Adenovirus encoding XAF-1 and TNF‑α in the same open reading frame efficiently inhibits hepatocellular cancer cells

  • Authors:
    • Kai Li
    • Xinhong Li
    • Zhongjun Wu
    • Liansheng Zheng
    • Yuqin Cui
    • Jun Wang
    • Yin Huang
    • Zhihong Yan
  • View Affiliations

  • Published online on: April 26, 2016     https://doi.org/10.3892/mmr.2016.5193
  • Pages: 5169-5176
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

X‑linked inhibitor of apoptosis (XIAP)‑associated factor 1 (XAF‑1), a tumor suppressor, is downregulated in most human malignant tumors. However, the tumor suppressive role of XAF‑1 in hepatocellular carcinoma (HCC) and its therapeutic value require further elucidation. The present study examined the expression of XAF‑1 at the mRNA and protein level in the HCC and paired peritumor tissue specimens, as well as in HCC cell lines and a normal liver cell line. A recombinant adenovirus which co‑expressed XAF‑1 and TNF‑α was then constructed, and its effects on the proliferation and colony formation ability of the MHCC97H HCC cell line were assessed using apoptosis induction, flow cytometry, trypan blue staining assay and a clonogenic assay. The results demonstrated that the expression of XAF‑1 was significantly reduced in HCC tissues compared with that in their matched peritumor specimens, and a significant correlation with the tumor size, stage and tumor ‑ nodes ‑ metastasis stage was identified. The reduced levels of XAF‑1 were further confirmed the HCC cell lines MHCC97L, HepG2 and MHCC97H compared with those in the L02 normal liver cell line. The recombinant adenovirus Ad‑XAF‑1&TNF‑α, which co‑expressed XAF‑1 and TNF‑α, was shown to efficiently express the two proteins at the mRNA and protein level. Furthermore, infection with Ad‑XAF‑1&TNF‑α synergistically induced apoptosis, reduced the proliferation and colony formation ability of MHCC97L cells to a significantly greater extent than overexpression of XAF‑1 or TNF‑α individually. To the best of our knowledge, the present study was the first to construct an adenovirus which co‑expressed XAF‑1 and TNF‑α in the same open reading frame and expressed them proportionally. As Ad‑XAF‑1&TNF‑α inhibited HCC cells with enhanced efficiency, it may be applicable for the treatment of HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common malignant cancer types worldwide and is accountable for almost 600,000 mortalities each year worldwide (1); it is also has the second highest mortality rate amongst all cancer types in China (2). The main risk factors for HCC development include chronic hepatitis B and C infection, alcohol abuse and aflatoxin intake (3,4), as they induce liver cirrhosis, from which 80% of HCCs are derived (5). Activation of oncogenes and inactivation of tumor suppressor genes have been identified to be associated with carcinogenesis and progression of HCC. Various genes have been identified to be differentially expressed in HCC tissues compared with paratumor tissues, including HIWI IGF2, FAT10, SCARA5, DLK1, p53 and ZNF267 (612), which have either oncogenic or tumor suppressive roles, indicating that HCC is based on complex oncogenic factors.

Besides oncogene activation and deregulation of apoptosis-associated genes, inactivation of tumor suppressor genes has also been associated with HCC (13). Evasion of apoptosis and angiogenesis are typical cancer-associated processes, whose reversal is an efficient therapeutic strategy for HCC (14) and other tumor types (15). Inhibitors of apoptosis (IAPs) are characterized by highly conserved baculoviral IAP repeats (16), belonging to a family of endogenous inhibitors of caspases (17,18). X-linked IAP (XIAP) prevents the activities of caspase-3, -7 and -9 via directly binding to these caspases (19). Overexpression of XIAP has been reported in most human cancer types, including HCC, and to be an independent prognostic factor for HCC patients (20). Inhibition of XIAP induces apoptosis and inhibits the growth of HCC cells (21), implying that targeting XIAP may be a promising approach for HCC therapy. XIAP-associated factor (XAF)-1 specifically inhibits IAP and sensitizes cancer cells to apoptosis (22), resulting in a pro-apoptotic effect (23). Thus, this antagonist may have significant value in the treatment of cancer.

