158). Compounds and small molecules targeting E3 ligases may become underlying templates for the synthesis of targeted therapeutic drugs in GC. However, there are still many obstacles to overcome before the application of compounds targeting E3 ligases in GC, such as the detection and analysis of their complex functional mechanisms and molecular structures. Therefore, further studies should aim to reveal the molecular mechanism of individual E3 ligases in different subtypes of GC, and determine the structure of these targeting compounds to facilitate further synthesis of such targeted therapy drugs. In conclusion, E3 ligases are crucial tumor regulatory factors and potential therapeutic targets in GC.

Acknowledgements

Not applicable.

Funding

This research was funded by The National Natural Science Foundation of China (grant no. 30871207), The National Natural Science Foundation of China (grant nos. 81874063 and 81672389) and Anhui Province Science and Technology Key Project (grant no. 1704a0802176).

Availability of data and materials

Not applicable.

Authors' contributions

MW and YL developed the concept of the manuscript. MW, WD and ZK were responsible for writing, reviewing and editing the manuscript. WD participated in revising the manuscript. ZK supervised the project. YL was involved with the project administration. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

Cbl

casitas B lineage lymphoma

GC

gastric cancer

HECT

homologous to E6-associated protein C-terminus

NEDD

neural precursor cell expressed developmentally downregulated protein

RBR

RING-in-between-RING

RING

really interesting new gene

RNF

RING finger domain

UPS

ubiquitin-proteasome system

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Wu Y, Fan Y, Jiang Y, Wang Y, Liu H and Wei M: Analysis of risk factors associated with precancerous lesion of gastric cancer in patients from eastern China: A comparative study. J Cancer Res Ther. 9:205–209. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Zeng H, Zheng R, Guo Y, Zhang S, Zou X, Wang N, Zhang L, Tang J, Chen J, Wei K, et al: Cancer survival in China, 2003–2005: A population-based study. Int J Cancer. 136:1921–1930. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Woo Y, Goldner B, Son T, Song K, Noh SH, Fong Y and Hyung WJ: Western validation of a novel gastric cancer prognosis prediction model in US gastric cancer patients. J Am Coll Surg. 226:252–258. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Wen T, Wang Z, Li Y, Li Z, Che X, Fan Y, Wang S, Qu J, Yang X, Hou K, et al: A four-factor immunoscore system that predicts clinical outcome for stage II/III gastric cancer. Cancer Immunol Res. 5:524–534. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Abramov IS, Emelyanova MA, Ryabaya OO, Krasnov GS, Zasedatelev AS and Nasedkina TV: Somatic mutations associated with metastasis in acral melanoma. Mol Biol (Mosk). 53:648–653. 2019.(In Russian). View Article : Google Scholar : PubMed/NCBI

7 

Hou YC and Deng JY: Role of E3 ubiquitin ligases in gastric cancer. World J Gastroenterol. 21:786–793. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Hickey CM, Xie Y and Hochstrasser M: DNA binding by the MATα2 transcription factor controls its access to alternative ubiquitin-modification pathways. Mol Biol Cell. 29:542–556. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Bulatov E, Valiullina A, Sayarova R and Rizvanov A: Promising new therapeutic targets for regulation of inflammation and immunity: RING-type E3 ubiquitin ligases. Immunol Lett. 202:44–51. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Lu W, Yang C, He H and Liu H: The CARM1-p300-c-Myc-Max (CPCM) transcriptional complex regulates the expression of CUL4A/4B and affects the stability of CRL4 E3 ligases in colorectal cancer. Int J Biol Sci. 16:1071–1085. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Liu L, Wong CC, Gong B and Yu J: Functional significance and therapeutic implication of ring-type E3 ligases in colorectal cancer. Oncogene. 37:148–159. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Uchida C and Kitagawa M: RING-, HECT-, and RBR-type E3 ubiquitin ligases: Involvement in human cancer. Curr Cancer Drug Targets. 16:157–174. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Johansson H, Isabella Tsai YC, Fantom K, Chung CW, Kümper S, Martino L, Thomas DA, Eberl HC, Muelbaier M, House D and Rittinger K: Fragment-based covalent ligand screening enables rapid discovery of inhibitors for the RBR E3 ubiquitin ligase HOIP. J Am Chem Soc. 141:2703–2712. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Yu JM, Sun W, Wang ZH, Liang X, Hua F, Li K, Lv XX, Zhang XW, Liu YY, Yu JJ, et al: TRIB3 supports breast cancer stemness by suppressing FOXO1 degradation and enhancing SOX2 transcription. Nat Commun. 10:57202019. View Article : Google Scholar : PubMed/NCBI

15 

Zhang J, Wu H, Yi B, Zhou J, Wei L, Chen Y and Zhang L: RING finger protein 38 induces gastric cancer cell growth by decreasing the stability of the protein tyrosine phosphatase SHP-1. FEBS Lett. 592:3092–3100. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Huang Z, Cai Y, Yang C, Chen Z, Sun H, Xu Y, Chen W, Xu D, Tian W and Wang H: Knockdown of RNF6 inhibits gastric cancer cell growth by suppressing STAT3 signaling. Onco Targets Ther. 11:6579–6587. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Berndsen CE and Wolberger C: New insights into ubiquitin E3 ligase mechanism. Nat Struct Mol Biol. 21:301–307. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Ohi MD, Vander Kooi CW, Rosenberg JA, Chazin WJ and Gould KL: Structural insights into the U-box, a domain associated with multi-ubiquitination. Nat Struct Biol. 10:250–255. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Leslie PL, Ke H and Zhang Y: The MDM2 RING domain and central acidic domain play distinct roles in MDM2 protein homodimerization and MDM2-MDMX protein heterodimerization. J Biol Chem. 290:12941–12950. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Genschik P, Sumara I and Lechner E: The emerging family of CULLIN3-RING ubiquitin ligases (CRL3s): Cellular functions and disease implications. EMBO J. 32:2307–2320. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Kelsall IR, Kristariyanto YA, Knebel A, Wood NT, Kulathu Y and Alpi AF: Coupled monoubiquitylation of the co-E3 ligase DCNL1 by Ariadne-RBR E3 ubiquitin ligases promotes cullin-RING ligase complex remodeling. J Biol Chem. 294:2651–2664. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Weber J, Polo S and Maspero E: HECT E3 ligases: A tale with multiple facets. Front Physiol. 10:370–377. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Lorenz S: Structural mechanisms of HECT-type ubiquitin ligases. Biol Chem. 399:127–145. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Geng R, Tan X, Wu J, Pan Z, Yi M, Shi W, Liu R, Yao C, Wang G, Lin J, et al: RNF183 promotes proliferation and metastasis of colorectal cancer cells via activation of NF-κB-IL-8 axis. Cell Death Dis. 8:e29942017. View Article : Google Scholar : PubMed/NCBI

