Open access peer-reviewed chapter

Oncogenic Secretory Clusterin: A Promising Therapeutic Target for Hepatocellular Carcinoma

Written By

Min Yao, Wenjie Zheng, Li Wang, Miao Fang, Dengfu Yao and Zhizheng Dong

Submitted: 26 May 2017 Reviewed: 14 September 2017 Published: 21 December 2017

DOI: 10.5772/intechopen.71007

From the Edited Volume

Hepatocellular Carcinoma - Advances in Diagnosis and Treatment

Edited by Costin Teodor Streba, Cristin Constantin Vere and Ion Rogoveanu

Chapter metrics overview

1,234 Chapter Downloads

View Full Metrics

Abstract

Oncogenic secretory clusterin (sCLU) is a stress-induced molecular chaperone that confers proliferative and survival advantages to hepatocellular carcinoma (HCC), plays a crucial role in cell proliferation, multiple drug resistance, metastasis, and tumor progression. However, the targeted effects and molecular mechanisms of sCLU for malignant tumor are still unknown. This chapter aims to review some progression of oncogenic sCLU as a promising therapeutic target for HCC. An English-language literature search was conducted using bibliographic databases on some valuable articles in focused review questions to analyze the interventions and findings of included studies using a conceptual framework. The positive rate of hepatic sCLU expression in cancerous tissues was significantly higher more than that in their surrounding non-cancerous ones at gene transcription level or at protein level, with increasing according to tumor-node-metastasis (TNM) staging. Abnormal expression of oncogenic sCLU associated with poor differentiation degree and TNM stage of HCC also has been considered as a valuable diagnostic or independent prognostic biomarker for HCC. Furthermore, silencing sCLU at mRNA level by specific shRNA or inhibition by OGX-011 suppressed the colony formation and proliferation of tumor cells with apoptosis increasing, cell cycle arrested, alterations of cell migration and invasion behaviors, decreasing phosphorylation level of Akt and GSK-3β in vitro, and significantly suppressing the xenograft tumor growth with decreasing expression of β-catenin, p-GSK3β, and cyclinD1 in vivo. The oncogenic sCLU expression was closely associated with tumor progression, and it should be a novel potential molecular-targeted therapy for HCC.

Keywords

  • hepatocellular carcinoma
  • secretory clusterin
  • targeted therapy

1. Introduction

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death worldwide [1]. Growing understanding of the multiple pathogenic factors including hepatitis B or C virus (HBV or HCV) infection, toxic, lipid accumulation, aflatoxin B1 intake, and so on with complex molecular mechanisms underlying HCC reveal that hepatocarcinogenesis is a multistep process including lots of activated or suppressed oncogenes or anticancer genes [2, 3, 4]. Some techniques for HCC therapy have experienced great progress. However, the prognosis of HCC patients is still very poor due to the high rates of tumor recurrence and metastasis. Effective therapy of HCC is dependent on early specific diagnosis, therefore, to provide optimal treatment for patients, more precise and effective markers are urgently needed in all phases of management from early detection to staging, treatment monitoring, and prognosis [5, 6, 7]. Numerous studies have shown the clinical utility of novel blood-based markers, such as circulating tumor cells, key signal molecules, long non-coding RNA, and microRNA with great potential for HCC [8, 9].

Molecular chaperones are proteins that response to cellular stresses including genotoxic agents, nutrient starvation, and heat shock, with cellular stresses-induced protein misfolding, aggregation, and denaturation [10, 11]. To date, only few specific markers such as hepatoma-specific γ-glutamyl transferase [12, 13], oncofetal antigen glypican-3 (GPC-3) [14, 15], hepatoma-specific alpha-fetoprotein (HS-AFP or AFP-L3) [16], member 3a of Wingless-type MMTV integration site family (Wnt3a) [17, 18], and molecular chaperones like heat shock proteins (Hsp27 or Hsp90) [19] and clusterin have been developed as valuable biomarkers for primary hepatocellular carcinoma (PHC) diagnosis and surveillance. The clusterin (CLU) that was first detected in HCC tissues by Tobe et al., who found that SP40-40 gene in hepatoma cells was located in human chromosome 8, also designated as apolipoprotein J (APOJ), SP-40, sulfated glycoprotein 2 (SGP2), and testosterone-repressed prostate message 2 (TRPM2) [20]. Following the detection of their complete cDNA cloning, sequencing and comparison, secretory CLU (sCLU) is found to be the mature isoform of cytoplasm endoplasmic reticulum (ER)-Golgi CLU, which is over-expressed in a wide variety of tumors with oncogenicity [21, 22]. Recently, the mechanisms of abnormal sCLU expression and its targeted effects for HCC have been explored [23, 24]. This article summarizes some progression of sCLU as a promising target for HCC gene therapy.

Advertisement

2. Gene structure and functions of sCLU

Human CLU gene is a single-copy gene on chromosome 8p21-p12 including 9 exons and 8 introns, encoding an mRNA of 2877 bp and translating to a polypeptide with 449 amino acids (a.a.) [25]. The secretory glycoprotein (1st a.a.) is a signal sequence of hydrophobic leader, and targets the ER protein. CLU gene encodes two isoforms with distinct functions as a result of alternative splicing and post-translational modifications: cytoplasm sCLU (75–80 kDa) and nuclear CLU (nCLU, 55 kDa), which is mainly located in the nucleus. The sCLU molecule is a highly conserved heterodimeric disulfide-linked 449 amino acid polypeptide that represents the major product of CLU gene. The ER-Golgi sCLU is considered to influence immune regulation, transformation, tissue remodeling, lipid transport, membrane recycling, complements cascade, DNA repair, cell adhesion, and cell-cell interactions, indicated that sCLU is widely distributed in tissues and body fluids involved in various physiological processes [11, 26, 27].