In the present study, a recombinant adenovirus was constructed, which carries a coding sequence for XAF-1 and another sequence encoding tumor necrosis factor (TNF)-α, which induces apoptosis similarly to XAF-1, with the 2A peptide coding sequence (24). The anti-tumor effects of this recombinant adenovirus was then assessed in HCC cells in vitro. The present study provided a novel strategy for the treatment of HCC.

Materials and methods

Tissue specimens, cell lines and culture

A total of 56 HCC intratumor specimens and 56 paired peritumor specimens (as controls; obtained at a distance of >10 mm from the tumor edge) were included in the present study. All specimens were obtained from the pathological archives of Baotou Cancer Hospital (Batou, China) and had been obtained between May 2009 and June 2014 with informed consent of the patients. The HCC specimens had been obtained by surgical resection, immediately frozen in liquid nitrogen and stored at −80°C prior to radiotherapy or chemotherapy. Clinico-pathological characteristics of each patient are listed in Table I. The present study was approved by the Medical Ethics Committee of Baotou Cancer Hospital (Batou, China).

Table I

Association of XAF-1 mRNA with clinico-pathological characteristics of hepatocellular carcinoma patients [mean age, 53.4±10.3 for XAF-1 levels ≥1 and 51.5±9.6 years for XAF-1 levels <1 (P=0.7620)].

Table I

Association of XAF-1 mRNA with clinico-pathological characteristics of hepatocellular carcinoma patients [mean age, 53.4±10.3 for XAF-1 levels ≥1 and 51.5±9.6 years for XAF-1 levels <1 (P=0.7620)].

CharacteristicXAF-1 mRNA levels
P-value
≥1 (n=20)<1 (n=36)
Age (years)0.2012
 ≥501317
 <50719
HBsAg0.1878
 Negative23
 Positive813
AFP0.1385
 ≥200 ng/ml37
 <200 ng/ml79
Tumor size0.0406
 ≥5 cm310
 <5 cm76
BCLC stage0.0470
 0-B88
 C-D28
TNM stage0.0274
 I+II87
 III+IV29
Vascular invasion0.0731
 Yes15
 No911

[i] The XAF-1 mRNA levels were relative to β-actin. HBsAG, hepatitis B surface antigen; BCLC, Barcelona clinic liver cancer; TNM, tumor, nodes, metastasis; AFP, alpha-fetoprotein.

The MHCC97L, HepG2 and MHCC97H human HCC cell lines and the L02 control liver cell line were purchased from the cell resource center of the Chinese Academy of Medical Sciences (Beijing, China). Each cell line was cultured in Dulbecco's modified Eagle's medium (DMEM; Ameresco, Inc., Framingham, MA, USA) with 10% fetal bovine serum (FBS; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) at 37°C in a humidified atmosphere containing 5% CO2.

Construction of an adenovirus co-expressing XAF-1 and TNF-α (Ad-XAF-1&TNF-α)

The open reading frame (ORF) of human XAF-1 (NM_017523) and TNF-α (NM_000594) was amplified by polymerase chain reaction (PCR) with primers that deleted the stop codon, and was overlapped with a sequence encoding a 2A peptide linker (24). The overlapped XAF-1 - 2A - TNF-α nucleotide was inserted into the pShuttle-cytomegalovirus (CMV) vector (Qbiogene, Inc., Irvine, CA, USA) to generate the recombinant pShuttle-CMV - XAF-1 - 2A - TNF-α. The adenovirus Ad-XAF-1&TNF-α and the Ad-control (Ad-con) virus were enveloped via co-transfecting the pShuttle-CMV - XAF-1 - 2A - TNF-α and the pAdeasy-1 (the viral DNA plasmid) into 293GPG retrovirus packaging cell line (Cell Resource Center of the Chinese Academy of Medical Sciences) using Lipofectamine™ 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). To co-express XAF-1 and TNF-α in HCC cells, MHCC97L cells were infected with Ad-XAF-1&TNF-α at a multiplicity of infection (MOI) of 1 or 10 for 2 h, followed by culture in fresh DMEM containing 2% FBS.