25 

Peng R, Zhang PF, Yang X, Wei CY, Huang XY, Cai JB, Lu JC, Gao C, Sun HX, Gao Q, et al: Overexpression of RNF38 facilitates TGF-β signaling by Ubiquitinating and degrading AHNAK in hepatocellular carcinoma. J Exp Clin Cancer Res. 38:1132019. View Article : Google Scholar : PubMed/NCBI

26 

Macdonald DH, Lahiri D, Sampath A, Chase A, Sohal J and Cross NC: Cloning and characterization of RNF6, a novel RING finger gene mapping to 13q12. Genomics. 58:94–97. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Eisenberg I, Hochner H, Levi T, Yelin R, Kahan T and Mitrani-Rosenbaum S: Cloning and characterization of a novel human gene RNF38 encoding a conserved putative protein with a RING finger domain. Biochem Biophys Res Commun. 294:1169–1176. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Katoh M: Molecular cloning and characterization of RNF26 on human chromosome 11q23 region, encoding a novel RING finger protein with leucine zipper. Biochem Biophys Res Commun. 282:1038–1044. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Jongsma ML, Berlin I, Wijdeven RH, Janssen L, Janssen GM, Garstka MA, Janssen H, Mensink M, van Veelen PA, Spaapen RM and Neefjes J: An ER-associated pathway defines endosomal architecture for controlled cargo transport. Cell. 166:152–166. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Qin Y, Zhou MT, Hu MM, Hu YH, Zhang J, Guo L, Zhong B and Shu HB: RNF26 temporally regulates virus-triggered type I interferon induction by two distinct mechanisms. PLoS Pathog. 10:e10043582014. View Article : Google Scholar : PubMed/NCBI

31 

Wang R, Zhao X, Xu J, Wen Y, Li A, Lu M and Zhou J: Astrocytic JWA deletion exacerbates dopaminergic neurodegeneration by decreasing glutamate transporters in mice. Cell Death Dis. 9:3522018. View Article : Google Scholar : PubMed/NCBI

32 

Zhu T, Chen R, Li A, Liu J, Gu D, Liu Q, C Chang H and Zhou J: JWA as a novel molecule involved in oxidative stress-associated signal pathway in myelogenous leukemia cells. J Toxicol Environ Health A. 69:1399–1411. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Qiu D, Wang Q, Wang Z, Chen J, Yan D, Zhou Y, Li A, Zhang R, Wang S and Zhou J: RNF185 modulates JWA ubiquitination and promotes gastric cancer metastasis. Biochim Biophys Acta Mol Basis Dis. 1864:1552–1561. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Gao Y, Cai A, Xi H, Li J, Xu W, Zhang Y, Zhang K, Cui J, Wu X, Wei B and Chen L: Ring finger protein 43 associates with gastric cancer progression and attenuates the stemness of gastric cancer stem-like cells via the Wnt-β/catenin signaling pathway. Stem Cell Res Ther. 8:982017. View Article : Google Scholar : PubMed/NCBI

35 

Xi HQ, Cai AZ, Wu XS, Cui JX, Shen WS, Bian SB, Wang N, Li JY, Lu CR, Song Z, et al: Leucine-rich repeat-containing G-protein-coupled receptor 5 is associated with invasion, metastasis, and could be a potential therapeutic target in human gastric cancer. Br J Cancer. 110:2011–2020. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Li XB, Yang G, Zhu L, Tang YL, Zhang C, Ju Z, Yang X and Teng Y: Gastric Lgr5(+) stem cells are the cellular origin of invasive intestinal-type gastric cancer in mice. Cell Res. 26:838–849. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Barker N, Huch M, Kujala P, van de Wetering M, Snippert HJ, van Es JH, Sato T, Stange DE, Begthel H, van den Born M, et al: Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 6:25–36. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Zhou Y, Lan J, Wang W, Shi Q, Lan Y, Cheng Z and Guan H: ZNRF3 acts as a tumour suppressor by the Wnt signalling pathway in human gastric adenocarcinoma. J Mol Histol. 44:555–563. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Nanki K, Toshimitsu K, Takano A, Fujii M, Shimokawa M, Ohta Y, Matano M, Seino T, Nishikori S, Ishikawa K, et al: Divergent routes toward Wnt and R-spondin niche independency during human gastric carcinogenesis. Cell. 174:856–869.e17. 2018. View Article : Google Scholar : PubMed/NCBI

40 

Hao HX, Jiang X and Cong F: Control of Wnt receptor turnover by R-spondin-ZNRF3/RNF43 signaling module and its dysregulation in cancer. Cancers (Basel). 8(pii): E542016. View Article : Google Scholar : PubMed/NCBI