Multiple reports have shown that the cytoplasm sCLU is cytoprotective and anti-apoptotic [28, 29], whereas nCLU protein is proapoptotic. Abnormal oncogenic sCLU expression was reported to correlate closely with HCC progression, such as inducing epithelial-mesenchymal transition (EMT) [30], formation of multiple drug resistance (MDR) [31], distal metastasis of tumor cells, and malignant transformation of hepatocytes, interaction with oncogenes or suppressor genes, and related signal pathways (Figure 1) [32, 33]. Because of ischemic or hypoxic microenvironment existence in cancerous tissues, the sCLU are often adaptively over-expressed and closely related with increased tumorigenicity, metastatic potential, and MDR to chemotherapy. As a stress-induced chaperone that inhibits protein misfolding and aggregation in a manner similar to small heat shock proteins (HSPs) [34, 35], its promoter region contains an element recognized by heat shock factor 1 (HSF-1) [36]. Cytoplasm sCLU-inhibited apoptosis by interacting with activated Bax, and protects HCC cells from ER stress-induced apoptosis through a physical interaction with glucose-regulated prptein78 (GRP78) [29, 37].

Figure 1.

Potential mechanisms of hepatic sCLU in hepatocellular carcinoma. Cyto C, cytochrome c; ER, endoplasmic reticulum; Gene Reg., gene regulation; HIF-1α, hypoxia inducible factor-1α; IGF, insulin-like factor; MDR, multiple drug resistance; MMP, matrix metalloproteinase; nCLU, nuclear clusterin; NF-κB, nuclear factor-κB; PKC, protein kinase C; PI3K, phosphatidylinositol 3-kinase; Scr, sarcoma gene; sCLU, secretory clusterin.

Hepatic sCLU has been confirmed that it was physically associated with eukaryotic translation initiation factor 3 subunit I (EIF3I), and might protect EIF3I protein from degradation. A positive correlation was founded between sCLU and EIF3I, and both of their functions might be as a cooperative unit in HCC. The levels of sCLU and EIF3I expression were investigated in HCC using tissue microarray (TMA) and the patients with high EIF3I level exhibited poor prognosis. After silenced EIF3I, Akt phosphorylation was significantly inhibited. The EIF3I-Akt complex could prevent PP2A-mediated dephosphorylation, which in turn led to a constitutive Akt signal activation, suggesting that the CLU-EIF3I complex might prevent EIF3I degradation, and then contribute to Akt upregulation [33, 38].

Advertisement

3. Biological behaviors of sCLU expression in cancerous tissues

Although great efforts have been made to explore molecular mechanism of HCC invasion and metastasis in the past decade [39, 40, 41], the mechanism of HCC remains incompletely understood. The alterations of sCLU expression at messenger RNA (mRNA) or protein level were investigated in HCC- and their non-tumorous tissues (NT) with self-control [42]. The overall level of sCLU mRNA in the HCC group was 75% up-regulated, 7.5% down-regulated, and 17.5% non-changed. Although no significant difference of the sCLU mRNA level at staging I was found between NT and HCC, they were drastically up-regulated expression from staging II to IV. The staining of sCLU mainly presented in the cytoplasm at protein level in HCC and their NT tissues were analyzed by tissue microarray (TMA) with immunohistochemistry (IHC). Its incidence in the HCC group (73.3%), with 37.5, 68 and 88.9% at staging I, II and III & IV, was significantly higher than that in the NT group (23.3%), respectively. The levels of sCLU protein consistent with their mRNA expression were gradually upregulation with increasing HCC staging [42, 43], indicated that high sCLU should be a valuable biomarker to distinguish malignant from benign liver nodular lesions [44].

The sCLU as a functional homolog of HSPs is a stress-induced chaperone that confers proliferative and closely associates with poor prognosis of HCC. Recurrence and metastasis are the most causes of poor prognosis of HCC. Clinicopathological features of sCLU revealed that its high expression was significantly linked to poor differentiation and advanced TNM stage [45]. There was a trend toward a poorer overall survival in HCC with high sCLU expression. Besides, survival time of HCC with high TNM stage was significantly shorter than that of cases with low stage. Moreover, in the subset of HCC patients with III and IV stage, high sCLU expression was prone to result in a shorter survival time (Table 1). There is a closely positive correlation between abnormal sCLU expression and HCC. High sCLU expression has more invasive phenotype for HCC [46]. The upregulation of sCLU is associated with HCC progression by contributing to angiogenesis, chemoresistance, cells survival, and metastasis. Growing evidences showed that sCLU with molecular chaperones played an important role in MDR formation, cells proliferation, metastasis of HCC [47]. Furthermore, univariate and multivariate analyses indicated that sCLU might be an independent prognostic indicator, in line with the factor of lymph node metastasis.

GroupnPos. n (%)χ2 valueP value
AFP (μg /L)≤503731 (83.78)1.7330.118
>503827 (71.05)
Portal vein invasionWith76 (85.71)0.3090.578
Without6852 (76.47)
HBsAgNegative4636 (78.26)0.0580.809
Positive2922 (75.86)
Tumor size≤5 cm4533 (73.33)1.0270.311
>5 cm3025 (83.33)
Liver cirrhosisWith5745 (78.95)0.3530.552
Without1813 (72.22)
Lymph node metastasisWith2322 (95.65)6.3510.012*
Without5236 (69.23)
DifferentiationWell & moderate5843 (74.14)1.4910.222
Poor1715 (88.24)
Gross classificationUnifocal6246 (74.19)1.7440.187
Multifocal1312 (92.31)
TNMI & II4530 (66.67)7.6830.021*
III & IV3028 (93.33)
Child degreeA4430 (68.18)5.0860.024*
B&C3129 (90.32)
5 years’ survivalNo5143 (84.31)4.4300.035*
Yes2415 (62.50)

Table 1.

