Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2020 Jun 1;10(6):1647–1657.

The multiple roles of deubiquitinases in liver cancer

Xin-You Lv 1, Ting Duan 2,3,4, Jin Li 1
PMCID: PMC7339268  PMID: 32642281

Abstract

Primary liver cancer ranks the second leading cause of death associated with cancer in the world and therefore a major public health challenge. The mortality rates of liver cancer has been increasing during the past decades with the reality that the alternative therapeutic drugs are not available. Although growing numbers of proteins involved in liver cancer progression have been identified, many of these are not suitable drug targets, which hinders the development of new drugs to cure liver cancer. It is in urgent demand that novel therapeutic approaches should be explored. Deubiquitinases (DUBs), specifically removing ubiquitin chains from the target protein, have showed vital roles for protein homeostasis and quality control by rigidly regulating the balance between ubiquitination and deubiquitination in normal physiology. Recent studies have revealed deregulation or dysfunction of DUBs always associates with cancer and other diseases. Targeting certain DUBs, leading to degradation or loss function of the key oncoproteins, including undruggable ones, seems to provide a potential therapy for cancer patients. In liver cancer, numberous of DUBs are demonstrated to participate in hepatocarcinogenesis, metastasis and so on. Depending on the substrates, some DUBs may suppress liver cancers while others promote. In this review, we primarily summarize the roles of DUBs in liver tumors, and illustrate opportunities for the application of targeting DUBs for cancer therapy.

Keywords: Primary liver cancer, deubiquitinases, cancer therapy

Introduction

Protein ubiquitination, one of the most versatile of post-translational modifications, plays an important role in regulation of both proteolytic and non-proteolytic process, such as proteasomal or lysosomal degradation of targeted proteins, influencing protein activity, protein interactions and protein localization [1]. Ubiquitin modification includes the attachment of ubiquitin to target proteins by ubiquitinating enzymes, and the reversible ubiquitin removal, which is mediated by deubiquitinating enzymes (DUBs) [2,3].

Ubiquitin is a highly conserved 76-amino acid polypeptide, and it is universally distributed among eukaryotes. The ubiquitin-protein conjugation system constitutes by cascades of steps including activating ubiquitin by the ubiquitin-activation enzyme (E1), then transfer the activated ubiquitin to the target proteins on a lysine residue by ubiquitin-conjugating enzymes (E2) and ubiquitin ligases (E3) [4,5]. Sometimes, only one single ubiquitin is attached to one or multiple lysines (mono-ubiquitination), whereas in others, ubiquitin chains (poly-ubiquitination) is added to target proteins with each ubiquitin attached to the prior (Figure 1). Depending on the lysine for inter-ubiquitin linkages, ubiquitin chains are divided into several different types. As reported, lysine 6, (K6), K11, K27, K29, K33, K48, and K63 are used for chain formation. Among these, the best studyed is K63- and K48-linked ubiquitination. Polyubiquition chains linked by K48, and also K27, K6, K33, K11, and K29 mainly function during proteasomal degradation [6].

Figure 1.

Figure 1

Ubiquitin ligases and deubiquitinases in the ubiquitination proteasomal system. Ubiquitin is firstly activated by the ubiquitin-activating enzyme (E1), followed by its transfer to a lysine residue on target proteins by ubiquitin-conjugating enzymes (E2) and ubiquitin ligases (E3). DUBs reverse this process by cleaving monoubiquitin or polyubiquitin chains from substrates, and therefore prevent proteins from degradation.

Ubiquitination of targeted proteins is reversed by deubiquitinating enzymes (DUBs), a superfamily of metalloproteases and cysteine proteases that catalytically cleave ubiquitin-protein bonds [7]. Up to date, more than one hundred human DUBs have been identified. Based on sequence and conserved domain, they can be classified into six families including ubiquitin-specific proteases (USPs), ovarian tumor proteases (OTUs), ubiquitin carboxy-terminal hydrolases (UCHs), Machado-Joseph disease protein domain proteases (MJDs), JAMM/MPN domain-associated metallopeptidases (JAMMs), and the monocyte chemotactic protein-induced proteases family (MINDYs) [8-11]. USPs, UCHs, OTUs, MJDs and MINDYs are cysteine peptidases, while JAMMs are zinc metallopeptidases [12].

DUBs regulate proteasomal or lysosomal degradation, proteins localization and recycling, which is crucial for protein homeostasis and normal functioning of the cell. Growing evidence indicates dysfunction of DUBs leads to amounts of genetic and sporadic diseases. For example, BRCA1-associated protein 1 (BAP1), one of the UCH family member, is mutated in mesothelioma, melanoma and renal cell carcinoma [13]; USP6 was reported to be translocated in aneurysmal bone cysts [14]; USP7 has shown mutation in neurological disorders [15]; USP8 is found to mutate in Cushing disease [16,17]; USP9X cause developmental disorders with mutation [18] and shows dysregulated expression in cancer [19]. CYLD, commonly mutated in cylindromatosis [20]; and USP15 is indicative of amplification in certain glioblastoma, ovarian and breast cancers [21]. In addition, expansion of DNA ‘CAG’ trinucleotide repeats in ataxin 3 (ATXN3) leads to Machado-Joseph disease (also called spinocerebellar ataxia 3) [22]. Moreover, mutations in the JAMM family member result in microcephaly-capillary malformation syndrome [23]. More and more interest has been focusing on exploiting the regulation of DUBs on vital proteins and pathways involved in cancers. In this paper, we mainly focus on the role of DUBs in liver cancer.

DUBs involved in human liver cancer

Plenty of DUBs have displayed tumour-suppressing or tumor-promoting functions, and therefore may represent potential drug targets for treatment [24]. DUBs related to liver cancer progression are primarily discussed below.

CYLD

CYLD, a K63 linkage-specific deubiquitinase, is one unique member of the USP family, whose zinc finger domain responsible for distal ubiquitin interaction was deleted. The cylindromatosis gene (CYLD) was reported to suppress tumor, whose mutation was found in familial cylindromatosis (also known as Brooke-Spiegler syndrome), an autosomal-dominant predisposition to various tumors of the skin appendages [25].

CYLD is an essential modifier of NF-κB signaling and the ubiquitination state of the NF-κB-activating molecule NEMO [26]. During liver tumorigenesis, CYLD function as tumour suppressor. By conditional knockout of CYLD in parenchymal liver cells, Bergkamen et al. reported the mutant CYLD in CYLDxAlbCre animals resulted in a chronic inflammatory response characterized by prominent ductular reaction and biliary fibrosis. NF-κB signaling was demonstrated to increase in livers of CYLD (FF) xAlbCre mice and may contribute to the fibrotic and inflammatory response. CYLD-mutant form did not contribute to spontaneous hepatocellular carcinomas (HCC), but showed a significantly increased sensitivity to liver cancers induced by the chemical carcinogen diethylnitrosamine (DEN), which proved to be associated with sustained c-Jun N-terminal kinase 1 (JNK1)-mediated signaling by ubiquitination of TNF receptor-associated factor 2 and expression of c-MYC [27,28]. Through regulation the level of hepatocyte growth factor, CYLD alleviated liver damage and hepatocellular fibrogenesis [29]. Once expressing a deubiquitinase-deficient form of CYLD, which has similar oncogenic mutations in humans, it leads to spontaneous hepatic fibrosis and liver tumours by activation of c-Jun N-terminal kinase (JNK) and TGF-β activated kinase 1 (TAK1) [30,31]. Overexpression of CYLD in hepatocytes inhibits both inflammation and fibrosis in mice with nonalcoholic steatohepatitis, whose progression lead to liver cancer finally [32].