RNA isolation and reverse-transcription quantitative PCR RT-qPCR

Cellular mRNA was isolated from tissues or cell lines using TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's manual, following homogenization of tissues. RT-qPCR was performed using the One Step SYBR® Green RT-qPCR kit (Sigma-Aldrich, St. Louis, MO, USA) following the manufacturer's instructions. The PCR reaction conditions were as follows: Initial denaturation, 5 min at 95°C; 40 cycles of denaturation for 20 sec at 94°C, annealing for 20 sec at 61°C and extension for 20 sec at 72°C; and a final extension for 5 min at 72°C. The primers for XAF-1, TNF-α and β-actin were synthesized by Invitrogen; Thermo Fisher Scientific, Inc., and were as follows: Forward, 5′-CCCAGGGACCTCTCTCTAATC-3′ and reverse, 5′-ATGGGCTACAGGCTTGTCACT-3′ for TNF-α; forward, 5′-AGAATTCCCCATTCAGTAAG-3′ and reverse, 5′-GTGTAAGGAAGTGGTTCAGT-3′ for XAF-1; and forward, 5′-CATTAAGGAGAAGCTGTGCT-3′ and reverse, 5′-GTTGAAGGTAGTTTCGTGGA-3′ for β-actin. RT-qPCR was performed in an ABI PRISM 7000 (Applied Biosystems; Thermo Fisher Scientific, Inc.). Expression levels were normalized to the internal control β-actin, expressed as the fold change compared with the control and calculated using the ∆∆Ct method (25), subsequent to confirm the target PCR product with melting curve analysis.

Western blot analysis

Intratumor or peritumor specimens from HCC patients were homogenized prior to protein extraction. Lysis was then performed with a Cell Lysis and Protein Extraction kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions, followed by addition of protease inhibitor cocktail (Sigma-Aldrich). Proteins were quanitified using the BCA Protein assay reagent kit (Thermo Fisher Scientific, Inc.) and 25 μg of each sample was separated by 10% sodium dodecyl sulfate polyacrylamide gel (Thermo Fisher Scientific, Inc.) electrophoresis and then transferred onto a nitrocellulose membrane (EMD Millipore, Billerica, MA, USA). Non-specific binding was blocked with 2% bovine serum albumin (Ameresco, Inc.) overnight at 4°C, and membranes were subsequently probed with rabbit polyclonal antibody to XAF-1 (Abcam, Cambridge, MA, USA; cat. no. ab81353; 1:500 dilution) β-actin (Abcam; cat. no. ab8227; 1:200 dilution) or TNF-α (Cell Signaling Technology Inc., Danvers, MA, USA; cat. no. 3727; 1:500 dilution) at 4°C overnight. The membrane was finally incubated with goat anti-rabbit horseradish peroxidase-conjugated secondary antibody (Promega Corp., Madison, WI, USA; cat. no. W4011) at 4°C for 2 h, and antibodies were visualized using an enhanced chemiluminescence detection system (GE Healthcare, Little Chalfont, UK) following the manufacturer's instructions. The images of the blots were captured on a UVP BioSpectrum 500 imaging system (UVP, LLC, Upland, CA, USA) and the bands were analyzed using Image J (imagej.nih.gov/ij/). The protein levels of XAF-1 or TNF-α were expressed as a percentage to β-actin.

Apoptosis assay via Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) kit

The apoptosis of MHCC97L cells with or without infection (1 MOI, 24 h) with Ad-Con, Ad-XAF-1, Ad-TNF-α or Ad-XAF-1&TNF-α was examined with an ApoDETECT Annexin V-FITC kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Briefly, MHCC97L cells either without or post-infection were incubated at 37°C for 24 h, and then were harvested and suspended in binding buffer (5×105 cells/ml). The suspended cells were mixed with 5 μl Annexin V-FITC and 10 μl of PI and incubated for 15 min in the dark at room temperature. The stained cells were detected using a FACScan flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA). The results were calculated using the CellQuest™ Pro software (BD Biosciences) and were presented as the percentage of apoptotic cells to total cells.