Clinicopathological features of hepatic sCLU expression in HCC.

Pos. n (%), positive number (%). *P<0.05

Advertisement

4. Circulating sCLU as diagnostic marker

The observations were in accordance with the early literature showing that upregulation of sCLU-positive expression might be associated with poor clinical outcome in HCC patients [21, 48]. According to previous clinical studies, average serum sCLU level was significantly higher in the HCC group more than that in any of groups with cirrhosis, chronic hepatitis, or healthy control (Table 2). The area under receiver operating characteristic (ROC) curve and diagnostic sensitivity were 0.75 and 74.7% in serum sCLU, and 0.74 and 58.7% in serum AFP, respectively. The incidence of both combining detection rose up to 90.7% for HCC diagnosis (Table 3). High-circulating sCLU levels were observed in HCC patients, consistent with a recent study using a three-step serum proteome analysis, which showed that serum sCLU levels in HCC were significantly higher than those in benign liver diseases [42].

GroupnMean ± SDPositive n (%)χ2 valueP value
sCLUμg/mL
HCC75119.21 ± 16.6756 (74.67)
LC3097.78 ± 19.068 (26.67)**20.744<0.001
CH30106.30 ± 19.2212 (40.00)**11.285<0.001
NC3689.96 ± 7.270 (0.00)**54.249<0.001
AFPng/mL
HCC752177.32 ± 3757.9944 (58.67)
LC30126.84 ± 244.7610 (33.33)*5.5050.019
CH3030.27 ± 50.095 (16.67)**15.188<0.001
NC367.1 ± 3.501 (2.78)**31.520<0.001

Table 2.

Serum sCLU or AFP levels among patients with chronic liver diseases and comparative analysis of both diagnostic values for HCC.

P < 0.05, compared with HCC group.


P < 0.01, compared with HCC group.


Serum sCLU values >104.0 μg/mL or AFP values >50 ng/mL were considered positive.

sCLU, secretory clusterin; LC, liver cirrhosis; CH, chronic hepatitis; NC, normal control; and AFP, α-fetoprotein.

sCLU level > 104.2 μg/mL (%)AFP level > 50 ng/mL (%)Both (%)
Sensitivity (%)74.6758.6790.67
Specificity (%)66.6775.0060.00
Accuracy (%)71.1165.9377.03
Positive predictive value (%)73.6874.5873.91
Negative predictive value (%)67.8059.2183.72

Table 3.

Assessment diagnostic validity of serum sCLU or AFP level for HCC.

AFP, α-fetoprotein; sCLU, secretory clusterin.

CLU is related to reverse cholesterol transport, platelet degranulation and human immune response pathways [49]. Protein-protein interaction analysis and pathway assessment showed a closed molecular relationship between cirrhosis and HCC [50]. Serum samples collected from HCCLM3-R metastatic HCC tumor model at specific stages of metastasis (1., 3 and 6 weeks) were subjected to iTRAQ labeling followed by 2DLC-ESI-MS/MS analysis. Circulating sCLU was significantly up-regulated during cancer progression and metastasis. The expression of sCLU was significantly higher in metastatic HCC cells and samples from HCC patients. Serum sCLU was highly increased with tumor size, tumor number, lymph node infiltration (Table 4) [42], and showed that the ROC area under curve value was 0.95 in sCLU more than that (0.85) in AFP. If 128 μg/mL as a cutoff value, the sensitivity or specificity of serum sCLU level for predicting HCC was 90 or 87%, respectively. The data indicated that circulating sCLU is a promising molecular marker of diagnosis or predicting metastasis for HCC [22, 51, 52].

GroupnAverage (μg/mL)t valueP valuePositive, n (%)χ2 valueP value
Age (years)
≤6047118.09 ± 16.450.7510.45533 (70.21)1.3200.251
>6028121.09 ± 17.1623 (82.14)
AFP (μg/L)
≤5031120.15 ± 17.120.4050.68624 (77.42)0.2120.645
>5044118.55 ± 16.5032 (72.73)
Portal vein invasion
With28119.34 ± 14.520.0500.96022 (78.57)0.3600.548
Without47119.14 ± 17.9734 (72.34)
Tumor size
≤5 cm34114.63 ± 18.102.2210.029a21 (61.76)5.4730.019
>5 cm41123.00 ± 14.5235 (85.37)
Lymph node metastasis
Without44114.84 ± 15.212.8260.006b29 (65.91)4.3160.038
With31125.40 ± 16.9127 (87.10)
Differentiation
Well & moderate57117.43 ± 16.341.6620.10142 (73.68)0.1210.728
Poor18124.84 ± 16.9014 (77.78)
Gross classification
Unifocal36114.02 ± 18.572.6980.009c23 (63.89)4.2510.039
Multifocal39124.00 ± 13.2033 (84.62)
TNM stage
I & II31113.93 ± 14.362.375 0.02d20 (64.52)2.8780.09
III & IV44122.93 ± 17.3136 (81.82)
Child classification
A55117.60 ± 14.561.3950.16740 (72.73)0.4100.522
B&C20123.63 ± 21.2316 (80.00)

Table 4.

Clinicopathologic features of serum sCLU expression in HCC patients (Mean ± SD).

With the tumor size ≥5 cm group.


The lymph node metastasis group.


With the multifocal group.


With TNM III & IV group.


AFP, α-fetoprotein; TNM, tumor node metastasis.