41 

Wang K, Yuen ST, Xu J, Lee SP, Yan HH, Shi ST, Siu HC, Deng S, Chu KM, Law S, et al: Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet. 46:573–582. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Liu H, Mintern JD and Villadangos JA: MARCH ligases in immunity. Curr Opin Immunol. 58:38–43. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Wang Q, Chen Q, Zhu L, Chen M, Xu W, Panday S, Wang Z, Li A, Røe OD, Chen R, et al: JWA regulates TRAIL-induced apoptosis via MARCH8-mediated DR4 ubiquitination in cisplatin-resistant gastric cancer cells. Oncogenesis. 6:e3532017. View Article : Google Scholar : PubMed/NCBI

44 

Yin J, Ji Z, Hong Y, Song Z, Hu N, Zhuang M, Bian B, Liu Y and Wu F: Sh-MARCH8 inhibits tumorigenesis via PI3K pathway in gastric cancer. Cell Physiol Biochem. 49:306–321. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Sun M, Li S, Yu K, Xiang J and Li F: An E3 ubiquitin ligase TRIM9 is involved in WSSV infection via interaction with β-TrCP. Dev Comp Immunol. 97:57–63. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Sun Y, Keown JR, Black MM, Raclot C, Demarais N, Trono D, Turelli P and Goldstone DC: A dissection of oligomerization by the TRIM28 tripartite motif and the interaction with members of the Krab-ZFP family. J Mol Biol. 431:2511–2527. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Yokoe T, Toiyama Y, Okugawa Y, Tanaka K, Ohi M, Inoue Y, Mohri Y, Miki C and Kusunoki M: KAP1 is associated with peritoneal carcinomatosis in gastric cancer. Ann Surg Oncol. 17:821–828. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Kosaka Y, Inoue H, Ohmachi T, Yokoe T, Matsumoto T, Mimori K, Tanaka F, Watanabe M and Mori M: Tripartite motif-containing 29 (TRIM29) is a novel marker for lymph node metastasis in gastric cancer. Ann Surg Oncol. 14:2543–2549. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Zhou Z, Ji Z, Wang Y, Li J, Cao H, Zhu HH and Gao WQ: TRIM59 is up-regulated in gastric tumors, promoting ubiquitination and degradation of p53. Gastroenterology. 147:1043–1054. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Ma X, Zhang S, Zhang M, Zhu Y, Ma P, Yang S, Su L, Li Z, Lv W and Luan W: TRIM28 down-regulation on methylation imprints in bovine preimplantation embryos. Zygote. 26:449–456. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Zhang P, Zhang H, Wang Y, Zhang P and Qi Y: Tripartite motif-containing protein 59 (TRIM59) promotes epithelial ovarian cancer progression via the focal adhesion kinase(FAK)/AKT/matrix metalloproteinase (MMP) pathway. Med Sci Monit. 25:3366–3373. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Shen H, Zhang J, Zhang Y, Feng Q, Wang H, Li G, Jiang W and Li X: Knockdown of tripartite motif 59 (TRIM59) inhibits proliferation in cholangiocarcinoma via the PI3K/AKT/mTOR signaling pathway. Gene. 698:50–60. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Chen G, Chen W, Ye M, Tan W and Jia B: TRIM59 knockdown inhibits cell proliferation by down-regulating the Wnt/β-catenin signaling pathway in neuroblastoma. Biosci Rep. 39(pii): BSR201812772019. View Article : Google Scholar : PubMed/NCBI

54 

Cui Z, Liu Z, Zeng J, Chen L, Wu Q, Mo J, Zhang G, Song L, Xu W, Zhang S and Guo X: Eugenol inhibits non-small cell lung cancer by repressing expression of NF-κB-regulated TRIM59. Phytother Res. 33:1562–1569. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Halaby MJ, Hakem R and Hakem A: Pirh2: An E3 ligase with central roles in the regulation of cell cycle, DNA damage response, and differentiation. Cell Cycle. 12:2733–2737. 2013. View Article : Google Scholar : PubMed/NCBI

56 

Bao Y, Wu X, Yuan D, Shi W and Shi J: High expression of Pirh2 is associated with poor prognosis in glioma. Cell Mol Neurobiol. 37:1501–1509. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Daks A, Petukhov A, Fedorova O, Shuvalov O, Merkulov V, Vasileva E, Antonov A and Barlev NA: E3 ubiquitin ligase Pirh2 enhances tumorigenic properties of human non-small cell lung carcinoma cells. Genes Cancer. 7:383–393. 2016.PubMed/NCBI

58 

Yang S, Chen Y, Sun F, Ni Q, Wang H, Huang Y, Zhang C, Liu K, Wang S, Qiu J, et al: Downregulated pirh2 can decrease the proliferation of breast cancer cells. Arch Med Res. 47:186–195. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Yang G, Gong Y, Wang Q, Wang L and Zhang X: miR-100 antagonism triggers apoptosis by inhibiting ubiquitination-mediated p53 degradation. Oncogene. 36:1023–1037. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Eichinger L, Pachebat JA, Glöckner G, Rajandream MA, Sucgang R, Berriman M, Song J, Olsen R, Szafranski K, Xu Q, et al: The genome of the social amoeba Dictyostelium discoideum. Nature. 435:43–57. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Langdon WY, Hyland CD, Grumont RJ and Morse HC III: The c-cbl proto-oncogene is preferentially expressed in thymus and testis tissue and encodes a nuclear protein. J Virol. 63:5420–5424. 1989. View Article : Google Scholar : PubMed/NCBI

62 

Lee H and Tsygankov AY: Cbl-family proteins as regulators of cytoskeleton-dependent phenomena. J Cell Physiol. 228:2285–2293. 2013. View Article : Google Scholar : PubMed/NCBI

63 

Kamei T, Machida K, Nimura Y, Senga T, Yamada I, Yoshii S, Matsuda S and Hamaguchi M: C-Cbl protein in human cancer tissues is frequently tyrosine phosphorylated in a tumor-specific manner. Int J Oncol. 17:335–339. 2000.PubMed/NCBI