Cell proliferation assay and colony formation assay

The proliferation of HCC cells was evaluated using a cell counting assay and a colony formation assay. The cell counting assay was performed as follows: Cells (103/ml) were seeded into 12-well plates and then infected with Ad-XAF-1&TNF-α or Ad-con virus at an MOI of 1 or 10 for 2 h, followed by further incubation in medium for 1, 3 or 5 days. The cells were trypsinized and the number of viable cells was counted using a hemocytometer (Reichert, Inc., Depew, NY, USA) following trypan blue (Thermo Fisher Scientific, Inc.) staining. For the colony formation assay, 1,000 cells were seeded into a 12-well plate and infected with Ad-XAF, Ad-TNF-α, Ad-XAF-1&TNF-α or Ad-con virus at an MOI of 1 or 10 for 2 h, followed by incubation in medium for another five days. The cell colonies were stained with 0.5% crystal violet (Sigma-Aldrich) in methanol for 10 min and colonies were counted on the plate by the naked eye.

Statistical analysis

Values are expressed as the mean ± standard error of the mean. Differences between two groups were evaluated using Student's unpaired t-test for the cell viability assay, and the paired-samples t-test was used for comparison of expression levels in the tumor and peritumor tissues or among the cell lines. Statistical analysis was performed using GraphPad Prism software (version 5; GraphPad Inc., La Jolla, CA, USA) and P<0.05 was considered to indicate a statistically significant difference between values.

Results

XAF-1 is downregulated in HCC specimens and cell lines

To confirm the tumor suppressive role of XAF-1 in HCC, its expression was determined in 56 HCC specimens and paired peritumor tissues. As shown in Fig. 1A, compared to the levels in the peritumor tissues, the relative XAF-1 mRNA levels (to β-actin) in the HCC specimens were significantly reduced by ~40% (P<0.01). Furthermore, western blot analysis confirmed a ~50% reduction of XAF-1 protein expression in the HCC group compared with that in the peritumor samples (P<0.001) (Fig. 1B). Furthermore, the expression of XAF-1 in the HCC cell lines MHCC97L, HepG2 and MHCC97H was significantly reduced at the mRNA and protein level compared to that in the L02 human hepatic cell line (P<0.05 or P<0.01) (Fig. 1C and D). Thus, the significant downregulation of XAF-1 in HCC specimens and cell lines was confirmed.

Downregulation of XAF-1 is associated with the degree of malignancy of HCC

To assess the association of the reduced XAF-1 with the malignant characteristics of HCC, the correlation of XAF-1 expression with clinico-pathological features, including tumor size, Barcelona clinic liver cancer (BCLC) stage, tumor -nodes -metastasis (TNM) stage and vascular invasion, was assessed. As shown in Table I, there was no significant difference in age, hepatitis B surface antigen positivity or alpha-fetoprotein levels between the groups with XAF-1 levels <1 and XAF-1 levels ≥1. However, XAF-1 expression was negatively associated with the tumor size, BCLC stage, TMN stage (P<0.05, respectively). However, the association of XAF-1 mRNA levels with vascular invasion was not significant (P>0.05). In conclusion, these results confirmed the association of reduced XAF-1 mRNA levels with the degree of malignancy of HCC.

Construction of adenovirus co-expressing XAF-1 and TNF-α

To further identify the suppressive role of XAF-1 in HCC, an adenovirus co-expressing XAF-1 and TNF-α was constructed. XAF-1 and TNF-α cDNA were amplified by PCR and then linked with a 2A peptide coding sequence (24). The construction strategy of the recombinant adenovirus Ad-XAF-1&TNF-α was illustrated in Fig. 2A. The adenovirus expressing green fluorescence protein (Ad-con), XAF-1 (Ad-XAF-1) or TNF-α (Ad-TNF-α) were used as controls. Each recombinant adenovirus was enveloped via co-transfection of the respective adenoviral genomic plasmid and the shuttle plasmid into BJ5183 bacterial cells. The efficiency of the adenovirus to co-express XAF-1 and TNF-α was evaluated in MHCC97L cells at an MOI of 1 or 10. At 24 h post-infection, the mRNA levels of the two genes were significantly enhanced (P<0.01 or P<0.0001, respectively) (Fig. 2B). Furthermore, western blot analysis indicated that the protein levels of XAF-1 and TNF-α were significantly enhanced by the adenovirus (P<0.01, P<0.001 or P<0.0001) (Fig. 2C and D).