Advertisement

5. Application prospect of targeting CLU gene

5.1. Antisense oligonucleotide (ASO) therapy

ASO is a useful technique to inhibit specific-targeted CLU genes, with a small synthetic natural nucleic acid analogue, that can complementary to CLU mRNA that induce degradation or inhibit translation into protein [52]. It is considered to be a potent inhibitor of sCLU expression in vitro, in vivo, and in human clinical trials, with no apparent effect on the expression of nCLU. Custirsen (OGX-011, 5′-CAGCAGCA GAGTCTTCATCAT-3′, 50 nM) is a novel 2′-methoxy-ethyl-modified phosphorothioate ASO, which is a 21-nucleoside complement to target the translation initiation site of CLU gene exon II mRNA translation initiation site with one CpG motif [53]. Hence, OGX-011 plays the role of chemosensitization by influencing the anti-apoptotic protein sCLU instead of the proapoptotic protein nCLU. Xiu et al. have reviewed the current state of research on clusterin, to predict future research directions and to analyze the potential of the clinical application of custirsen in HCC [54]. However, the median overall survival of HCC cases were ms 23.4 mo in the group of the OGX-011 combining anti-cancer drugs (docetaxel/prednisone) and ms 22.2 mo in the other group of cases with docetaxel/prednisone alone. No significant difference was found between two groups. Some potential key factors might contribute to its results, and still want to do more clinical trials to be ongoing [55].

5.2. Reversal MDR by specific shRNA-targeted sCLU

Resistance of tumor cells to chemotherapy continues to be a major clinical obstacle to extend the survival rate of patients with HCC. Recently, one of the major strategies for liver cancer is surgical resection with adjuvant anti-HCC drug chemotherapy [10]. However, the HCC patients always tend to acquire MDR during tumor progression. MDR-related P-glycoprotein (P-gp), encoded by MDR1 gene is positively linked closely with chronic liver diseases, because of its drug efflux via ATP-binding cassette (ABC) family transporters, which can decrease intracellular concentration of anti-HCC drugs. Targeting sCLU to sensitize cancer cells to chemotherapy has become an attractive new strategy for cancer treatment. Previous studies found sCLU expressed in line with P-gp via immunohistochemical staining. The sCLU expression was analyzed in human hepatoma cells and chemoresistant counter-part HepG2/ADM cells. After transfection of shRNA-1 (5′-GTAAGTACGTC AATAAGGA-3′) into HepG2/ADM cells, the inhibition of CLU expression was 73.68% at mRNA level with obvious enhancement in chemosensitivity, and increasing apoptosis induced by doxorubicin [56]. Knockdown CLU also significantly decreased the drug efflux pump activity through the depression of MDR1/P-gp. Moreover, silencing CLU led to downregulation of β-catenin, suggesting that downregulation of CLU might be a key point to reverse MDR of HepG2/ADM cells [57].

5.3. Blockade-related pathway

5.3.1. PI3K/Akt/NF-κB pathway

Previous data revealed that CLU promoted cell survival through the PI3K/Akt pathway and induced MMP-9 expression via ERK1/2 and PI3K/Akt/NF-κB pathways [32]. CLU could increase p-Akt and MMP13 expression. A positive correlation between CLU expression and p-Akt level was observed in cohort of HCC tissues. Where CLU knockdown using OGX-011 significantly decreased p-Akt and MMP13 levels and suppressed HCC metastasis in two metastatic models through inhibiting EIF3I/Akt/MMP13 signaling. The related signaling molecule blockade of the PI3K-Akt pathway could significantly inhibited MMP13 expression in human HepG2-CLU or HCCLM3 cells [38, 58]. Decreased level of CLU accompanied with downregulation of MMP13 and p-Akt was observed in tumors derived from HCCLM3-shCLU group, revealed that p-Akt level was significantly correlated with poor prognosis and indicated that CLU may play a crucial role in HCC metastasis [38].

5.3.2. Wnt/β-catenin pathway

Wnt canonical pathway is often constitutively active in neoplastic cells, although, normally β-catenin is negatively regulated by GSK-3β that phosphorylates β-catenin to drive it for proteosomal degradation [30, 59]. Previous data emphasized sCLU modification after being exposed to Wnt⁄β-catenin inhibitor, and expressions of the crucial β-catenin and GSK-3β genes were detected in cases of sCLU depletion. The data indicated that sCLU suppression might lead to the inhibition of Wnt/β-catenin pathway in reverse. Hence, sCLU might play an important role in chemoresistance of HepG2/ADM cells together with Wnt/β-catenin signaling molecules [31, 56].

5.3.3. IGF-1/IGF-1R/Src/Mek/Erk pathway

Hepatic sCLU is a general genotoxic stress-induced, prosurvival gene product implicated in cancer [36, 37]. The regulatory signal transduction processes that control sCLU expression, the induction of sCLU is delayed, peaking 72 h after low doses of ionizing radiation, and is dependent on up-regulating IGF-1 and phosphorylation-dependent IGF-1R activation [23, 27] that stimulates the downstream Src-Mek-Erk signal transduction cascade to ultimately transactivate the early growth response-1 (Egr-1) transcription factor. Thus, the ionizing radiation exposure causes stress-induced IGF-1R-Src-Mek-Erk-Egr-1 activation that regulates the sCLU prosurvival cascade pathway for radiation resistance in HCC therapy [60].

5.4. Suppressed HCC growth in vivo by silencing sCLU

The inhibition of sCLU gene transcription by specific shRNA-1 on effects of forming time, growth curve, and size of orthotopic xenograft tumors after sacrifice of the mice at the 34th day with injection are shown in Figure 2. The mean weight of the xenograft tumors in the shRNA-1 group was significantly less than that of the control or NC-shRNA group, respectively. The curves of xenograft tumor growth indicated that tumor sizes in the shRNA-1 group with lower mRNA level were significantly smaller less than those of the control or NC-shRNA group [31, 46]. Consistently, the sCLU protein expression in the shRNA-1 group was also lower than that in the control or the NC-shRNA group; and the sCLU staining in the control or NC-shRNA group was stronger than that in the shRNA-1 group by immunohistochemistry [46]. Specific shRNA-mediated downregulation of sCLU resulted in a reduced migratory capacity in HCC cell lines, as well as a reduction in pulmonary metastasis in vivo [38]. Overexpression of sCLU in HepG2 cell line showed increased cell migratory ability. In addition, sCLU also plays an important role in the regulation of TGF-β1-smad3 signaling pathway, suggested that oncogenic sCLU might promote HCC metastasis via the induction of EMT process and could be a promising candidate target for HCC therapy [32, 61, 62, 63].