64 

Dong Q, Liu YP, Qu XJ, Hou KZ and Li LL: Expression of c-Cbl, Cbl-b, and epidermal growth factor receptor in gastric carcinoma and their clinical significance. Chin J Cancer. 29:59–64. 2010. View Article : Google Scholar : PubMed/NCBI

65 

Feng D, Ma Y, Liu J, Xu L, Zhang Y, Qu J, Liu Y and Qu X: Cbl-b enhances sensitivity to 5-fluorouracil via EGFR- and mitochondria-mediated pathways in gastric cancer cells. Int J Mol Sci. 14:24399–24411. 2013. View Article : Google Scholar : PubMed/NCBI

66 

Yu P, Fan Y, Qu X, Zhang J, Song N, Liu J and Liu Y: Cbl-b regulates the sensitivity of cetuximab through ubiquitin-proteasome system in human gastric cancer cells. J BUON. 21:867–873. 2016.PubMed/NCBI

67 

CHe X, Zhang Y, Qu X, Guo T, Ma Y, Li C, Fan Y, Hou K, Cai Y, Yu R, et al: The E3 ubiquitin ligase Cbl-b inhibits tumor growth in multidrug-resistant gastric and breast cancer cells. Neoplasma. 64:887–892. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Xu L, Zhang Y, Qu X, Che X, Guo T, Cai Y, Li A, Li D, Li C, Wen T, et al: E3 ubiquitin ligase Cbl-b prevents tumor metastasis by maintaining the epithelial phenotype in multiple drug-resistant gastric and breast cancer cells. Neoplasia. 19:374–382. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Zhang Y, Qu X, Hu X, Yang X, Hou K, Teng Y, Zhang J, Sada K and Liu Y: Reversal of P-glycoprotein-mediated multi-drug resistance by the E3 ubiquitin ligase Cbl-b in human gastric adenocarcinoma cells. J Pathol. 218:248–255. 2009. View Article : Google Scholar : PubMed/NCBI

70 

Lai AZ, Durrant M, Zuo D, Ratcliffe CD and Park M: Met kinase-dependent loss of the E3 ligase Cbl in gastric cancer. J Biol Chem. 287:8048–8059. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Gao YJ, Xin Y, Zhang JJ and Zhou J: Mechanism and pathobiologic implications of CHFR promoter methylation in gastric carcinoma. World J Gastroenterol. 14:5000–5007. 2008. View Article : Google Scholar : PubMed/NCBI

72 

Li Y, Yang Y, Lu Y, Herman JG, Brock MV, Zhao P and Guo M: Predictive value of CHFR and MLH1 methylation in human gastric cancer. Gastric Cancer. 18:280–287. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Satoh A, Toyota M, Itoh F, Sasaki Y, Suzuki H, Ogi K, Kikuchi T, Mita H, Yamashita T, Kojima T, et al: Epigenetic inactivation of CHFR and sensitivity to microtubule inhibitors in gastric cancer. Cancer Res. 63:8606–8613. 2003.PubMed/NCBI

74 

Kashima L, Idogawa M, Mita H, Shitashige M, Yamada T, Ogi K, Suzuki H, Toyota M, Ariga H, Sasaki Y and Tokino T: CHFR protein regulates mitotic checkpoint by targeting PARP-1 protein for ubiquitination and degradation. J Biol Chem. 287:12975–12984. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Kim JH, Park SM, Kang MR, Oh SY, Lee TH, Muller MT and Chung IK: Ubiquitin ligase MKRN1 modulates telomere length homeostasis through a proteolysis of hTERT. Genes Dev. 19:776–781. 2005. View Article : Google Scholar : PubMed/NCBI

76 

Ko A, Shin JY, Seo J, Lee KD, Lee EW, Lee MS, Lee HW, Choi IJ, Jeong JS, Chun KH and Song J: Acceleration of gastric tumorigenesis through MKRN1-mediated posttranslational regulation of p14ARF. J Natl Cancer Inst. 104:1660–1672. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Jang JH: FIGC, a novel FGF-induced ubiquitin-protein ligase in gastric cancers FEBS. Lett. 578:21–25. 2004.

78 

Wu CE, Esfandiari A, Ho YH, Wang N, Mahdi AK, Aptullahoglu E, Lovat P and Lunec J: Targeting negative regulation of p53 by MDM2 and WIP1 as a therapeutic strategy in cutaneous melanoma. Br J Cancer. 118:495–508. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Hu C, Ni Z, Li BS, Yong X, Yang X, Zhang JW, Zhang D, Qin Y, Jie MM, Dong H, et al: hTERT promotes the invasion of gastric cancer cells by enhancing FOXO3a ubiquitination and subsequent ITGB1 upregulation. Gut. 66:31–42. 2017. View Article : Google Scholar : PubMed/NCBI

80 

Chi XZ, Kim J, Lee YH, Lee JW, Lee KS, Wee H, Kim WJ, Park WY, Oh BC, Stein GS, et al: Runt-related transcription factor RUNX3 is a target of MDM2-mediated ubiquitination. Cancer Res. 69:8111–8119. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Jung CR, Lim JH, Choi Y, Kim DG, Kang KJ, Noh SM and Im DS: Enigma negatively regulates p53 through MDM2 and promotes tumor cell survival in mice. J Clin Invest. 120:4493–4506. 2010. View Article : Google Scholar : PubMed/NCBI

82 

Feng Y, Gao S, Gao Y, Song D, Wang X and Chen Z: Runx3 expression in rectal cancer cells and its effect on cell invasion and proliferation. Oncol Lett. 18:3290–3294. 2019.PubMed/NCBI

83 

Connell P, Ballinger CA, Jiang J, Wu Y, Thompson LJ, Höhfeld J and Patterson C: The co-chaperone CHIP regulates protein triage decisions mediated by heat-shock proteins. Nat Cell Biol. 3:93–96. 2001. View Article : Google Scholar : PubMed/NCBI