In addition, to compare the effects of Ad-XAF-1&TNF-α with those of XAF-1 or TNF-α alone, MHCC97L cells were infected with Ad-XAF-1 or Ad-TNF-α. As expected, XAF-1 was only overexpressed following infection with Ad-XAF-1, while TNF-α was only overexpressed following infection with Ad-TNF-α at the mRNA and protein level (P<0.001), while Ad-con had no effect (Fig. 3). While XAF-1 as well as TNF-α were significantly overexpressed following infection with Ad-XAF-1&TNF-α (P<0.001), their expression levels were significantly lower than those following infection with the respective mono-overexpression vectors.

In addition, apoptosis in the MHCC97L cells that were infected with 1 MOI Ad-Con, Ad-XAF-1, Ad-TNF-α or Ad-XAF-1&TNF-α was examined. MHCC97L cells without infection served as a blank control. As indicated in Fig. 3D and E), compared with the Ad-Con, Ad-XAF-1 or Ad-TNF-α induced a significantly increased level of apoptosis in MHCC97L cells (P<0.05 or P<0.01). Furthermore, the Ad-XAF-1&TNF-α infection induced more apoptotic cells than the infection with Ad-XAF-1 or Ad-TNF-α (P<0.01). Thus, the co-expression of XAF-1 and TNF-α synergistically induced apoptosis in MHCC97L cells.

Co-expression of XAF-1 and TNF-α inhibits the growth of HCC cells

The present study then investigated the effects of XAF-1 and TNF-α co-expression on the growth of HCC cells. The growth of MHCC97L cells was assessed in vitro using a cell counting assay and a colony formation assay. It was revealed that following infection with Ad-XAF-1&TNF-α, the proliferation of MHCC97L cells was reduced compared with that of the cells infected with Ad-con, Ad-XAF-1 or Ad-TNF-α at either 3 days (P<0.05 or P<0.001) or 5 days (P<0.05, P<0.01 or P<0.001) post-infection, while infection with Ad-XAF-1 or Ad-TNF-α also significantly inhibited the proliferation of MHCC97L cells (P<0.05 or P<0.01) (Fig. 4A and B). Similarly, the colony formation assay showed that the clonogenicity of MHCC97L cells following infection with Ad-XAF-1&TNF-α was significantly reduced compared with that following infection with Ad-XAF-1 or Ad-TNF-α (P<0.05; Fig. 4C and D), while mono-infection still significantly reduced the number of colonies formed (P<0.05 or P<0.01; Fig. 4C and D). These results indicated that co-expression of XAF-1 and TNF-α inhibited the growth of HCC MHCC97L cells more efficiently than either protein alone, even though their co-expression was lower than that following infection with Ad-XAF-1 or Ad-TNF-α.

Discussion

XAF-1 has been identified as a tumor suppressor gene (8) and has been reported to be deregulated in gastric (26), renal (27), pancreatic (28) and esophageal (29) cancers as well as in HCCs (14). The present study reconfirmed the downregulation of XAF-1 in HCCs at the mRNA as well as at the protein level, which was demonstrated in HCC tissues and paired peritumor specimens as well as in cell lines. Of note, the downregulation of XAF-1 was associated with the degree of malignancy of HCC, as a significant correlation of XAF-1 downregulation with the clinico-pathological characteristics of tumor size, BCLC stage and TMN stage was identified. However, the association of XAF-1 mRNA level with the vascular invasion was not significant, possibly due to the small sample size.

XAF-1 has been shown to inhibit the proliferation of lung cancer cells (30), to suppress colon cancer growth and trigger tumor regression (31), and to induce cell apoptosis in gastric and colorectal cancer cell lines; furthermore, XAF-1 was reported to enhance the apoptotic effects of chemotherapeutic drugs and TNF-related apoptosis-inducing ligand (31,32). Recombinant adenoviral vector-mediated XAF-1 overexpression was previously shown to significantly suppress tumor growth in gastric and colon cancer in vitro and in vivo (14,3134). The present study confirmed that the co-expression of XAF-1 and TNF-α by the Ad-XAF-1&TNF-α infection synergistically induced apoptosis in the HCC MHCC97L cells and inhibited the proliferation of HCC cells. To amplify the inhibitory effects of XAF-1 on HCC cell growth, a co-expressing strategy was utilized to overexpress XAF-1 and TNF-α by a singular adenovirus with a 2A peptide linker.