In addition, CYLD is suggested to involve in the apoptosis resistance of hepatocellular carcinoma cells by enhancing NF-κB activity [33]. Therefore, CYLD plays key roles in liver inflammation and cancer. It represents a promising therapeutic target for liver cancer.

UCHs

UCHs have recently drawn much attention because of their diverse functions in cell biology [34,35]. Until now, four members of UCHs have been identified including UCH-L1/PGP9.5 (protein gene product 9.5), UCH-L3, UCHL5/UCH37, and BRCA1-associated protein-1 (BAP1) [36-39]. The biological effects of the four UCH enzymes are complicated, as these proteins play quite different roles in different tumor progression.

UCHL1

UCHL1 has been studied extensively. Yu et al. showed hypermethylation of UCHL1 promoter CpG downregulated or silenced its expression in liver cancer, which provided tumor suppression evidence of UCHL1. Previous study has demonstrated that UCHL1 can directly interact with p53 and stabilize p53 through its hydrolase activity, and then decrease MDM2 by its E3 ligase activity. Subsequently, UCHL1 increases the p21 expression in HCC cells, resulting in G2/M phase arrest and hence suppressing proliferation. Moreover, re-expression of UCHL1 activates caspase-9 and induces PARP cleavage, leading to cell apoptosis [40].

Recently, the methylation level of UCHL1 was observed higher in tumor tissues compared with adjacent normal tissues. Cholangiocarcinoma patients with low methylation of UCHL1 survived longer overall, indicating the methylation level of UCHL1 may represent a potential biomarker for Cholangiocarcinoma prediction [41]. Yang et al. reported upregulation of UCHL1 gene promoted apoptosis in hepatocellular carcinoma cells treated with adriamycin and verapamil, while UCHL1 knockdown by siRNA weakened the effect, indicating UCHL1 was involved in the reversal effect of verapamil on Adriamycin-resistant hepatocellular carcinoma cells by promoting apoptosis [42].

UCH37

UCH37 has unique isopeptidase activity of the 19S proteasome complex, which is special for the UCH members. As one subunit of the 19S regulatory particle, hRpn13 interacted with UCH37 via KEKE motifs in the C-terminal regions, then UCH37 is recruited and activated to show deubiquitination activity [43-47]. Fang et al. firstly observed higher UCH37 expression in liver cancer tissues than adjacent para-cancerous tissues, which indicated UCH37 could be a predictor of recurrence after radical resection in HCC patients. Moreover, UCH37 promoted migration and invasion of HCC cells by deubiquitination of PRP19 (essential RNA splicing factor) [48]. By a functional proteomic analysis and screening, glucose-regulated protein 78 was identified as UCH37-interacting protein in HCC, but how the interaction regulates HCC progression remains further investigation [49].

BAP1

It has been found that BAP1 associates with multi-protein complexes that regulate key cellular pathways, including the cell differentiation, cell death, cell cycle, and the DNA damage response (DDR) [50]. Recently, Mosbeh et al. suggested that BAP1 may regulate the pathogenesis of biliary and pancreatic cancers, a subset of hepatocellular carcinoma. High frequency of BAP1 loss in intrahepatic cholangicarcinoma was identified, while the frequency was lower in hepatocellular carcinoma and extrahepatic biliary cancer. For HCC tumors with decrease or loss of BAP1, expression of bile duct (cytokeratin 7) and hepatocytic (HepPar1) markers were higher than those with preserved BAP1 [51]. These studies all indicate BAP1 may be a novel therapy target.

USP9X

USP9X, which belongs to the X-linked USP family, can remove mono-ubiquitin and a number of ubiquitin chains, such as K29, K48 and K63 linkages [52-56]. It has been found to interact with more than 35 proteins. By regulating the protein claspin during S phase [57], USP9X maintains DNA damage checkpoint responses and DNA replication fork stability. Long noncoding RNA LNC473 enhanced HCC cell proliferation, invasion and epithelial-mesenchymal transition process by recruiting USP9X to survivin, which lead to increased survivin expression, a result of ubiquitination inhibition [58]. Overexpression of miR-26b inhibited the endogenous expression level of USP9X protein, leading to the epithelial-mesenchymal transition inhibition of hepatocytes [59]. Above studies indicate USP9X may be a crutial factor involved in epithelial-mesenchymal transition process of HCC. Also, USP9X palys roles in cell death. Zhang et al. reported that glycogen synthase kinase-3β (GSK-3β), a multifunctional kinase, suppressed hydrogen peroxide (H2O2)-induced cell death in HepG2 cells through inhibition of USP9X, and subsequently ubiquitination and proteasome-dependent degradation of ASK1 [60]. HCC cells with p53 expression showed enhanced response to combined treatment with WP1130, inhibitor of USP9X, and doxorubicin compared with p53-deficient cells. Mechanistically, USP9X inhibition promoted ubiquitin-proteasome dependent degradation of p53 [61]. Taken together, USP9X represents another potential therapeutic target.

USP7

USP7, also known as HAUSP, is an evolutionarily conserved protein that was first identified as a molecular partner of the herpes simplex virus protein, Vmw110 [62]. USP7 is required for cell growth, development and stress [63]. In human HCC tissues, the expression of USP7 is higher than in matched peritumoral tissues. Ectopic expression of USP7 promotes HCC cells growth both in vitro and in vivo. Mechanistically, USP7 overexpression stabilizes thyroid hormone receptor-interacting protein 12 (TRIP12) by deubiquitination, thus constitutively inactivating the tumor suppressor p14 (ARF) [64]. Zhu et al. identified that in HCC cells, USP7 protein level was inhibited by microRNA-205 (miR-205), thereby impairing the p53 signaling pathway and facilitating cell proliferation [65]. Moreover, by facilitating the interaction of USP7 with p53, Abraxas brother 1 suppresses HCC cell proliferation and tumour formation [66]. Base on above, USP7 may play vital role during HCC development, which needs further evidence.

A20

A20, also known as TNFAIP3 (Tumor necrosis factor α-induced protein 3), is originally discoved as a primary gene product after tumor necrosis factor α (TNFα) treatment in human umbilical vein endothelial cells [67]. A20 hydrolyzes K48, K11 and K63 polyubiquitin, while it displays enhanced activity towards K63-polyubiquitinated substrates in cells [68-71]. It has been shown that A20 is an important regulator of cellular inflammation signaling [72]. Over the past few years, growing evidence suggests that A20 also plays a functional role in cancer development. Catrysse et al. showed the mice lacking A20 specifically in liver parenchymal cells spontaneously develop chronic liver inflammation, hepatocyte apoptosis and lethality following treatment with sublethal doses of TNF. Besides, these mice are more susceptive to hepatocellular carcinoma development induced by chemical or high fat-diet [73]. Another study revealed silence of A20 accompanied with IFN-γ exposure obviously repressed cell viability, and induced cell-cycle arrest and apoptosis in HCC cells through restraining phosphoinositide 3-kinase/Akt pathway and antiapoptotic B-cell lymphoma 2 proteins [74].