Figure 2.

Inhibition of sCLU gene transcription by specific shRNA-1 on effects of forming time, growth curve and size of orthotopic xenograft tumors. The control, neg-shRNA, or sCLU-shRNA groups were injected with the plasmid contained specific shRNA-1 into nude mice. (A) The forming time of the orthotopic xenograft tumors from each group. (B) The growth curves of the xenograft tumors from each group. And the tumor volumes were measured at the indicated time point. (C) The representative photographs of the nude mice and corresponding dissected tumors from each group. 1 and 2, the xenografts in the control group; 3 and 4, the xenografts in the neg-shRNA group; 5 and 6, the xenografts in the sCLU-shRNA group. The data are presented as the mean ± SD. Compared with the control group, **P < 0.01.

Advertisement

6. Conclusions

In conclusion, the upregulation of sCLU expression at early staging of HCC is considered to promote tumor development, which may be related to the phosphorylation of AKT/GSK-3β. An increasing number of reports have provided evidence that sCLU level could be a novel biomarker for HCC diagnosis and prognosis, and there will be of great significance for the individualized treatment in HCC patients. The sCLU regulating signaling pathways could be critical to unraveling the solution for MDR in HCC. Therefore, silencing sCLU gene transcription and inhibiting sCLU expression by specific Custirsen inhibition have provided a new mechanism insight into molecular-targeted therapy for HCC in injected- or orthotopic model, indicated that sCLU gene would be a potential molecular-targeted for HCC therapy. Further study found that sCLU contributed to HCC migration and EMT in vitro, and metastasis in vivo. Although additional preclinical and clinical trials are necessary to explore the sCLU role in HCC, targeting the oncogenic sCLU could validate the approach as a systemic therapy to increase chemotherapy sensitivity.

Advertisement

Acknowledgments

This work was supported by grants-in-aid from Projects of the National Natural Science Foundation (81673241, 81702419), the Jiangsu Health Plans (2014-YY-028 and BE2016698), the Jiangsu Graduate Innovation Plan (KYCX17_1934), the Nantong Science Foundation of Health and Family Planning Commission (WQ2016083), and the International Science & Technology Cooperation Program (2013DFA32150) of China.

Advertisement

Abbreviations

AFPalpha-fetoprotein
HCChepatocellular carcinoma
IHCimmunohistochemistry
MDRmultiple drug resistance
miRNAmicroRNA
sCLUsecretory clusterin
shRNAshort hairpin RNA
TMAtissue microarray
VEGFvascular endothelial growth factor
WntWnt/β-catenin signaling pathway