The 2A peptide is a 'self-cleavage' peptide, which is encoded by the foot-and-mouth disease virus. The 2A peptide links two coding sequences in one ORF, which is transcribed into one mRNA molecule, whereas it is translated into two different, function-independent proteins (24). The 'self-cleavage' characteristic of 2A peptide qualifies it to co-express two separate molecules by same vector efficiently (35,36). The present study was the first to constructed an adenovirus, Ad-XAF-1&TNF-α, which co-expressed XAF-1 and TNF-α efficiently. The expression of the two genes was significantly increased at the mRNA as well as the protein level by infection of the Ad-XAF-1&TNF-α into HCC MHCC97L cells. Furthermore, infection with Ad-XAF-1&TNF-α significantly reduced the proliferation and clonogenicity of HCC MHCC97L cells to a greater extent than infection with the Ad-XAF-1 or Ad-TNF-α virus individually. The present study provides a method by which XAF-1 and TNF-α were expressed simultaneously per transcription. The co-expression vector presents an advantage as a potential anti-tumor strategy, as a single administration simultaneously presents two different anti-tumor effectors in the same tumor cell.

In conclusion, the present study was the first to construct an adenovirus which co-expressed XAF-1 and TNF-α in same ORF and expressed them proportionally. This Ad-XAF-1&TNF-α co-expression virus inhibited the proliferation of HCC cells more efficiently than infection with Ad-XAF-1 or Ad-TNF-α alone, suggesting that it may be a promising therapeutic for the treatment of HCC.

Acknowledgments

The present study was supported by a grant from the Baotou Bureau of Science and Technology (grant no. 2012-BT039.

References

1 

Aravalli RN, Steer CJ and Cressman EN: Molecular mechanisms of hepatocellular carcinoma. Hepatology. 48:2047–2063. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Niu J, Lin Y, Guo Z, Niu M and Su C: The epidemiological investigation on the risk factors of hepatocellular carcinoma: A case-control study in Southeast China. Medicine (Baltimore). 95:e27582016. View Article : Google Scholar

3 

Schafer DF and Sorrell MF: Hepatocellular carcinoma. Lancet. 353:1253–1257. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Thorgeirsson SS and Grisham JW: Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet. 31:339–346. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Llovet JM, Burroughs A and Bruix J: Hepatocellular carcinoma. Lancet. 362:1907–1917. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Huang J, Zhang X, Zhang M, Zhu JD, Zhang YL, Lin Y, Wang KS, Qi XF, Zhang Q, Liu GZ, et al: Up-regulation of DLK1 as an imprinted gene could contribute to human hepatocellular carcinoma. Carcinogenesis. 28:1094–1103. 2007. View Article : Google Scholar

7 

Huang J, Zheng DL, Qin FS, Cheng N, Chen H, Wan BB, Wang YP, Xiao HS and Han ZG: Genetic and epigenetic silencing of SCARA5 may contribute to human hepatocellular carcinoma by activating FAK signaling. J Clin Invest. 120:223–241. 2010. View Article : Google Scholar :

8 

Iizuka N, Oka M, Yamada-Okabe H, Mori N, Tamesa T, Okada T, Takemoto N, Tangoku A, Hamada K, Nakayama H, et al: Comparison of gene expression profiles between hepatitis B virus- and hepatitis C virus-infected hepatocellular carcinoma by oligonucleotide microarray data on the basis of a supervised learning method. Cancer Res. 62:3939–3944. 2002.PubMed/NCBI

9 

Okada T, Iizuka N, Yamada-Okabe H, Mori N, Tamesa T, Takemoto N, Tangoku A, Hamada K, Nakayama H, Miyamoto T, et al: Gene expression profile linked to p53 status in hepatitis C virus-related hepatocellular carcinoma. FEBS Lett. 555:583–590. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Oliva J, Bardag-Gorce F, French BA, Li J, McPhaul L, Amidi F, Dedes J, Habibi A, Nguyen S and French SW: Fat10 is an epigenetic marker for liver preneoplasia in a drug-primed mouse model of tumorigenesis. Exp Mol Pathol. 84:102–112. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Schnabl B, Valletta D, Kirovski G and Hellerbrand C: Zinc finger protein 267 is up-regulated in hepatocellular carcinoma and promotes tumor cell proliferation and migration. Exp Mol Pathol. 91:695–701. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Jiang J, Zhang H, Tang Q, Hao B and Shi R: Expression of HIWI in human hepatocellular carcinoma. Cell Biochem Biophys. 61:53–58. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Jain S, Singhal S, Lee P and Xu R: Molecular genetics of hepatocellular neoplasia. Am J Transl Res. 2:105–118. 2010.PubMed/NCBI