In addition, A20 plays a role in HCC metastasis. In 89 tissue samples from HCC patients, the expression of A20 was decreased in invasive cells compared with the noninvasive cells. Overexpression of A20 significantly inhibited the proliferation, epithelial-mesenchymal transition and migration of HCC cells both in vitro and in vivo [75,76].

Interestingly, Chen et al. found patients with higher A20 expression had a prolonged overall survival and disease-free survival than those with lower A20 expression [76]. Meanwhile, a recent study showed in HCC and liver cirrhosis patients, A20 mRNA level in peripheral blood mononuclear cells was higher than patients with chronic hepatitis B, who showed significantly higher A20 mRNA level than the healthy. For HCC patients with vascular invasion, liver cirrhosis and ascites, A20 mRNA level was also elevated compared with those without [77]. Therefore, A20 might represent a potential biomarker to differentiate the stages of HCC and evaluate prognosis.

Other DUBs involved in liver cancer

There are growing DUBs reported to promote HCC progression recently. By unbiased siRNA screening, Kim et al. found that YOD1 enhances the stability of ITCH, an E3 ligase of LATS, and facilitates LATS1/2 ubiquitination and degradation, subsequently increasing YAP/TAZ levels. Overexpression of YOD1 enhances the hepatocytes proliferation and leads to hepatomegaly [78]. It has been shown that the expression of USP14 in tumor tissues of HCC patients was much higher than para-carcinoma and normal liver tissues. Knockdown or inhibiton of USP14 with b-AP15, the potent and selective inhibitor of USP14, in human hepatocarcinoma SMMC7721 cells both significantly suppressed cell proliferation and induced apoptosis [79,80]. USP4 was up-regulated in mesenchymal-type liver-tumor cells, facilitating proliferation and migration [81]. Downregulation of USP39 significantly enhanced apoptosis, inhibited cell growth in HCC cells, and reduced xenograft tumor growth in nude mice [82,83]. By directly targeting and inhibiting USP28, miR-363-3p destabilizes Myc and prevented hepatocellular tumorigenesis [84]. The EEF1A2/PI3K/AKT/mTOR axis has been suggested to promote the protumorigenic stabilization of the protooncogene MDM4 in human HCC by way of USP2a and AKT-mediated phosphorylation [85].

However, other DUBs have emerged as tumor suppressor. For example, USP16 was frequently downregulated in human HCCs, and the reduced expression of USP16 was correlated with poor differentiation status. In tumour cells, Inhibition of USP16 promoted stem-like properties, ectopic expression of USP16 significantly decreased cell viability and tumour growth [86]. DUBs also regulate drug resistance, which has been a challenge during cancer treament. USP22 promotes the multidrug resistance in HCC cells by activating the SIRT1/AKT/MRP1 pathway via direct interaction with SIRT1 and upregulation of SIRT1 protein [87]. In chemoresistant HCC cells, silencing USP22 dramatically suppressed tumorigenic and metastatic capacities in vivo, as well as inhibited proliferation and epithelial-mesenchymal transition in vitro [88].

DUBs may represent valuable biomarker for HCC patients in near future. HCC samples with higher OTUB1 and USP11 expression have shorter overall survival time [89,90]. HCC patients with Cezanne reduction, in which cancer cells showed high invasiveness, had shorter time to recurrence and poor overall survival [91]. HCC patients with high expression of USP22 showed poor prognosis, reduction of USP22 suppressed cell growth [92]. Tumor metabolism is the key event for unconstrained proliferation of tumor cells. DUBs may impact this process to alter HCC progression. USP2a inhibited fatty acid synthase ubiquitination and subsequently promoted lipogenesis and HCC development [93].

Development of DUBs inhibitors for cancer therapy

Bortezomib, the proteasome inhibitor, has been approved by FDA for multiple myeloma treatment, which makes the ubiquitin-proteasome system as a promising target for new anticancer treatment development [94]. Considering the key roles of DUBs in the ubiquitin-proteasome system and diverse functions in cancer, growing interest has been focused on exploiting DUBs as therapeutic targets [95]. However, the development of selective DUBs inhibitors has been hampered by lack of sufficient knowledge about DUBs biology, difficulties in establishing suitable biochemical methods for compound screening, limitations in models of in vitro and in vivo to measure DUBs activity. Over the past few years, the progress in DUBs drug discovery has accelerated with many issues overcome. Although no DUBs inhibitors have successfully entered clinical trials yet, endeavor is being made to develop them as treatment strategies.

Currently, both pan-DUBs inhibitors and specific inhibitors of single DUB have been identified. The first DUBs active-site inhibitors were cyclopentenone prostaglandins, which induce accumulation of polyubiquitylated proteins and cause p53-dependent apoptosis in colon cancer cells [96,97]. P5091, the inhibitor of USP7, lead multiple myeloma cells to apoptosis [98]. WP1130, which inhibits USP5, USP9X, USP14, and UCH37, was found to trigger rapid polyubiquitinated proteins accumulation in aggresomes and induced apoptosis of tumor cell [99].

In brief, the development of DUBs inhibitors are in the early stages and numerous researches have proved selective inhibitors of cancer-promoting DUBs, such as USP7, efficiently induced cancer cell death. More efforts need to be made for improving the specificity, efficacy, and safety. There is no doubt DUBs inhibitors are emerging as attractive druggable targets and new agents for the treatment of cancer.

Conclusions and perspectives

Liver cancer is predicted to be the fourth leading cause of cancer death worldwide in 2018 and therefore a major public health challenge. Because of rapid progression and lack of targeted drugs, the survival rate of liver cancer is extremely low. Recently, growing evidence has demonstrated the vital role of DUBs in liver cancer progression, which may represent novel targets for cancer therapy. With dramatic advances in DUB screening technologies and biochemical assays, increasing numbers of DUB inhibitors have been developed. Such inhibitors provide the basis for drug-like molecules suitable for clinical evaluation and also provide versatile tools to further investigate DUB cell biology.

Although much progress has been made in exploring the roles of DUBs in liver cancer progression, why these DUBs function differently in the same disease needs further investigation. And we know little about what function the same one DUB has through the pathological process, including liver fibrosis, liver cirrhosis and final hepatocellular carcinoma. In addition, knowledge about whether DUBs affect the tumor microenvironment during liver cancer progression is deficient. Also, specificity and selectivity of DUBs inhibitors needs improvement. Summary, we will witness rapid expansion in the arenas of DUB biology and drug discovery in the next few years.

Acknowledgements

We thank Zikai Zhou for helpful discussion and advice. The research is supported by China Postdoctoral Science Foundation (2018M642468).

Disclosure of conflict of interest

None.