References

  1. 1. Jayachandran M. An updated portrait of pathogenesis, molecular markers and signaling pathways of hepatocellular carcinoma. Current Pharmaceutical Design. 2017. 23(16):2356-2365 [PMID: 28356044]
  2. 2. Kirstein MM, Vogel A. The pathogenesis of hepatocellular carcinoma. Digestive Diseases. 2014;32(5):545-553 [PMID: 25034287]
  3. 3. Nuño Solinís R, Arratibel Ugarte P, Rojo A, Sanchez Gonzalez Y. Value of treating all stages of chronic hepatitis C: A comprehensive review of clinical and economic evidence. Infectious Disease and Therapy. 2016;5(5):491-508 [PMID: 27783223]
  4. 4. Bellentani S. The epidemiology of non-alcoholic fatty liver disease. Liver International. 2017;37(S1):81-84 [PMID: 24843434
  5. 5. Bang CS, Song IH. Impact of antiviral therapy on hepatocellular carcinoma and mortality in patients with chronic hepatitis C: Systematic review and meta-analysis. BMC Gastroenterology. 2017;17(1):46-65 [PMID: 28376711]
  6. 6. Hollebecque A, Malka D, Ferte C, Ducreux M, Boige V. Systemic treatment of advanced hepatocellular carcinoma: From disillusions to new horizons. European Journal of Cancer. 2015;51(3):327-339 [PMID: 25559615]
  7. 7. Choi SH, Park JY. Regulation of the hypoxic tumor environment in hepatocellular carcinoma using RNA interference. Cancer Cell International. 2017;17(1):3-11 [PMID: 28053598]
  8. 8. Bruix J, Reig M, Sherman M. Evidence-based diagnosis, staging, and treatment of patients with hepatocellular carcinoma. Gastroenterology. 2016;150(4):835-853 [PMID: 26795574]
  9. 9. Juárez-Hernández E, Motola-Kuba D, Chávez-Tapia NC, Uribe M, Barbero Becerra V. Biomarkers in hepatocellular carcinoma: An overview. Expert Review of Gastroenterology & Hepatology. 2017;11(6):549-558 [PMID: 28347162]
  10. 10. Zoubeidi A, Chi K, Gleave M. Targeting the cytoprotective chaperone, clusterin, for treatment of advanced cancer. Clinical Cancer Research. 2010;16(4):1088-1093 [PMID: 20145158]
  11. 11. Trougakos IP, Djeu JY, Gonos ES, Boothman DA. Advances and challenges in basic and translational research on clusterin. Cancer Research. 2009;69(2):403-406 [PMID: 19147550]
  12. 12. Yao DF, Huang ZW, Chen SZ, Huang JF, Lu JX, Xiao MB, Meng XY. Diagnosis of hepatocellular carcinoma by quantitative detection of hepatoma-specific bands of serum gamma-glutamyl transferase. American Journal of Clinical Pathology. 1998;110(6):743-749 [PMID: 9844586]
  13. 13. Yao DF, Jiang DR, Huang ZW, Lu JX, Tao QY, Yu ZJ, Meng XY. Abnormal expression of hepatoma specific γ-glutamyl transferase and alteration of γ-glutamyl transferase gene methylation status in patients with hepatocellular carcinoma. Cancer. 2000;88(4):761-769 [PMID: 10679644]
  14. 14. Yao M, Wang L, Fang M, Zheng W, Dong Z, Yao D. Advances in the study of oncofetal antigen glypican-3 expression in HBV-related hepatocellular carcinoma. Bioscience Trends. 2016;10(5):337-343 [PMID: 27795482]
  15. 15. Wang L, Pan L, Yao M, Cai Y, Dong Z, Yao D. Expression of oncofetal antigen glypican-3 associates significantly with poor prognosis in HBV-related hepatocellular carcinoma. Oncotarget. 2016;7(27):42150-42158 [PMID: 27286460]
  16. 16. Wu W, Yao DF, Yuan YM, Fan JW, Lu XF, Li XH, Qiu LW, Zong L, Wu XH. Combined serum hepatoma-specific alpha-fetoprotein and circulating alpha-fetoprotein-mRNA in diagnosis of hepatocellular carcinoma. Hepatobiliary & Pancreatic Diseases International. 2006;5(4):538-544 [PMID: 17085339]
  17. 17. Pan LH, Yao M, Zheng WJ, Gu JJ, Yang XL, Qiu LW, Cai Y, Wu W, Yao DF. Abnormality of Wnt3a expression as novel specific biomarker for diagnosis and differentiation of hepatocellular carcinoma. Tumour Biology. 2016;37:5561-5568 [PMID: 26577850]
  18. 18. Pan LH, Yao M, Cai Y, Gu JJ, Yang XL, Wang L, Yao DF. Oncogenic Wnt3a expression as an estimable prognostic marker for hepatocellular carcinoma. World Journal of Gastroenterology. 2016;22:3829-3837 [PMID: 27076768]
  19. 19. Malhi H, Kaufman RJ. Endoplasmic reticulum stress in liver disease. Journal of Hepatology. 2011;54:795-809 [PMID: 21145844]
  20. 20. Tobe T, Minoshima S, Yamase S, Choi NH, Tomita M, Shimizu N. Assignment of a human serum glycoprotein SP-40,40 gene (CLI) to chromosome 8. Cytogenetics and Cell Genetics. 1991;57:193-195 [PMID: 1660393]
  21. 21. Bertuzzi M, Marelli C, Bagnati R, Colombi A, Fanelli R, Saieva C, Ceroti M, Bendineli B, Caini S, Airoldi L, Palli D. Plasma clusterin as a candidate pre-diagnosis marker of colorectal cancer risk in the florence cohort of the European prospective investigation into cancer and nutrition: A pilot study. BMC Cancer. 2015;15(1):56-67 [PMID: 25884309]
  22. 22. Nafee AM, Pasha HF, Abd EL, Aal SM, Mostafa NA. Clinical significance of serum clusterin as a biomarker for evaluating diagnosis and metastasis potential of viral-related hepatocellular carcinoma. Clinical Biochemistry. 2012;45(13-14):1070-1074 [PMID: 22580393
  23. 23. Luo X, Suzuki M, Ghandhi SA, Amundson SA, Boothman DA. ATM regulates insulin-like growth factor 1-secretory clusterin (IGF-1-sCLU) expression that protects cells against senescence. PLoS One. 2014;9(6):e99983 [PMID: 24937130]