84 

Xiao M, Yan M, Zhang J, Xu Q and Chen W: Carboxy-terminus Hsc70 interacting protein exerts a tumor inhibition function in head and neck cancer. Oncol Rep. 38:1629–1636. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Liu F, Zhou J, Zhou P, Chen W and Guo F: The ubiquitin ligase CHIP inactivates NF-κB signaling and impairs the ability of migration and invasion in gastric cancer cells. Int J Oncol. 46:2096–2106. 2015. View Article : Google Scholar : PubMed/NCBI

86 

Zheng N, Schulman BA, Song L, Miller JJ, Jeffrey PD, Wang P, Chu C, Koepp DM, Elledge SJ, Pagano M, et al: Structure of the Cul1-Rbx1-Skp1-F boxSkp2 SCF ubiquitin ligase complex. Nature. 416:703–709. 2002. View Article : Google Scholar : PubMed/NCBI

87 

Lisztwan J, Marti A, Sutterlüty H, Gstaiger M, Wirbelauer C and Krek W: Association of human CUL-1 and ubiquitin-conjugating enzyme CDC34 with the F-box protein p45(SKP2): Evidence for evolutionary conservation in the subunit composition of the CDC34-SCF pathway. EMBO J. 17:368–383. 1998. View Article : Google Scholar : PubMed/NCBI

88 

Uddin S, Bhat AA, Krishnankutty R, Mir F, Kulinski M and Mohammad RM: Involvement of F-BOX proteins in progression and development of human malignancies. Semin Cancer Biol. 36:18–32. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Jiang ZH, Peng T, Qian HL, Lu CD, Qiu F and Zhang SZ: DNA damage-induced activation of ATM promotes β-TRCP-mediated ARID1A ubiquitination and destruction in gastric cancer cells. Cancer Cell Int. 19:1622019. View Article : Google Scholar : PubMed/NCBI

90 

Milne AN, Leguit R, Corver WE, Morsink FH, Polak M, de Leng WW, Carvalho R and Offerhaus GJ: Loss of CDC4/FBXW7 in gastric carcinoma. Cell Oncol. 32:347–359. 2010.PubMed/NCBI

91 

Li H, Wang Z, Zhang W, Qian K, Xu W and Zhang S: Fbxw7 regulates tumor apoptosis, growth arrest and the epithelial-to-mesenchymal transition in part through the RhoA signaling pathway in gastric cancer. Cancer Lett. 370:39–55. 2016. View Article : Google Scholar : PubMed/NCBI

92 

Huang LY, Zhao J, Chen H, Wan L, Inuzuka H, Guo J, Fu X, Zhai Y, Lu Z, Wang X, et al: SCFFBW7-mediated degradation of Brg1 suppresses gastric cancer metastasis. Nat Commun. 9:35692018. View Article : Google Scholar : PubMed/NCBI

93 

Kuai X, Li L, Chen R, Wang K, Chen M, Cui B, Zhang Y, Li J, Zhu H, Zhou H, et al: SCFFBXW7/GSK3β-Mediated GFI1 Degradation Suppresses Proliferation of Gastric Cancer Cells. Cancer Res. 79:4387–4398. 2019. View Article : Google Scholar : PubMed/NCBI

94 

Zhou J, Hayakawa Y, Wang TC and Bass AJ: RhoA mutations identified in diffuse gastric cancer. Cancer Cell. 26:9–11. 2014. View Article : Google Scholar : PubMed/NCBI

95 

Gong J, Cui Z, Li L, Ma Q, Wang Q, Gao Y and Sun H: MicroRNA-25 promotes gastric cancer proliferation, invasion, and migration by directly targeting F-box and WD-40 domain protein 7, FBXW7. Tumour Biol. 36:7831–7840. 2015. View Article : Google Scholar : PubMed/NCBI

96 

Lv Z, Zhang Y, Yu X, Lin Y and Ge Y: RETRACTED: The function of long non-coding RNA MT1JP in the development and progression of gastric cancer. Pathol Res Pract. 214:1218–1223. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Frescas D and Pagano M: Deregulated proteolysis by the F-box proteins SKP2 and beta-TrCP: Tipping the scales of cancer. Nat Rev Cancer. 8:438–449. 2008. View Article : Google Scholar : PubMed/NCBI

98 

Saitoh T and Katoh M: Expression profiles of betaTRCP1 and betaTRCP2, and mutation analysis of betaTRCP2 in gastric cancer. Int J Oncol. 18:959–964. 2001.PubMed/NCBI

99 

Gao G, Kun T, Sheng Y, Qian M, Kong F, Liu X, Yu Z, Zhang H, Zhang Q, Gu J and Zhang X: SGT1 regulates Akt signaling by promoting beta-TrCP-dependent PHLPP1 degradation in gastric cancer cells. Mol Biol Rep. 40:2947–2953. 2013. View Article : Google Scholar : PubMed/NCBI

100 

Wang S, Zhang Y, Wang Y, Ye P, Li J, Li H, Ding Q and Xia J: Amphiregulin confers regulatory T cell suppressive function and tumor invasion via the EGFR/GSK-3β/Foxp3 axis. J Biol Chem. 291:21085–21095. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Li LQ, Pan D, Chen H, Zhang L and Xie WJ: F-box protein FBXL2 inhibits gastric cancer proliferation by ubiquitin-mediated degradation of forkhead box M1. FEBS Lett. 590:445–452. 2016. View Article : Google Scholar : PubMed/NCBI

102 

Cen G, Ding HH, Liu B and Wu WD: FBXL5 targets cortactin for ubiquitination-mediated destruction to regulate gastric cancer cell migration. Tumour Biol. 35:8633–8638. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Wu W, Ding H, Cao J and Zhang W: FBXL5 inhibits metastasis of gastric cancer through suppressing Snail1. Cell Physiol Biochem. 35:1764–1772. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Zou S, Ma C, Yang F, Xu X, Jia J and Liu Z: FBXO31 Suppresses gastric cancer EMT by targeting Snail1 for proteasomal degradation. Mol Cancer Res. 16:286–295. 2018. View Article : Google Scholar : PubMed/NCBI