References

  • 1.Swatek KN, Komander D. Ubiquitin modifications. Cell Res. 2016;26:399–422. doi: 10.1038/cr.2016.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Hershko A, Ciechanover A. The ubiquitin system. Annu Rev Biochem. 1998;67:425–479. doi: 10.1146/annurev.biochem.67.1.425. [DOI] [PubMed] [Google Scholar]
  • 3.Komander D, Rape M. The ubiquitin code. Annu Rev Biochem. 2012;81:203–229. doi: 10.1146/annurev-biochem-060310-170328. [DOI] [PubMed] [Google Scholar]
  • 4.Hershko A, Heller H, Elias S, Ciechanover A. Components of ubiquitin-protein ligase system. Resolution, affinity purification, and role in protein breakdown. J Biol Chem. 1983;258:8206–8214. [PubMed] [Google Scholar]
  • 5.Pickart CM. Mechanisms underlying ubiquitination. Annu Rev Biochem. 2001;70:503–533. doi: 10.1146/annurev.biochem.70.1.503. [DOI] [PubMed] [Google Scholar]
  • 6.Dammer EB, Na CH, Xu P, Seyfried NT, Duong DM, Cheng D, Gearing M, Rees H, Lah JJ, Levey AI, Rush J, Peng J. Polyubiquitin linkage profiles in three models of proteolytic stress suggest the etiology of Alzheimer disease. J Biol Chem. 2011;286:10457–10465. doi: 10.1074/jbc.M110.149633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wilkinson KD. Regulation of ubiquitin-dependent processes by deubiquitinating enzymes. FASEB J. 1997;11:1245–1256. doi: 10.1096/fasebj.11.14.9409543. [DOI] [PubMed] [Google Scholar]
  • 8.Abdul RS, Kristariyanto YA, Choi SY, Nkosi PJ, Weidlich S, Labib K, Hofmann K, Kulathu Y. MINDY-1 is a member of an evolutionarily conserved and structurally distinct new family of deubiquitinating enzymes. Mol Cell. 2016;63:146–155. doi: 10.1016/j.molcel.2016.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Harrigan JA, Jacq X, Martin NM, Jackson SP. Deubiquitylating enzymes and drug discovery: emerging opportunities. Nat Rev Drug Discov. 2018;17:57–78. doi: 10.1038/nrd.2017.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.He M, Zhou Z, Wu G, Chen Q, Wan Y. Emerging role of DUBs in tumor metastasis and apoptosis: therapeutic implication. Pharmacol Ther. 2017;177:96–107. doi: 10.1016/j.pharmthera.2017.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ramakrishna S, Suresh B, Baek KH. Biological functions of hyaluronan and cytokine-inducible deubiquitinating enzymes. Biochim Biophys Acta. 2015;1855:83–91. doi: 10.1016/j.bbcan.2014.11.006. [DOI] [PubMed] [Google Scholar]
  • 12.Ronau JA, Beckmann JF, Hochstrasser M. Substrate specificity of the ubiquitin and Ubl proteases. Cell Res. 2016;26:441–456. doi: 10.1038/cr.2016.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Murali R, Wiesner T, Scolyer RA. Tumours associated with BAP1 mutations. Pathology. 2013;45:116–126. doi: 10.1097/PAT.0b013e32835d0efb. [DOI] [PubMed] [Google Scholar]
  • 14.Oliveira AM, Chou MM. USP6-induced neoplasms: the biologic spectrum of aneurysmal bone cyst and nodular fasciitis. Hum Pathol. 2014;45:1–11. doi: 10.1016/j.humpath.2013.03.005. [DOI] [PubMed] [Google Scholar]
  • 15.Hao YH, Fountain MJ, Fon TK, Xia F, Bi W, Kang SH, Patel A, Rosenfeld JA, Le Caignec C, Isidor B, Krantz ID, Noon SE, Pfotenhauer JP, Morgan TM, Moran R, Pedersen RC, Saenz MS, Schaaf CP, Potts PR. USP7 acts as a molecular rheostat to promote WASH-dependent endosomal protein recycling and is mutated in a human neurodevelopmental disorder. Mol Cell. 2015;59:956–969. doi: 10.1016/j.molcel.2015.07.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ma ZY, Song ZJ, Chen JH, Wang YF, Li SQ, Zhou LF, Mao Y, Li YM, Hu RG, Zhang ZY, Ye HY, Shen M, Shou XF, Li ZQ, Peng H, Wang QZ, Zhou DZ, Qin XL, Ji J, Zheng J, Chen H, Wang Y, Geng DY, Tang WJ, Fu CW, Shi ZF, Zhang YC, Ye Z, He WQ, Zhang QL, Tang QS, Xie R, Shen JW, Wen ZJ, Zhou J, Wang T, Huang S, Qiu HJ, Qiao ND, Zhang Y, Pan L, Bao WM, Liu YC, Huang CX, Shi YY, Zhao Y. Recurrent gain-of-function USP8 mutations in Cushing’s disease. Cell Res. 2015;25:306–317. doi: 10.1038/cr.2015.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Reincke M, Sbiera S, Hayakawa A, Theodoropoulou M, Osswald A, Beuschlein F, Meitinger T, Mizuno-Yamasaki E, Kawaguchi K, Saeki Y, Tanaka K, Wieland T, Graf E, Saeger W, Ronchi CL, Allolio B, Buchfelder M, Strom TM, Fassnacht M, Komada M. Mutations in the deubiquitinase gene USP8 cause Cushing’s disease. Nat Genet. 2015;47:31–38. doi: 10.1038/ng.3166. [DOI] [PubMed] [Google Scholar]
  • 18.Homan CC, Kumar R, Nguyen LS, Haan E, Raymond FL, Abidi F, Raynaud M, Schwartz CE, Wood SA, Gecz J, Jolly LA. Mutations in USP9X are associated with X-linked intellectual disability and disrupt neuronal cell migration and growth. Am J Hum Genet. 2014;94:470–478. doi: 10.1016/j.ajhg.2014.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Murtaza M, Jolly LA, Gecz J, Wood SA. La FAM fatale: USP9X in development and disease. Cell Mol Life Sci. 2015;72:2075–2089. doi: 10.1007/s00018-015-1851-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bignell GR, Warren W, Seal S, Takahashi M, Rapley E, Barfoot R, Green H, Brown C, Biggs PJ, Lakhani SR, Jones C, Hansen J, Blair E, Hofmann B, Siebert R, Turner G, Evans DG, Schrander-Stumpel C, Beemer FA, van Den Ouweland A, Halley D, Delpech B, Cleveland MG, Leigh I, Leisti J, Rasmussen S. Identification of the familial cylindromatosis tumour-suppressor gene. Nat Genet. 2000;25:160–165. doi: 10.1038/76006. [DOI] [PubMed] [Google Scholar]
  • 21.Eichhorn PJ, Rodon L, Gonzalez-Junca A, Dirac A, Gili M, Martinez-Saez E, Aura C, Barba I, Peg V, Prat A, Cuartas I, Jimenez J, Garcia-Dorado D, Sahuquillo J, Bernards R, Baselga J, Seoane J. USP15 stabilizes TGF-beta receptor I and promotes oncogenesis through the activation of TGF-beta signaling in glioblastoma. Nat Med. 2012;18:429–435. doi: 10.1038/nm.2619. [DOI] [PubMed] [Google Scholar]