  24. 24. Essabbani A, Garcia L, Zonetti MJ, Fisco T, Pucci S, Chiocchia G. Exon-skipping strategy by ratio modulation between cytoprotective versus pro-apoptotic clusterin forms increased sensitivity of LNCaP to cell death. PLoS One. 2013;8(2):e54920 [PMID: 23418433]
  25. 25. Novinec M, Lenarčič B, Baici A. Clusterin is a specific stabilizer and liberator of extracellular cathepsin K. FEBS Letters. 2012;586(7):1062-1066 [PMID: 22569264]
  26. 26. Liao FT, Lee YJ, Ko JL, Tsai CC, Tseng CJ, Sheu GT. Hepatitis delta virus epigenetically enhances clusterin expression via histone acetylation in human hepatocellular carcinoma cells. The Journal of General Virology. 2009;90(Pt 5):1124-1134 [PMID:19264665]
  27. 27. Goetz EM, Shankar B, Zou Y, Morales JC, Luo X, Araki S, Bachoo R, Mayo LD, Boothman DA. ATM-dependent IGF-1 induction regulates secretory clusterin expression after DNA damage and in genetic instability. Oncogene. 2011;30(35):3745-3754 [PMID:21460853]
  28. 28. Wang X, Luo L, Dong D, Yu Q, Zhao K. Clusterin plays an important role in clear renal cell cancer metastasis. Urologia Internationalis. 2014;92(1):95-103 [PMID:24008723]
  29. 29. Zhang H, Kim JK, Edwards CA, Xu Z, Taichman R, Wang CY. Clusterin inhibits apoptosis by interacting with activated Bax. Nature Cell Biology. 2005;7(9):909-915 [PMID: 16113678]
  30. 30. Wang C, Jiang K, Kang X, Gao D, Sun C, Li Y, Sun C, Zhang S, Liu X, Wu W, Yang P, Guo K, Liu Y. Tumor-derived secretory clusterin induces epithelial-mesenchymal transition and facilitates hepatocellular carcinoma metastasis. The International Journal of Biochemistry & Cell Biology. 2012;44(12):2308-2320 [PMID: 23010347]
  31. 31. Zheng WJ, Sai WL, Yao M, Cai Y, Pan LH, Gu JJ, Wu W, Yao DF. Down-regulated clusterin expression enhances sensitivity of hepatoma cells to anti-cancer drugs. Zhonghua Gan Zang Bing Za Zhi. 2015;23(11):844-848 [PMID: 26743245]
  32. 32. Shim YJ, Kang BH, Jeon HS, Park IS, Lee KU, Lee IK, Park GH, Lee KM, Schedin P, Min BH. Clusterin induces matrix metalloproteinase-9 expression via ERK1/2 and PI3K/Akt/NF-kappaB pathways in monocytes/macrophages. Journal of Leukocyte Biology. 2011;90(4):761-769 [PMID: 21742938]
  33. 33. Ammar H, Closset JL. Clusterin activates survival through the phosphatidylinositol 3-kinase/ Akt pathway. The Journal of Biological Chemistry. 2008;283(19):12851-12861 [PMID: 18321852]
  34. 34. Wang C, Zhang Y, Guo K, Wang N, Jin H, Liu Y, Qin W. Heat shock proteins in hepatocellular carcinoma: Molecular mechanism and therapeutic potential. International Journal of Cancer. 2016;138(8):1824-1834 [PMID: 26853533]
  35. 35. Lamoureux F, Thomas C, Yin MJ, Fazli L, Zoubeidi A, Gleave ME. Suppression of heat shock protein 27 using OGX-427 induces endoplasmic reticulum stress and potentiates heat shock protein 90 inhibitors to delay castrate-resistant prostate cancer. European Urology. 2014;66(1):145-155 [PMID: 24411988]
  36. 36. Loison F, Debure L, Nizard P, le Goff P, Michel D, le Drean Y. Up-regulation of the clusterin gene after proteotoxic stress: Implication of HSF1-HSF2 heterocomplexes. The Biochemical Journal. 2006;395(1):223-231 [PMID: 16336210]
  37. 37. Wang C, Jiang K, Gao D, Kang X, Sun C, Zhang Q, Li Y, Sun L, Zhang S, Guo K, Liu Y. Clusterin protects hepatocellular carcinoma cells from endoplasmic reticulum stress induced apoptosis through GRP78. PLoS One. 2013;8(2):e55981 [PMID: 23457489]
  38. 38. Wang C, Jin G, Jin H, Wang N, Luo Q, Zhang Y, Gao D, Jiang K, Gu D, Shen Q, Huo X, Hu F, Ge T, Zhao F, Chu W, Shu H, Yao M, Cong W, Qin W. Clusterin facilitates metastasis by EIF3I/Akt/MMP13 signaling in hepatocellular carcinoma. Oncotarget. 2015;6(5):2903-2916 [PMID: 25609201]
  39. 39. Maluccio M, Covey A. Recent progress in understanding, diagnosing, and treating hepato-cellular carcinoma. CA: A Cancer Journal for Clinicians. 2012;62(6):394-399 [PMID: 23070690]
  40. 40. Qu Z, Yuan CH, Yin CQ, Quan Q, Chen H, Wang FB. Meta-analysis of the prognostic value of abnormally expressed lncRNAs in hepatocellular carcinoma. OncoTargets and Therapy. 2016;9(8):5143-5152 [PMID: 27574455]
  41. 41. Wang Y, Zeng LI, Chen W. HBV X gene point mutations are associated with the risk of hepatocellular carcinoma: A systematic review and meta-analysis. Molecular and Clinical Oncology. 2016;4(6):1045-1051 [PMID: 27284442]
  42. 42. Zheng W, Yao M, Sai W, Qian Q, Pan L, Qiu L, Huang J, Wu W, Yao D. Diagnostic and prognostic significance of secretory clusterin expression in patients with hepatocellular carcinoma. Tumour Biology. 2016;37(1):999-1008 [PMID: 26264614]
  43. 43. Kang YK, Hong SW, Lee H, Kim WH. Overexpression of clusterin in human hepatocellular carcinoma. Human Pathology. 2004;35(11):1340-1346 [PMID: 15668890]
  44. 44. Lai JP, Chen ZM, Lok T, Chan OT, Himmelfarb E, Zhai Q, Lin F, Wang HL. Immunohistochemical stains of proliferating cell nuclear antigen, insulin-like growth factor 2 and clusterin help distinguish malignant from benign liver nodular lesions. Journal of Clinical Pathology. 2014;67(6):464-469 [PMID: 24407433]
  45. 45. Lau SH, Sham JS, Xie D, Tzang CH, Tang D, Ma N, Hu L, Wang Y, Wen JM, Xiao G, Zhang WM, Lau GK, Yang M, Guan XY. Clusterin plays an important role in hepatocellular carcinoma metastasis. Oncogene. 2006;25(8):1242-1250 [PMID: 16247463]