14 

Zhu LM, Shi DM, Dai Q, Cheng XJ, Yao WY, Sun PH, Ding Y, Qiao MM, Wu YL, Jiang SH and Tu SP: Tumor suppressor XAF1 induces apoptosis, inhibits angiogenesis and inhibits tumor growth in hepatocellular carcinoma. Oncotarget. 5:5403–5415. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Henry LR, Lee HO, Lee JS, Klein-Szanto A, Watts P, Ross EA, Chen WT and Cheng JD: Clinical implications of fibroblast activation protein in patients with colon cancer. Clin Cancer Res. 13:1736–1741. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Salvesen GS and Duckett CS: IAP proteins: Blocking the road to death's door. Nat Rev Mol Cell Biol. 3:401–410. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Yang YL and Li XM: The IAP family: Endogenous caspase inhibitors with multiple biological activities. Cell Res. 10:169–177. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Ngan CY, Yamamoto H, Seshimo I, Tsujino T, Man-i M, Ikeda JI, Konishi K, Takemasa I, Ikeda M, Sekimoto M, et al: Quantitative evaluation of vimentin expression in tumour stroma of colorectal cancer. Br J Cancer. 96:986–992. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Zhu L, Cheng X, Ding Y, Shi J, Jin H, Wang H, Wu Y, Ye J, Lu Y, Wang TC, et al: Bone marrow-derived myofibroblasts promote colon tumorigenesis through the IL-6/JAK2/STAT3 pathway. Cancer Lett. 343:80–89. 2014. View Article : Google Scholar

20 

Wu WY, Kim H, Zhang CL, Meng XL and Wu ZS: Clinical significance of autophagic protein LC3 levels and its correlation with XIAP expression in hepatocellular carcinoma. Med Oncol. 31:1082014. View Article : Google Scholar : PubMed/NCBI

21 

Pan Q, Liu B, Liu J, Cai R, Liu X and Qian C: Synergistic antitumor activity of XIAP-shRNA and TRAIL expressed by oncolytic adenoviruses in experimental HCC. Acta Oncol. 47:135–144. 2008. View Article : Google Scholar

22 

Plenchette S, Cheung HH, Fong WG, LaCasse EC and Korneluk RG: The role of XAF1 in cancer. Curr Opin Investig Drugs. 8:469–476. 2007.PubMed/NCBI

23 

Leaman DW, Chawla-Sarkar M, Vyas K, Reheman M, Tamai K, Toji S and Borden EC: Identification of X-linked inhibitor of apoptosis-associated factor-1 as an interferon-stimulated gene that augments TRAIL Apo2L-induced apoptosis. J Biol Chem. 277:28504–28511. 2002. View Article : Google Scholar : PubMed/NCBI

24 

Szymczak AL, Workman CJ, Wang Y, Vignali KM, Dilioglou S, Vanin EF and Vignali DA: Correction of multi-gene deficiency in vivo using a single 'self-cleaving' 2A peptide-based retroviral vector. Nat Biotechnol. 22:589–594. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Schmittgen TD and Livak KJ: Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 3:1101–1108. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Wang J, Gu Q, Li M, Zhang W, Yang M, Zou B, Chan S, Qiao L, Jiang B, Tu S, et al: Identification of XAF1 as a novel cell cycle regulator through modulating G(2)/M checkpoint and interaction with checkpoint kinase 1 in gastrointestinal cancer. Carcinogenesis. 30:1507–1516. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Kempkensteffen C, Fritzsche FR, Johannsen M, Weikert S, Hinz S, Dietel M, Riener MO, Moch H, Jung K, Krause H, et al: Down-regulation of the pro-apoptotic XIAP associated factor-1 (XAF1) during progression of clear-cell renal cancer. BMC Cancer. 9:2762009. View Article : Google Scholar : PubMed/NCBI