105 

Petroski MD and Deshaies RJ: Function and regulation of cullin-RING ubiquitin ligases. Nat Rev Mol Cell Biol. 6:9–20. 2005. View Article : Google Scholar : PubMed/NCBI

106 

Morimoto M, Nishida T, Honda R and Yasuda H: Modification of cullin-1 by ubiquitin-like protein Nedd8 enhances the activity of SCF(skp2) toward p27(kip1). Biochem Biophys Res Commun. 270:1093–1096. 2000. View Article : Google Scholar : PubMed/NCBI

107 

Cheng Q and Yin G: Cullin-1 regulates MG63 cell proliferation and metastasis and is a novel prognostic marker of osteosarcoma. Int J Biol Markers. 32:e202–e209. 2017. View Article : Google Scholar : PubMed/NCBI

108 

Zhou YH, Xia J, Xu WH, Zhu X, Wu XH, Hua D and Xing C: Cullin-1 promotes cell proliferation in human breast cancer and is related to diabetes. Int J Biol Markers. 31:e375–e381. 2016. View Article : Google Scholar : PubMed/NCBI

109 

Jiang H, He D, Xu H, Liu J, Qu L and Tong S: Cullin-1 promotes cell proliferation via cell cycle regulation and is a novel in prostate cancer. Int J Clin Exp Pathol. 8:1575–1583. 2015.PubMed/NCBI

110 

Chen TJ, Gao F, Yang T, Thakur A, Ren H, Li Y, Zhang S, Wang T and Chen MW: CDK-associated Cullin 1 promotes cell proliferation with activation of ERK1/2 in human lung cancer A549 cells. Biochem Biophys Res Commun. 437:108–113. 2013. View Article : Google Scholar : PubMed/NCBI

111 

Michail O, Moris D, Theocharis S and Griniatsos J: Cullin-1 and −2 protein expression in colorectal cancer: Correlation with clinicopathological variables. In Vivo. 32:391–396. 2018.PubMed/NCBI

112 

Bai J, Zhou Y, Chen G, Zeng J, Ding J, Tan Y, Zhou J and Li G: Overexpression of Cullin1 is associated with poor prognosis of patients with gastric cancer. Hum Pathol. 42:375–383. 2011. View Article : Google Scholar : PubMed/NCBI

113 

Clifford SC, Cockman ME, Smallwood AC, Mole DR, Woodward ER, Maxwell PH, Ratcliffe PJ and Maher ER: Contrasting effects on HIF-1alpha regulation by disease-causing pVHL mutations correlate with patterns of tumourigenesis in von Hippel-Lindau disease. Hum Mol Genet. 10:1029–1038. 2001. View Article : Google Scholar : PubMed/NCBI

114 

Yokoe S, Nakagawa T, Kojima Y, Higuchi K and Asahi M: Indomethacin-induced intestinal epithelial cell damage is mediated by pVHL activation through the degradation of collagen I and HIF-1α. Biochem Biophys Res Commun. 468:671–676. 2015. View Article : Google Scholar : PubMed/NCBI

115 

Clifford SC, Astuti D, Hooper L, Maxwell PH, Ratcliffe PJ and Maher ER: The pVHL-associated SCF ubiquitin ligase complex: molecular genetic analysis of elongin B and C, Rbx1 and HIF-1alpha in renal cell carcinoma. Oncogene. 20:5067–5074. 2001. View Article : Google Scholar : PubMed/NCBI

116 

Gao L, Wu GJ, Liu B, Shen MZ, Pan TJ, Yu CG, Wang QH, Ru Y, Liu XP, Niu TS, et al: Up-regulation of pVHL along with down-regulation of HIF-1α by NDRG2 expression attenuates proliferation and invasion in renal cancer cells. PLoS One. 8:e841272013. View Article : Google Scholar : PubMed/NCBI

117 

Bowers AJ and Boylan JF: Nek8, a NIMA family kinase member, is overexpressed in primary human breast tumors. Gene. 328:135–142. 2004. View Article : Google Scholar : PubMed/NCBI

118 

Ding XF, Zhou J, Hu QY, Liu SC and Chen G: The tumor suppressor pVHL down-regulates never-in-mitosis A-related kinase 8 via hypoxia-inducible factors to maintain cilia in human renal cancer cells. J Biol Chem. 290:1389–1394. 2015. View Article : Google Scholar : PubMed/NCBI

119 

Ding XF, Chen J, Zhou J, Chen G and Wu YL: Never-in-mitosis A-related kinase 8, a novel target of von-Hippel-Lindau tumor suppressor protein, promotes gastric cancer cell proliferation. Oncol Lett. 16:5900–5906. 2018.PubMed/NCBI

120 

Bianchi E, Denti S, Catena R, Rossetti G, Polo S, Gasparian S, Putignano S, Rogge L and Pardi R: Characterization of human constitutive photomorphogenesis protein 1, a RING finger ubiquitin ligase that interacts with Jun transcription factors and modulates their transcriptional activity. J Biol Chem. 278:19682–19690. 2003. View Article : Google Scholar : PubMed/NCBI

121 

Zhu D, Maier A, Lee JH, Laubinger S, Saijo Y, Wang H, Qu LJ, Hoecker U and Deng XW: Biochemical characterization of Arabidopsis complexes containing CONSTITUTIVELY PHOTOMORPHOGENIC1 and SUPPRESSOR OF PHYA proteins in light control of plant development. Plant Cell. 20:2307–2323. 2008. View Article : Google Scholar : PubMed/NCBI

122 

Saijo Y, Zhu D, Li J, Rubio V, Zhou Z, Shen Y, Hoecker U, Wang H and Deng XW: Arabidopsis COP1/SPA1 complex and FHY1/FHY3 associate with distinct phosphorylated forms of phytochrome A in balancing light signaling. Mol Cell. 31:607–613. 2008. View Article : Google Scholar : PubMed/NCBI