  • 22.Kawaguchi Y, Okamoto T, Taniwaki M, Aizawa M, Inoue M, Katayama S, Kawakami H, Nakamura S, Nishimura M, Akiguchi I, Et A. CAG expansions in a novel gene for Machado-Joseph disease at chromosome 14q32.1. Nat Genet. 1994;8:221–228. doi: 10.1038/ng1194-221. [DOI] [PubMed] [Google Scholar]
  • 23.Mcdonell LM, Mirzaa GM, Alcantara D, Schwartzentruber J, Carter MT, Lee LJ, Clericuzio CL, Graham JJ, Morris-Rosendahl DJ, Polster T, Acsadi G, Townshend S, Williams S, Halbert A, Isidor B, David A, Smyser CD, Paciorkowski AR, Willing M, Woulfe J, Das S, Beaulieu CL, Marcadier J, Geraghty MT, Frey BJ, Majewski J, Bulman DE, Dobyns WB, O’Driscoll M, Boycott KM. Mutations in STAMBP, encoding a deubiquitinating enzyme, cause microcephaly-capillary malformation syndrome. Nat Genet. 2013;45:556–562. doi: 10.1038/ng.2602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Clague MJ, Barsukov I, Coulson JM, Liu H, Rigden DJ, Urbe S. Deubiquitylases from genes to organism. Physiol Rev. 2013;93:1289–1315. doi: 10.1152/physrev.00002.2013. [DOI] [PubMed] [Google Scholar]
  • 25.Bignell GR, Warren W, Seal S, Takahashi M, Rapley E, Barfoot R, Green H, Brown C, Biggs PJ, Lakhani SR, Jones C, Hansen J, Blair E, Hofmann B, Siebert R, Turner G, Evans DG, Schrander-Stumpel C, Beemer FA, van Den Ouweland A, Halley D, Delpech B, Cleveland MG, Leigh I, Leisti J, Rasmussen S. Identification of the familial cylindromatosis tumour-suppressor gene. Nat Genet. 2000;25:160–165. doi: 10.1038/76006. [DOI] [PubMed] [Google Scholar]
  • 26.Kovalenko A, Chable-Bessia C, Cantarella G, Israel A, Wallach D, Courtois G. The tumour suppressor CYLD negatively regulates NF-kappaB signalling by deubiquitination. Nature. 2003;424:801–805. doi: 10.1038/nature01802. [DOI] [PubMed] [Google Scholar]
  • 27.Urbanik T, Boger RJ, Longerich T, Becker K, Ehrenberg KR, Hovelmeyer N, Hahn M, Schuchmann M, Jager D, Waisman A, Worns MA, Schulze-Bergkamen H. Liver specific deletion of CYLDexon7/8 induces severe biliary damage, fibrosis and increases hepatocarcinogenesis in mice. J Hepatol. 2012;57:995–1003. doi: 10.1016/j.jhep.2012.06.017. [DOI] [PubMed] [Google Scholar]
  • 28.Pannem RR, Dorn C, Ahlqvist K, Bosserhoff AK, Hellerbrand C, Massoumi R. CYLD controls c-MYC expression through the JNK-dependent signaling pathway in hepatocellular carcinoma. Carcinogenesis. 2014;35:461–468. doi: 10.1093/carcin/bgt335. [DOI] [PubMed] [Google Scholar]
  • 29.Pannem RR, Dorn C, Hellerbrand C, Massoumi R. Cylindromatosis gene CYLD regulates hepatocyte growth factor expression in hepatic stellate cells through interaction with histone deacetylase 7. Hepatology. 2014;60:1066–1081. doi: 10.1002/hep.27209. [DOI] [PubMed] [Google Scholar]
  • 30.Nikolaou K, Tsagaratou A, Eftychi C, Kollias G, Mosialos G, Talianidis I. Inactivation of the deubiquitinase CYLD in hepatocytes causes apoptosis, inflammation, fibrosis, and cancer. Cancer Cell. 2012;21:738–750. doi: 10.1016/j.ccr.2012.04.026. [DOI] [PubMed] [Google Scholar]
  • 31.Trompouki E, Tsagaratou A, Kosmidis SK, Dolle P, Qian J, Kontoyiannis DL, Cardoso WV, Mosialos G. Truncation of the catalytic domain of the cylindromatosis tumor suppressor impairs lung maturation. Neoplasia. 2009;11:469–476. doi: 10.1593/neo.81424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ji YX, Huang Z, Yang X, Wang X, Zhao LP, Wang PX, Zhang XJ, Alves-Bezerra M, Cai L, Zhang P, Lu YX, Bai L, Gao MM, Zhao H, Tian S, Wang Y, Huang ZX, Zhu XY, Zhang Y, Gong J, She ZG, Li F, Cohen DE, Li H. The deubiquitinating enzyme cylindromatosis mitigates nonalcoholic steatohepatitis. Nat Med. 2018;24:213–223. doi: 10.1038/nm.4461. [DOI] [PubMed] [Google Scholar]
  • 33.Urbanik T, Kohler BC, Boger RJ, Worns MA, Heeger S, Otto G, Hovelmeyer N, Galle PR, Schuchmann M, Waisman A, Schulze-Bergkamen H. Down-regulation of CYLD as a trigger for NF-kappaB activation and a mechanism of apoptotic resistance in hepatocellular carcinoma cells. Int J Oncol. 2011;38:121–131. [PubMed] [Google Scholar]
  • 34.Nijman SM, Luna-Vargas MP, Velds A, Brummelkamp TR, Dirac AM, Sixma TK, Bernards R. A genomic and functional inventory of deubiquitinating enzymes. Cell. 2005;123:773–786. doi: 10.1016/j.cell.2005.11.007. [DOI] [PubMed] [Google Scholar]
  • 35.Sowa ME, Bennett EJ, Gygi SP, Harper JW. Defining the human deubiquitinating enzyme interaction landscape. Cell. 2009;138:389–403. doi: 10.1016/j.cell.2009.04.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lam YA, Xu W, Demartino GN, Cohen RE. Editing of ubiquitin conjugates by an isopeptidase in the 26S proteasome. Nature. 1997;385:737–740. doi: 10.1038/385737a0. [DOI] [PubMed] [Google Scholar]
  • 37.Larsen CN, Krantz BA, Wilkinson KD. Substrate specificity of deubiquitinating enzymes: ubiquitin C-terminal hydrolases. Biochemistry. 1998;37:3358–3368. doi: 10.1021/bi972274d. [DOI] [PubMed] [Google Scholar]
  • 38.Doran JF, Jackson P, Kynoch PA, Thompson RJ. Isolation of PGP 9.5, a new human neurone-specific protein detected by high-resolution two-dimensional electrophoresis. J Neurochem. 1983;40:1542–1547. doi: 10.1111/j.1471-4159.1983.tb08124.x. [DOI] [PubMed] [Google Scholar]
  • 39.Jensen DE, Proctor M, Marquis ST, Gardner HP, Ha SI, Chodosh LA, Ishov AM, Tommerup N, Vissing H, Sekido Y, Minna J, Borodovsky A, Schultz DC, Wilkinson KD, Maul GG, Barlev N, Berger SL, Prendergast GC, Rauscher FR. BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene. 1998;16:1097–1112. doi: 10.1038/sj.onc.1201861. [DOI] [PubMed] [Google Scholar]