  46. 46. Zheng W, Yao M, Qian Q, Sai W, Qiu L, Yang J, Wu W, Dong Z, Yao D. Oncogenic secretory clusterin in hepatocellular carcinoma: Expression at early staging and emerging molecular target. Oncotarget. 2017;8(32):52321-52323 [PMID: 28881732]
  47. 47. Klokov D, Leskov K, Araki S, Zou Y, Goetz EM, Luo X, Willson D, Boothman DA. Low dose IR-induced IGF-1-sCLU expression: A p53-repressed expression cascade that interferes with TGFbeta1 signaling to confer a pro-survival by stander effect. Oncogene. 2013;32(4):479-490 [PMID: 22391565]
  48. 48. Kimura A, Sogawa K, Satoh M, Kodera Y, Yokosuka O, Tomonaga T, Nomura F. The application of a three-step serum proteome analysis for the discovery and identification of novel biomarkers of hepatocellular carcinoma. International Journal of Proteomics. 2012;2012:623190 [PMID: 22957256]
  49. 49. Wang Y, Liu YH, Mai SJ, He LJ, Liao YJ, Deng HX, Guan XY, Zeng YX, Kung HF, Xie D. Evaluation of serum clusterin as a surveillance tool for human hepatocellular carcinoma with hepatitis B virus related cirrhosis. Journal of Gastroenterology and Hepatology. 2010;25(6):1123-1128 [PMID: 20594228]
  50. 50. Ehsani Ardakani MJ, Safaei A, Arefi Oskouie A, Haghparast H, Haghazali M, Mohaghegh Shalmani H, Peyvandi H, Naderi N, Zali MR. Evaluation of liver cirrhosis and hepatocellular carcinoma using Protein-Protein Interaction Networks. Gastroenterology and Hepatology From Bed to Bench. 2016;9(Suppl1):S14-S22 [PMID: 28224023
  51. 51. Comunale MA, Wang M, Rodemich-Betesh L, Hafner J, Lamontagne A, Klein A, Marrero J, Di Bisceglie AM, Gish R, Block T, Mehta A. Novel changes in glycosylation of serum Apo-J in patients with hepatocellular carcinoma. Cancer Epidemiology, Biomarkers & Prevention. 2011;20(6):1222-1229 [PMID: 21467232]
  52. 52. Chen D, Wang Y, Zhang K, Jiao X, Yan B, Liang J. Antisense oligonucleotide against clusterin regulates human hepatocellular carcinoma invasion through transcriptional regulation of matrix metalloproteinase-2 and E-cadherin. International Journal of Molecular Sciences. 2012;13(8):10594-10607 [PMID: 22949882]
  53. 53. Lamoureux F, Thomas C, Yin MJ, Kuruma H, Beraldi E, Fazli L, Zoubeidi A, Gleave ME. Clusterin inhibition using OGX-011 synergistically enhances Hsp90 inhibitor activity by suppressing the heat shock response in castrate-resistant prostate cancer. Cancer Research. 2011;71(17):5838-5849 [PMID: 21737488]
  54. 54. Xiu P, Dong XF, Li XP, Li J. Clusterin: Review of research progress and looking ahead to direction in hepatocellular carcinoma. World Journal of Gastroenterology. 2015;21(27):8262-8270 [PMID: 26217078]
  55. 55. Sad F, Hotte S, North S, Eigl B, Chi K, Czaykowski P, Wood L, Pollak M, Berry S, Lattouf JB, Mukherjee SD, Gleave M, Winquist E. Randomized phase II trial of Custirsen (OGX-011) in combination with docetaxel or mitoxantrone as second-line therapy in patients with metastatic castrate-resistant prostate cancer progressing after first-line docetaxel: CUOG trial P-06c. Clinical Cancer Research. 2011;17(17):5765-5773 [PMID: 21788353]
  56. 56. Zheng W, Sai W, Yao M, Gu H, Yao Y, Qian Q, Yao D. Silencing clusterin gene transcription on effects of multidrug resistance reversing of human hepatoma HepG2/ADM cells. Tumour Biology. 2015;36(5):3995-4003 [PMID: 25600802]
  57. 57. Xiu P, Xu Z, Liu F, Li Z, Li T, Zou F, Sun X, Li J. Downregulating sCLU enhances the sensitivity of hepatocellular carcinoma cells to gemcitabine by activating the intrinsic apoptosis pathway. Digestive Diseases and Sciences. 2014;59(8):1798-1809 [PMID: 24671452]
  58. 58. Wang YW, Lin KT, Chen SC, Gu DL, Chen CF, Tu PH, Jou YS. Over-expressed-eIF3I interacted and activated oncogenic Akt1 is a theranostic target in human hepatocellular carcinoma. Hepatology. 2013;58(1):239-250 [PMID: 23460382]
  59. 59. Yao M, Wang L, Qiu L, Qian Q, Yao D. Encouraging microRNA-based therapeutic strategies for hepatocellular carcinoma. Anticancer Agents Medicinal Chemistry. 2015;15:453-460 [PMID: 25511513]
  60. 60. Criswell T, Beman M, Araki S, Leskov K, Cataldo E, Mayo LD, Boothman DA. Delayed activation of insulin-like growth factor-1 receptor/Src/MAPK/Egr-1 signaling regulates clusterin expression, a pro-survival factor. The Journal of Biological Chemistry. 2005;280(14):14212-14221 [PMID: 15689620]
  61. 61. Shiota M, Zardan A, Takeuchi A, Kumano M, Beraldi E, Naito S, Zoubeidi A, Gleave ME. Clusterin mediates TGF-beta-induced epithelial-mesenchymal transition and metastasis via Twist1 in prostate cancer cells. Cancer Research. 2012;72(20):5261-5272 [PMID: 22896337]
  62. 62. Nannuru KC, Futakuchi M, Varney ML, Vincent TM, Marcusson EG, Singh RK. Matrix metalloproteinase (MMP)-13 regulates mammary tumor-induced osteolysis by activating MMP9 and transforming growth factor-beta signaling at the tumor-bone interface. Cancer Research. 2010;70(9):3494-3504 [PMID: 20406980]
  63. 63. Kalyan A, Nimeiri H, Kulik L. Systemic therapy of hepatocellular carcinoma: Current and promising. Clinics in Liver Disease. 2015;19(2):421-432 [PMID: 25921671]

Written By

Min Yao, Wenjie Zheng, Li Wang, Miao Fang, Dengfu Yao and Zhizheng Dong

Submitted: 26 May 2017 Reviewed: 14 September 2017 Published: 21 December 2017