123 

Ka WH, Cho SK, Chun BN, Byun SY and Ahn JC: The ubiquitin ligase COP1 regulates cell cycle and apoptosis by affecting p53 function in human breast cancer cell lines. Breast Cancer. 25:529–538. 2018. View Article : Google Scholar : PubMed/NCBI

124 

Zou S, Zhu Y, Wang B, Qian F, Zhang X, Wang L, Fu C, Bao H, Xie M, Gao S, et al: The ubiquitin ligase COP1 promotes glioma cell proliferation by preferentially downregulating tumor suppressor p53. Mol Neurobiol. 54:5008–5016. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Li YF, Wang DD, Zhao BW, Wang W, Huang CY, Chen YM, Zheng Y, Keshari RP, Xia JC and Zhou ZW: High level of COP1 expression is associated with poor prognosis in primary gastric cancer. Int J Biol Sci. 8:1168–1177. 2012. View Article : Google Scholar : PubMed/NCBI

126 

Sawada G, Ueo H, Matsumura T, Uchi R, Ishibashi M, Mima K, Kurashige J, Takahashi Y, Akiyoshi S, Sudo T, et al: Loss of COP1 expression determines poor prognosis in patients with gastric cancer. Oncol Rep. 30:1971–1975. 2013. View Article : Google Scholar : PubMed/NCBI

127 

Rotin D and Kumar S: Physiological functions of the HECT family of ubiquitin ligases. Nat Rev Mol Cell Biol. 10:398–409. 2009. View Article : Google Scholar : PubMed/NCBI

128 

Zheng H, Ke X, Li D and Wang Q, Wang J, Liu X, Deng M, Deng X, Xue Y, Zhu Y and Wang Q: NEDD4 promotes cell growth and motility in hepatocellular carcinoma. Cell Cycle. 17:728–738. 2018. View Article : Google Scholar : PubMed/NCBI

129 

Shao G, Wang R, Sun A, Wei J, Peng K, Dai Q, Yang W and Lin Q: The E3 ubiquitin ligase NEDD4 mediates cell migration signaling of EGFR in lung cancer cells. Mol Cancer. 17:242018. View Article : Google Scholar : PubMed/NCBI

130 

Kim SS, Yoo NJ, Jeong EG, Kim MS and Lee SH: Expression of NEDD4-1, a PTEN regulator, in gastric and colorectal carcinomas. APMIS. 116:779–784. 2008. View Article : Google Scholar : PubMed/NCBI

131 

Zou X, Levy-Cohen G and Blank M: Molecular functions of NEDD4 E3 ubiquitin ligases in cancer. Biochim Biophys Acta. 1856:91–106. 2015.PubMed/NCBI

132 

Zhang L, Wu Z, Ma Z, Liu H, Wu Y and Zhang Q: WWP1 as a potential tumor oncogene regulates PTEN-Akt signaling pathway in human gastric carcinoma. Tumour Biol. 36:787–798. 2015. View Article : Google Scholar : PubMed/NCBI

133 

Ma L, Chen X, Li C, Cheng R, Gao Z, Meng X, Sun C, Liang C and Liu Y: miR-129-5p and −3p co-target WWP1 to suppress gastric cancer proliferation and migration. J Cell Biochem. Nov 11–2018.(Epub ahead of print).

134 

Li Q, Li Z, Wei S, Wang W, Chen Z, Zhang L, Chen L, Li B, Sun G, Xu J, et al: Overexpression of miR-584-5p inhibits proliferation and induces apoptosis by targeting WW domain-containing E3 ubiquitin protein ligase 1 in gastric cancer. J Exp Clin Cancer Res. 36:592017. View Article : Google Scholar : PubMed/NCBI

135 

Zhu H, Kavsak P, Abdollah S, Wrana JL and Thomsen GH: A SMAD ubiquitin ligase targets the BMP pathway and affects embryonic pattern formation. Nature. 400:687–693. 1999. View Article : Google Scholar : PubMed/NCBI

136 

Koganti P, Levy-Cohen G and Blank M: Smurfs in protein homeostasis, signaling, and cancer. Front Oncol. 8:2952018. View Article : Google Scholar : PubMed/NCBI

137 

Dote H, Toyooka S, Tsukuda K, Yano M, Ota T, Murakami M, Naito M, Toyota M, Gazdar AF and Shimizu N: Aberrant promoter methylation in human DAB2 interactive protein (hDAB2IP) gene in gastrointestinal tumour. Br J Cancer. 92:1117–1125. 2005. View Article : Google Scholar : PubMed/NCBI

138 

Li X, Dai X, Wan L, Inuzuka H, Sun L and North BJ: Smurf1 regulation of DAB2IP controls cell proliferation and migration. Oncotarget. 7:26057–26069. 2016. View Article : Google Scholar : PubMed/NCBI

139 

Tao Y, Sun C, Zhang T and Song Y: SMURF1 promotes the proliferation, migration and invasion of gastric cancer cells. Oncol Rep. 38:1806–1814. 2017. View Article : Google Scholar : PubMed/NCBI

140 

Yang M, Jiang N, Cao QW, Ma MQ and Sun Q: The E3 ligase UBR5 regulates gastric cancer cell growth by destabilizing the tumor suppressor GKN1. Biochem Biophys Res Commun. 478:1624–1629. 2016. View Article : Google Scholar : PubMed/NCBI

141 

Kozlov G, Nguyen L, Lin T, De Crescenzo G, Park M and Gehring K: Structural basis of ubiquitin recognition by the ubiquitin-associated (UBA) domain of the ubiquitin ligase EDD. J Biol Chem. 282:35787–35795. 2007. View Article : Google Scholar : PubMed/NCBI