  • 40.Yu J, Tao Q, Cheung KF, Jin H, Poon FF, Wang X, Li H, Cheng YY, Rocken C, Ebert MP, Chan AT, Sung JJ. Epigenetic identification of ubiquitin carboxyl-terminal hydrolase L1 as a functional tumor suppressor and biomarker for hepatocellular carcinoma and other digestive tumors. Hepatology. 2008;48:508–518. doi: 10.1002/hep.22343. [DOI] [PubMed] [Google Scholar]
  • 41.Nanok C, Jearanaikoon P, Proungvitaya S, Limpaiboon T. Aberrant methylation of HTATIP2 and UCHL1 as a predictive biomarker for cholangiocarcinoma. Mol Med Rep. 2018;17:4145–4153. doi: 10.3892/mmr.2017.8319. [DOI] [PubMed] [Google Scholar]
  • 42.Yang G, Fan G, Zhang T, Ma K, Huang J, Liu M, Teng X, Xu K, Fan P, Cheng D. Upregulation of ubiquitin carboxyl-terminal hydrolase L1 (UCHL1) mediates the reversal effect of verapamil on chemo-resistance to adriamycin of hepatocellular carcinoma. Med Sci Monit. 2018;24:2072–2082. doi: 10.12659/MSM.908925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Husnjak K, Elsasser S, Zhang N, Chen X, Randles L, Shi Y, Hofmann K, Walters KJ, Finley D, Dikic I. Proteasome subunit Rpn13 is a novel ubiquitin receptor. Nature. 2008;453:481–488. doi: 10.1038/nature06926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Schreiner P, Chen X, Husnjak K, Randles L, Zhang N, Elsasser S, Finley D, Dikic I, Walters KJ, Groll M. Ubiquitin docking at the proteasome through a novel pleckstrin-homology domain interaction. Nature. 2008;453:548–552. doi: 10.1038/nature06924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Jiao L, Ouyang S, Shaw N, Song G, Feng Y, Niu F, Qiu W, Zhu H, Hung LW, Zuo X, Eleonora SV, Zhu P, Dong YH, Xu R, Liu ZJ. Mechanism of the Rpn13-induced activation of Uch37. Protein Cell. 2014;5:616–630. doi: 10.1007/s13238-014-0046-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fang Y, Fu D, Shen XZ. The potential role of ubiquitin c-terminal hydrolases in oncogenesis. Biochim Biophys Acta. 2010;1806:1–6. doi: 10.1016/j.bbcan.2010.03.001. [DOI] [PubMed] [Google Scholar]
  • 47.Randles L, Anchoori RK, Roden RB, Walters KJ. The proteasome ubiquitin receptor hRpn13 and its interacting deubiquitinating enzyme uch37 are required for proper cell cycle progression. J Biol Chem. 2016;291:8773–8783. doi: 10.1074/jbc.M115.694588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fang Y, Fu D, Tang W, Cai Y, Ma D, Wang H, Xue R, Liu T, Huang X, Dong L, Wu H, Shen X. Ubiquitin C-terminal Hydrolase 37, a novel predictor for hepatocellular carcinoma recurrence, promotes cell migration and invasion via interacting and deubiquitinating PRP19. Biochim Biophys Acta. 2013;1833:559–572. doi: 10.1016/j.bbamcr.2012.11.020. [DOI] [PubMed] [Google Scholar]
  • 49.Fang Y, Mu J, Ma Y, Ma D, Fu D, Shen X. The interaction between ubiquitin C-terminal hydrolase 37 and glucose-regulated protein 78 in hepatocellular carcinoma. Mol Cell Biochem. 2012;359:59–66. doi: 10.1007/s11010-011-0999-7. [DOI] [PubMed] [Google Scholar]
  • 50.Carbone M, Yang H, Pass HI, Krausz T, Testa JR, Gaudino G. BAP1 and cancer. Nat Rev Cancer. 2013;13:153–159. doi: 10.1038/nrc3459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mosbeh A, Halfawy K, Abdel-Mageed WS, Sweed D, Rahman M. Nuclear BAP1 loss is common in intrahepatic cholangiocarcinoma and a subtype of hepatocellular carcinoma but rare in pancreatic ductal adenocarcinoma. Cancer Genet. 2018;224-225:21–28. doi: 10.1016/j.cancergen.2018.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Al-Hakim AK, Zagorska A, Chapman L, Deak M, Peggie M, Alessi DR. Control of AMPK-related kinases by USP9X and atypical Lys(29)/Lys(33)-linked polyubiquitin chains. Biochem J. 2008;411:249–260. doi: 10.1042/BJ20080067. [DOI] [PubMed] [Google Scholar]
  • 53.Dupont S, Mamidi A, Cordenonsi M, Montagner M, Zacchigna L, Adorno M, Martello G, Stinchfield MJ, Soligo S, Morsut L, Inui M, Moro S, Modena N, Argenton F, Newfeld SJ, Piccolo S. FAM/USP9x, a deubiquitinating enzyme essential for TGFbeta signaling, controls Smad4 monoubiquitination. Cell. 2009;136:123–135. doi: 10.1016/j.cell.2008.10.051. [DOI] [PubMed] [Google Scholar]
  • 54.Marx C, Held JM, Gibson BW, Benz CC. ErbB2 trafficking and degradation associated with K48 and K63 polyubiquitination. Cancer Res. 2010;70:3709–3717. doi: 10.1158/0008-5472.CAN-09-3768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mouchantaf R, Azakir BA, Mcpherson PS, Millard SM, Wood SA, Angers A. The ubiquitin ligase itch is auto-ubiquitylated in vivo and in vitro but is protected from degradation by interacting with the deubiquitylating enzyme FAM/USP9X. J Biol Chem. 2006;281:38738–38747. doi: 10.1074/jbc.M605959200. [DOI] [PubMed] [Google Scholar]
  • 56.Vong QP, Cao K, Li HY, Iglesias PA, Zheng Y. Chromosome alignment and segregation regulated by ubiquitination of survivin. Science. 2005;310:1499–1504. doi: 10.1126/science.1120160. [DOI] [PubMed] [Google Scholar]
  • 57.Mcgarry E, Gaboriau D, Rainey MD, Restuccia U, Bachi A, Santocanale C. The deubiquitinase USP9X maintains DNA replication fork stability and DNA damage checkpoint responses by regulating CLASPIN during S-phase. Cancer Res. 2016;76:2384–2393. doi: 10.1158/0008-5472.CAN-15-2890. [DOI] [PubMed] [Google Scholar]
  • 58.Chen H, Yang F, Li X, Gong ZJ, Wang LW. Long noncoding RNA LNC473 inhibits the ubiquitination of survivin via association with USP9X and enhances cell proliferation and invasion in hepatocellular carcinoma cells. Biochem Biophys Res Commun. 2018;499:702–710. doi: 10.1016/j.bbrc.2018.03.215. [DOI] [PubMed] [Google Scholar]
  • 59.Shen G, Lin Y, Yang X, Zhang J, Xu Z, Jia H. MicroRNA-26b inhibits epithelial-mesenchymal transition in hepatocellular carcinoma by targeting USP9X. BMC Cancer. 2014;14:393. doi: 10.1186/1471-2407-14-393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhang N, Liu L, Dou Y, Song D, Deng H. Glycogen synthase kinase-3beta antagonizes ROS-induced hepatocellular carcinoma cell death through suppression of the apoptosis signal-regulating kinase 1. Med Oncol. 2016;33:60. doi: 10.1007/s12032-016-0776-2. [DOI] [PubMed] [Google Scholar]