28 

Huang J, Yao WY, Zhu Q, Tu SP, Yuan F, Wang HF, Zhang YP and Yuan YZ: XAF1 as a prognostic biomarker and therapeutic target in pancreatic cancer. Cancer Sci. 101:559–567. 2010. View Article : Google Scholar

29 

Chen XY, He QY and Guo MZ: XAF1 is frequently methylated in human esophageal cancer. World J Gastroenterol. 18:2844–2849. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Yang WT, Chen DL, Zhang FQ, Xia YC, Zhu RY, Zhou DS and Chen YB: Experimental study on inhibition effects of the XAF1 gene against lung cancer cell proliferation. Asian Pac J Cancer Prev. 15:7825–7829. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Tu SP, Sun YW, Cui JT, Zou B, Lin MC, Gu Q, Jiang SH, Kung HF, Korneluk RG and Wong BC: Tumor suppressor XIAP-Associated factor 1 (XAF1) cooperates with tumor necrosis factor-related apoptosis-inducing ligand to suppress colon cancer growth and trigger tumor regression. Cancer. 116:1252–1263. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Tu SP, Liston P, Cui JT, Lin MC, Jiang XH, Yang Y, Gu Q, Jiang SH, Lum CT, Kung HF, et al: Restoration of XAF1 expression induces apoptosis and inhibits tumor growth in gastric cancer. Int J Cancer. 125:688–697. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Sun PH, Zhu LM, Qiao MM, Zhang YP, Jiang SH, Wu YL and Tu SP: The XAF1 tumor suppressor induces autophagic cell death via upregulation of Beclin-1 and inhibition of Akt pathway. Cancer Lett. 310:170–180. 2011.PubMed/NCBI

34 

Qi R, Gu J, Zhang Z, Yang K, Li B, Fan J, Wang C, He Z, Qiao L, Lin Z and Liu XY: Potent antitumor efficacy of XAF1 delivered by conditionally replicative adenovirus vector via caspase-independent apoptosis. Cancer Gene Ther. 14:82–90. 2007. View Article : Google Scholar

35 

De Giorgi M, Cinti A, Pelikant-Małecka I, Chisci E, Lavitrano M, Giovannoni R and Smolenski RT: Co-expression of functional human heme oxygenase 1, ecto-5′-nucleotidase and ecto-nucleoside triphosphate diphosphohydrolase-1 by 'self-cleaving' 2A peptide system. Plasmid. 79:22–29. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Chng J, Wang T, Nian R, Lau A, Hoi KM, Ho SC, Gagnon P, Bi X and Yang Y: Cleavage efficient 2A peptides for high level monoclonal antibody expression in CHO cells. MAbs. 7:403–412. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 13 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li K, Li X, Wu Z, Zheng L, Cui Y, Wang J, Huang Y and Yan Z: Adenovirus encoding XAF-1 and TNF‑α in the same open reading frame efficiently inhibits hepatocellular cancer cells. Mol Med Rep 13: 5169-5176, 2016
APA
Li, K., Li, X., Wu, Z., Zheng, L., Cui, Y., Wang, J. ... Yan, Z. (2016). Adenovirus encoding XAF-1 and TNF‑α in the same open reading frame efficiently inhibits hepatocellular cancer cells. Molecular Medicine Reports, 13, 5169-5176. https://doi.org/10.3892/mmr.2016.5193
MLA
Li, K., Li, X., Wu, Z., Zheng, L., Cui, Y., Wang, J., Huang, Y., Yan, Z."Adenovirus encoding XAF-1 and TNF‑α in the same open reading frame efficiently inhibits hepatocellular cancer cells". Molecular Medicine Reports 13.6 (2016): 5169-5176.
Chicago
Li, K., Li, X., Wu, Z., Zheng, L., Cui, Y., Wang, J., Huang, Y., Yan, Z."Adenovirus encoding XAF-1 and TNF‑α in the same open reading frame efficiently inhibits hepatocellular cancer cells". Molecular Medicine Reports 13, no. 6 (2016): 5169-5176. https://doi.org/10.3892/mmr.2016.5193