142 

Kim MS, Oh JE, Eom HS, Yoo NJ and Lee SH: Mutational analysis of UBR5 gene encoding an E3 ubiquitin ligase in common human cancers. Pathology. 42:93–94. 2010. View Article : Google Scholar : PubMed/NCBI

143 

Richardson PG, Hideshima T and Anderson KC: Bortezomib (PS-341): A novel, first-in-class proteasome inhibitor for the treatment of multiple myeloma and other cancers. Cancer Control. 10:361–369. 2003. View Article : Google Scholar : PubMed/NCBI

144 

Zhang B and Gu Y: Bortezomib inhibits gastric carcinoma HGC-27 cells through the phospho-Jun N-terminal kinase (p-JNK) pathway in vitro. Gene. 559:164–171. 2015. View Article : Google Scholar : PubMed/NCBI

145 

Yi H, Yan X, Luo Q, Yuan L, Li B, Pan W, Zhang L, Chen H, Wang J, Zhang Y, et al: A novel small molecule inhibitor of MDM2-p53 (APG-115) enhances radiosensitivity of gastric adenocarcinoma. J Exp Clin Cancer Res. 37:972018. View Article : Google Scholar : PubMed/NCBI

146 

Vassilev LT, Vu BT, Graves B, Carvajal D, Podlaski F, Filipovic Z, Kong N, Kammlott U, Lukacs C, Klein C, et al: In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 303:844–848. 2004. View Article : Google Scholar : PubMed/NCBI

147 

Impicciatore G, Sancilio S, Miscia S and Di Pietro R: Nutlins and ionizing radiation in cancer therapy. Curr Pharm Des. 16:1427–1442. 2010. View Article : Google Scholar : PubMed/NCBI

148 

Yee-Lin V, Pooi-Fong W and Soo-Beng AK: Nutlin-3, A p53-Mdm2 antagonist for nasopharyngeal carcinoma treatment. Mini Rev Med Chem. 18:173–183. 2018. View Article : Google Scholar : PubMed/NCBI

149 

Meijer A, Kruyt FA, van der Zee AG, Hollema H, Le P, ten Hoor KA, Groothuis GM, Quax WJ, de Vries EG and de Jong S: Nutlin-3 preferentially sensitises wild-type p53-expressing cancer cells to DR5-selective TRAIL over rhTRAIL. Br J Cancer. 109:2685–2695. 2013. View Article : Google Scholar : PubMed/NCBI

150 

Lee DM, Kim IY, Seo MJ, Kwon MR and Choi KS: Nutlin-3 enhances the bortezomib sensitivity of p53-defective cancer cells by inducing paraptosis. Exp Mol Med. 49:e3652017. View Article : Google Scholar : PubMed/NCBI

151 

Endo S, Yamato K, Hirai S, Moriwaki T, Fukuda K, Suzuki H, Abei M, Nakagawa I and Hyodo I: Potent in vitro and in vivo antitumor effects of MDM2 inhibitor nutlin-3 in gastric cancer cells. Cancer Sci. 102:605–613. 2011. View Article : Google Scholar : PubMed/NCBI

152 

Wei YS and Adachi I: Inhibitory effect of triptolide on colony formation of breast and stomach cancer cell lines. Zhongguo Yao Li Xue Bao. 12:406–410. 1991.PubMed/NCBI

153 

Jiang XH, Wong BC, Lin MC, Zhu GH, Kung HF, Jiang SH, Yang D and Lam SK: Functional p53 is required for triptolide-induced apoptosis and AP-1 and nuclear factor-kappaB activation in gastric cancer cells. Oncogene. 20:8009–8018. 2001. View Article : Google Scholar : PubMed/NCBI

154 

Wang BY, Cao J, Chen JW and Liu QY: Triptolide induces apoptosis of gastric cancer cells via inhibiting the overexpression of MDM2. Med Oncol. 31:2702014. View Article : Google Scholar : PubMed/NCBI

155 

Choi HS, Seo HS, Kim JH, Um JY, Shin YC and Ko SG: Ethanol extract of paeonia suffruticosa Andrews (PSE) induced AGS human gastric cancer cell apoptosis via fas-dependent apoptosis and MDM2-p53 pathways. J Biomed Sci. 19:822012. View Article : Google Scholar : PubMed/NCBI

156 

Lan H, Tang Z, Jin H and Sun Y: Neddylation inhibitor MLN4924 suppresses growth and migration of human gastric cancer cells. Sci Rep. 6:242182016. View Article : Google Scholar : PubMed/NCBI

157 

Eto K, Iwatsuki M, Watanabe M, Ishimoto T, Ida S, Imamura Y, Iwagami S, Baba Y, Sakamoto Y, Miyamoto Y, et al: The sensitivity of gastric cancer to trastuzumab is regulated by the miR-223/FBXW7 pathway. Int J Cancer. 136:1537–1545. 2015. View Article : Google Scholar : PubMed/NCBI

158 

Schneekloth JS Jr and Crews CM: Natural product inhibitors of the ubiquitin-proteasome pathway. Curr Drug Targets. 12:1581–1594. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2020
Volume 20 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang M, Dai W, Ke Z and Li Y: Functional roles of E3 ubiquitin ligases in gastric cancer (Review). Oncol Lett 20: 22, 2020
APA
Wang, M., Dai, W., Ke, Z., & Li, Y. (2020). Functional roles of E3 ubiquitin ligases in gastric cancer (Review). Oncology Letters, 20, 22. https://doi.org/10.3892/ol.2020.11883
MLA
Wang, M., Dai, W., Ke, Z., Li, Y."Functional roles of E3 ubiquitin ligases in gastric cancer (Review)". Oncology Letters 20.4 (2020): 22.
Chicago
Wang, M., Dai, W., Ke, Z., Li, Y."Functional roles of E3 ubiquitin ligases in gastric cancer (Review)". Oncology Letters 20, no. 4 (2020): 22. https://doi.org/10.3892/ol.2020.11883