  • 61.Liu H, Chen W, Liang C, Chen BW, Zhi X, Zhang S, Zheng X, Bai X, Liang T. WP1130 increases doxorubicin sensitivity in hepatocellular carcinoma cells through usp9x-dependent p53 degradation. Cancer Lett. 2015;361:218–225. doi: 10.1016/j.canlet.2015.03.001. [DOI] [PubMed] [Google Scholar]
  • 62.Everett RD, Meredith M, Orr A, Cross A, Kathoria M, Parkinson J. A novel ubiquitin-specific protease is dynamically associated with the PML nuclear domain and binds to a herpesvirus regulatory protein. EMBO J. 1997;16:566–577. doi: 10.1093/emboj/16.3.566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Meulmeester E, Maurice MM, Boutell C, Teunisse AF, Ovaa H, Abraham TE, Dirks RW, Jochemsen AG. Loss of HAUSP-mediated deubiquitination contributes to DNA damage-induced destabilization of Hdmx and Hdm2. Mol Cell. 2005;18:565–576. doi: 10.1016/j.molcel.2005.04.024. [DOI] [PubMed] [Google Scholar]
  • 64.Cai JB, Shi GM, Dong ZR, Ke AW, Ma HH, Gao Q, Shen ZZ, Huang XY, Chen H, Yu DD, Liu LX, Zhang PF, Zhang C, Hu MY, Yang LX, Shi YH, Wang XY, Ding ZB, Qiu SJ, Sun HC, Zhou J, Shi YG, Fan J. Ubiquitin-specific protease 7 accelerates p14 (ARF) degradation by deubiquitinating thyroid hormone receptor-interacting protein 12 and promotes hepatocellular carcinoma progression. Hepatology. 2015;61:1603–1614. doi: 10.1002/hep.27682. [DOI] [PubMed] [Google Scholar]
  • 65.Zhu L, Liu R, Zhang W, Qian S, Wang JH. MicroRNA-205 regulates ubiquitin specific peptidase 7 protein expression in hepatocellular carcinoma cells. Mol Med Rep. 2015;12:4652–4656. doi: 10.3892/mmr.2015.3998. [DOI] [PubMed] [Google Scholar]
  • 66.Zhang J, Cao M, Dong J, Li C, Xu W, Zhan Y, Wang X, Yu M, Ge C, Ge Z, Yang X. ABRO1 suppresses tumourigenesis and regulates the DNA damage response by stabilizing p53. Nat Commun. 2014;5:5059. doi: 10.1038/ncomms6059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dixit VM, Green S, Sarma V, Holzman LB, Wolf FW, O’Rourke K, Ward PA, Prochownik EV, Marks RM. Tumor necrosis factor-alpha induction of novel gene products in human endothelial cells including a macrophage-specific chemotaxin. J Biol Chem. 1990;265:2973–2978. [PubMed] [Google Scholar]
  • 68.Evans PC, Ovaa H, Hamon M, Kilshaw PJ, Hamm S, Bauer S, Ploegh HL, Smith TS. Zinc-finger protein A20, a regulator of inflammation and cell survival, has de-ubiquitinating activity. Biochem J. 2004;378:727–734. doi: 10.1042/BJ20031377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Komander D, Barford D. Structure of the A20 OTU domain and mechanistic insights into deubiquitination. Biochem J. 2008;409:77–85. doi: 10.1042/BJ20071399. [DOI] [PubMed] [Google Scholar]
  • 70.Boone DL, Turer EE, Lee EG, Ahmad RC, Wheeler MT, Tsui C, Hurley P, Chien M, Chai S, Hitotsumatsu O, Mcnally E, Pickart C, Ma A. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol. 2004;5:1052–1060. doi: 10.1038/ni1110. [DOI] [PubMed] [Google Scholar]
  • 71.Wertz IE, O’Rourke KM, Zhou H, Eby M, Aravind L, Seshagiri S, Wu P, Wiesmann C, Baker R, Boone DL, Ma A, Koonin EV, Dixit VM. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature. 2004;430:694–699. doi: 10.1038/nature02794. [DOI] [PubMed] [Google Scholar]
  • 72.Hymowitz SG, Wertz IE. A20: from ubiquitin editing to tumour suppression. Nat Rev Cancer. 2010;10:332–341. doi: 10.1038/nrc2775. [DOI] [PubMed] [Google Scholar]
  • 73.Catrysse L, Farhang GM, Vereecke L, Youssef SA, Mc GC, Sze M, Weber A, Heikenwalder M, de Bruin A, Beyaert R, van Loo G. A20 prevents chronic liver inflammation and cancer by protecting hepatocytes from death. Cell Death Dis. 2016;7:e2250. doi: 10.1038/cddis.2016.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Yin L, Fang Z, Shen NJ, Qiu YH, Li AJ, Zhang YJ. Downregulation of A20 increases the cytotoxicity of IFN-gamma in hepatocellular carcinoma cells. Drug Des Devel Ther. 2017;11:2841–2850. doi: 10.2147/DDDT.S135993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wang X, Ma C, Zong Z, Xiao Y, Li N, Guo C, Zhang L, Shi Y. A20 inhibits the motility of HCC cells induced by TNF-alpha. Oncotarget. 2016;7:14742–14754. doi: 10.18632/oncotarget.7521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chen H, Hu L, Luo Z, Zhang J, Zhang C, Qiu B, Dong L, Tan Y, Ding J, Tang S, Shen F, Li Z, Wang H. A20 suppresses hepatocellular carcinoma proliferation and metastasis through inhibition of Twist1 expression. Mol Cancer. 2015;14:186. doi: 10.1186/s12943-015-0454-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fan YC, Zhang YY, Sun YY, Wang N, Xiao XY, Wang K. Altered expression of A20 gene in peripheral blood mononuclear cells is associated with the progression of chronic hepatitis B virus infection. Oncotarget. 2016;7:68821–68832. doi: 10.18632/oncotarget.11993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kim Y, Kim W, Song Y, Kim JR, Cho K, Moon H, Ro SW, Seo E, Ryu YM, Myung SJ, Jho EH. Deubiquitinase YOD1 potentiates YAP/TAZ activities through enhancing ITCH stability. Proc Natl Acad Sci U S A. 2017;114:4691–4696. doi: 10.1073/pnas.1620306114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Huang G, Li L, Zhou W. USP14 activation promotes tumor progression in hepatocellular carcinoma. Oncol Rep. 2015;34:2917–2924. doi: 10.3892/or.2015.4296. [DOI] [PubMed] [Google Scholar]
  • 80.Ding Y, Chen X, Wang B, Yu B, Ge J. Deubiquitinase inhibitor b-AP15 activates endoplasmic reticulum (ER) stress and inhibits Wnt/Notch1 signaling pathway leading to the reduction of cell survival in hepatocellular carcinoma cells. Eur J Pharmacol. 2018;825:10–18. doi: 10.1016/j.ejphar.2018.02.020. [DOI] [PubMed] [Google Scholar]
  • 81.Heo MJ, Kim YM, Koo JH, Yang YM, An J, Lee SK, Lee SJ, Kim KM, Park JW, Kim SG. microRNA-148a dysregulation discriminates poor prognosis of hepatocellular carcinoma in association with USP4 overexpression. Oncotarget. 2014;5:2792–2806. doi: 10.18632/oncotarget.1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Pan Z, Pan H, Zhang J, Yang Y, Liu H, Yang Y, Huang G, Ni J, Huang J, Zhou W. Lentivirus mediated silencing of ubiquitin specific peptidase 39 inhibits cell proliferation of human hepatocellular carcinoma cells in vitro. Biol Res. 2015;48:18. doi: 10.1186/s40659-015-0006-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Yuan X, Sun X, Shi X, Jiang C, Yu D, Zhang W, Guan W, Zhou J, Wu Y, Qiu Y, Ding Y. USP39 promotes the growth of human hepatocellular carcinoma in vitro and in vivo. Oncol Rep. 2015;34:823–832. doi: 10.3892/or.2015.4065. [DOI] [PubMed] [Google Scholar]
  • 84.Han H, Sun D, Li W, Shen H, Zhu Y, Li C, Chen Y, Lu L, Li W, Zhang J, Tian Y, Li Y. A c-Myc-MicroRNA functional feedback loop affects hepatocarcinogenesis. Hepatology. 2013;57:2378–2389. doi: 10.1002/hep.26302. [DOI] [PubMed] [Google Scholar]
  • 85.Pellegrino R, Calvisi DF, Neumann O, Kolluru V, Wesely J, Chen X, Wang C, Wuestefeld T, Ladu S, Elgohary N, Bermejo JL, Radlwimmer B, Zornig M, Zender L, Dombrowski F, Evert M, Schirmacher P, Longerich T. EEF1A2 inactivates p53 by way of PI3K/AKT/mTOR-dependent stabilization of MDM4 in hepatocellular carcinoma. Hepatology. 2014;59:1886–1899. doi: 10.1002/hep.26954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Qian Y, Wang B, Ma A, Zhang L, Xu G, Ding Q, Jing T, Wu L, Liu Y, Yang Z, Liu Y. USP16 Downregulation by carboxyl-terminal truncated HBx promotes the growth of hepatocellular carcinoma cells. Sci Rep. 2016;6:33039. doi: 10.1038/srep33039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ling S, Li J, Shan Q, Dai H, Lu D, Wen X, Song P, Xie H, Zhou L, Liu J, Xu X, Zheng S. USP22 mediates the multidrug resistance of hepatocellular carcinoma via the SIRT1/AKT/MRP1 signaling pathway. Mol Oncol. 2017;11:682–695. doi: 10.1002/1878-0261.12067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhang J, Luo N, Tian Y, Li J, Yang X, Yin H, Xiao C, Sheng J, Li Y, Tang B, Li R. USP22 knockdown enhanced chemosensitivity of hepatocellular carcinoma cells to 5-Fu by up-regulation of Smad4 and suppression of Akt. Oncotarget. 2017;8:24728–24740. doi: 10.18632/oncotarget.15798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ni Q, Chen J, Li X, Xu X, Zhang N, Zhou A, Zhou B, Lu Q, Chen Z. Expression of OTUB1 in hepatocellular carcinoma and its effects on HCC cell migration and invasion. Acta Biochim Biophys Sin. 2017;49:680–688. doi: 10.1093/abbs/gmx056. [DOI] [PubMed] [Google Scholar]
  • 90.Zhang S, Xie C, Li H, Zhang K, Li J, Wang X, Yin Z. Ubiquitin-specific protease 11 serves as a marker of poor prognosis and promotes metastasis in hepatocellular carcinoma. Lab Invest. 2018;98:883–894. doi: 10.1038/s41374-018-0050-7. [DOI] [PubMed] [Google Scholar]
  • 91.Wang JH, Wei W, Guo ZX, Shi M, Guo RP. Decreased Cezanne expression is associated with the progression and poor prognosis in hepatocellular carcinoma. J Transl Med. 2015;13:41. doi: 10.1186/s12967-015-0396-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Tang B, Liang X, Tang F, Zhang J, Zeng S, Jin S, Zhou L, Kudo Y, Qi G. Expression of USP22 and Survivin is an indicator of malignant behavior in hepatocellular carcinoma. Int J Oncol. 2015;47:2208–2216. doi: 10.3892/ijo.2015.3214. [DOI] [PubMed] [Google Scholar]
  • 93.Calvisi DF, Wang C, Ho C, Ladu S, Lee SA, Mattu S, Destefanis G, Delogu S, Zimmermann A, Ericsson J, Brozzetti S, Staniscia T, Chen X, Dombrowski F, Evert M. Increased lipogenesis, induced by AKT-mTORC1-RPS6 signaling, promotes development of human hepatocellular carcinoma. Gastroenterology. 2011;140:1071–1083. doi: 10.1053/j.gastro.2010.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Manasanch EE, Orlowski RZ. Proteasome inhibitors in cancer therapy. Nat Rev Clin Oncol. 2017;14:417–433. doi: 10.1038/nrclinonc.2016.206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Cohen P, Tcherpakov M. Will the ubiquitin system furnish as many drug targets as protein kinases? Cell. 2010;143:686–693. doi: 10.1016/j.cell.2010.11.016. [DOI] [PubMed] [Google Scholar]
  • 96.Mullally JE, Fitzpatrick FA. Pharmacophore model for novel inhibitors of ubiquitin isopeptidases that induce p53-independent cell death. Mol Pharmacol. 2002;62:351–358. doi: 10.1124/mol.62.2.351. [DOI] [PubMed] [Google Scholar]
  • 97.Mullally JE, Moos PJ, Edes K, Fitzpatrick FA. Cyclopentenone prostaglandins of the J series inhibit the ubiquitin isopeptidase activity of the proteasome pathway. J Biol Chem. 2001;276:30366–30373. doi: 10.1074/jbc.M102198200. [DOI] [PubMed] [Google Scholar]
  • 98.Chauhan D, Tian Z, Nicholson B, Kumar KG, Zhou B, Carrasco R, Mcdermott JL, Leach CA, Fulcinniti M, Kodrasov MP, Weinstock J, Kingsbury WD, Hideshima T, Shah PK, Minvielle S, Altun M, Kessler BM, Orlowski R, Richardson P, Munshi N, Anderson KC. A small molecule inhibitor of ubiquitin-specific protease-7 induces apoptosis in multiple myeloma cells and overcomes bortezomib resistance. Cancer Cell. 2012;22:345–358. doi: 10.1016/j.ccr.2012.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kapuria V, Peterson LF, Fang D, Bornmann WG, Talpaz M, Donato NJ. Deubiquitinase inhibition by small-molecule WP1130 triggers aggresome formation and tumor cell apoptosis. Cancer Res. 2010;70:9265–9276. doi: 10.1158/0008-5472.CAN-10-1530. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES