Hepatitis C virus core protein induces hypoxia‑inducible factor 1α-mediated vascular endothelial growth factor expression in Huh7.5.1 cells

  • Authors:
    • Chengliang Zhu
    • Xinghui Liu
    • Shiqun Wang
    • Xiaohong Yan
    • Zhaoming Tang
    • Kailang Wu
    • Yan Li
    • Fang Liu
  • View Affiliations

  • Published online on: March 11, 2014     https://doi.org/10.3892/mmr.2014.2039
  • Pages: 2010-2014
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatitis C virus (HCV) infection is one of the major causes of hepatocellular carcinoma (HCC). It has been demonstrated that the overexpression of angiogenic factors are associated with the maintenance of liver neoplasia. Hypoxia‑inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF) are important regulators of angiogenesis and are important in wound healing, the regeneration of new vessels and reproductive functions. The present study investigated the role of the HCV core protein in the induction of HIF-1α and VEGF expression. The HCV core gene and HIF-1α siRNA were transfected into Huh7.5.1 cells. The results demonstrated that the induction of HCV core gene expression in Huh7.5.1 cells leads to the overexpression and stabilization of HIF-1α, and the activation of HIF-1α leads, in turn, to the stimulation of VEGF, which is one of the most important angiogenic factors. These results provide new information to facilitate the understanding of HCC oncogenesis.

Introduction

It is estimated that 2–3% of the world’s population are chronically infected with the hepatitis C virus (HCV), which is considered to be a major risk factor for the development of hepatocellular carcinoma (HCC) (1,2). HCV belongs to the Flaviviridae family of enveloped RNA viruses and contains a 9.6 kb single-stranded positive-sense RNA genome. This genome is translated into a large polyprotein which is then cleaved by viral and host proteases into structural (core, E1 and E2) and nonstructural (p7, NS2, NS3, NS4A, NS4B, NS5A and NS5B) proteins (35). In addition to their unique involvement in the life cycle and assembly of the virus, these HCV proteins also participate in processes, including transcriptional activation, cell signaling, apoptosis and transformation by way of interaction with host factors (610). In particular, the core gene product has long been proposed as a candidate protein implicated in liver oncogenesis.

Hypoxia-inducible factor 1 (HIF-1) is a heteroduplex that contains a constitutively expressed HIF-1β subunit and an oxygen- and growth factor-regulated HIF-1α subunit (the major determinant for the activity of HIF-1). HIF-1 is the most important regulator of oxygen homeostasis, which is required for cellular metabolism (11). Under physiological and normoxic conditions, HIF-1α is subjected to rapid degradation by ubiquitin-proteasome pathways (12). However, under hypoxic induction or non-hypoxic growth factor induction conditions, HIF-1α is overexpressed and stabilized, resulting in the activation of genes that stimulate angiogenesis, including vascular endothelial growth factor (VEGF) (13). In human cancers, HIF-1 mediated angiogenesis and metabolic adaptation are important in tumor formation, progression and metastasis (14).

Extensive investigations have been conducted to elucidate the inter-relationships between viral products and host cellular factors. Previous studies have demonstrated that HCV infection stabilizes HIF-1α and stimulates the synthesis of VEGF (15). The present study demonstrated that induction of HCV core protein expression in Huh7.5.1 cells enhances the transcriptional level and protein amount of HIF-1α, as well as VEGF, and also confirmed that the HCV core protein increases the expression of VEGF directly via the activation of HIF-1α. Thus, we propose a novel molecular mechanism of the core protein in modulating gene expression that is associated with HCC.

Materials and methods

Plasmids and siRNAs

The plasmid pCMV-Tag2B (Flag2B; Stratagene, La Jolla, CA, USA) was used to construct the HCV core expression plasmid pCMV-Tag2B-core (Flag2B-core). The selection of siRNAs against HIF-1α (HIF-1α siRNA) and negative control siRNA (NC siRNA) were based on the study by Gillespie et al (16).

Cell culture and transfection

The human hepatoma cell line, Huh7.5.1, was cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum, 100 U of penicillin/ml and 100 μg of streptomycin sulfate/ml at 37°C in a humidified 5% CO2 incubator. Transient transfections of Huh7.5.1 cells with the plasmids and siRNAs described above were conducted using Lipofectamine 2000 (Invitrogen Life Technologies, Carlsbad, CA, USA), according to the manufacturer’s instructions.

Reverse transcriptase (RT)-PCR

Following 48 h of transfection, total cellular RNAs were extracted using TRIzol (Invitrogen Life Technologies) and the cDNA was reverse transcribed from 1 μg of total RNA using an oligo (dT) primer. The resulting cDNA was PCR amplified with the following gene-specific primers: HIF-1α, forward 5′-TAGTGCCACATCATCACC-3′ and reverse 5′-ACATGCTAAATCAGAGGG-3′; VEGF, forward 5′-GGGCAGAATCATCACGAAGT-3′ and reverse 5′-GGCTCCAGGGCATTAGACA-3′. PCR amplification was performed under the following conditions: 10 min at 95°C, followed by 35 cycles of 94°C for 15 sec, 55°C for 30 sec and 72°C for 30 sec and finishing with a dissociation protocol. The PCR products were detected by 2% agarose gel electrophoresis and visualized under UV light with ethidium bromide staining.

Western blot analysis

Following 48 h of transfection, cell samples were lysed with Nonidet P-40 lysis buffer [10 mM of Tris-HCl (pH 7.4), 10 mM of NaCl, 3 mM of MgCl2 and 0.5% Nonidet P-40]. The cell lysates were then centrifuged at 3,000 × g for 10 min and the supernatants were used in the assay. Protein samples were separated by 12% SDS-polyacrylamide gel electrophoresis and then transferred onto nitrocellulose membranes. Following the inhibition of non-specific binding sites, western blot analysis was performed using specific antibodies against HIF-1α (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA), VEGF (Sigma, St. Louis, MO, USA), HCV core protein (Affinity Bioreagents, Golden, CO, USA) and, as an internal control, a monoclonal antibody against β-actin (Sigma). Following washing, blots were developed with horseradish peroxidase-labelled goat anti-rabbit IgG, using an enhanced chemiluminescence kit (Amersham Life Sciences, Piscataway, NJ, USA).

Enzyme-linked immunosorbent assay (ELISA) analysis

Following 48 h of transfection, the VEGF concentration in cell supernatants was measured by ELISA, which was performed according to the manufacturer’s instructions (R&D Systems, Minneapolis, MN, USA).

Statistical analysis

SPSS 13.0 software was used for statistical analysis. Values are expressed as the means ± SD. The comparison of two means was performed by t-tests. P<0.05 was considered to indicate a statistically significant difference.

Results

HCV core protein enhances the expression levels of HIF-1α mRNA and protein in Huh7.5.1 cells

HIF-1 is a heterodimeric (HIF-1α coupled with HIF-1β) protein that regulates oxygen homeostasis for cellular metabolism and acts as an inducer of angiogenic factors. Under physiological conditions, HIF-1α is constitutively expressed and degraded, however, under hypoxia or other conditions, HIF-1α is overexpressed and stabilized (11,13). In the present study, the expression levels of HIF-1α mRNA and protein were measured by RT-PCR and western blot analysis, respectively, in Huh7.5.1 cells transfected with the HCV core gene eukaryotic expression vector (Flag2B-core) or the empty vector (Flag2B). The results in Fig. 1A demonstrated a moderate increase of HIF-1α mRNA in HCV core induced Huh7.5.1 cells relative to non-induced cells. The western blot assay demonstrated a significant increase of HIF-1α protein in HCV core induced Huh7.5.1 cells compared with the control (Fig. 1B).

HCV core protein induces the expression and secretion of VEGF in Huh7.5.1 cells

VEGF stimulates angiogenesis and vascular permeability in neoplastic tissues, which means the expression and secretion of VEGF are increased significantly in numerous types of cancer (17,18). In order to investigate whether HCV core gene expression alone can induce the expression and secretion of VEGF, VEGF mRNA and protein levels in Huh7.5.1 cells were measured according to the same instructions used to measure HIF-1α expression levels. An increase in VEGF mRNA (Fig. 2A) and protein expression (Fig. 2B) was identified in HCV core induced Huh7.5.1 cells, indicating that the HCV core protein contributes to the biosynthesis of VEGF.

As VEGF can be secreted into the extracellular media, we further examined the concentrations of VEGF in cell supernatants by ELISA. The supernatant was removed from all wells and a human VEGF ELISA (R&D Systems) was performed on the cell supernatants 48 h post-transfection, as described in the Quantikine human VEGF ELISA instructions. Student’s t-test was used for statistical analysis. The results in Fig. 2C demonstrated that VEGF concentrations in the supernatants of HCV core induced Huh7.5.1 cells were significantly elevated compared with the controls (654.5±43.7 vs 365.9±26.8 pg/ml).

RNA interference disrupts HIF-1α-induced upregulation of VEGF

The present study utilized HIF-1α siRNA that, when transfected into cells, targets HIF-1α mRNA for degradation, thus reducing the expression of HIF-1α RNA and protein. The VEGF mRNA (Fig. 3A) and protein levels (Fig. 3B) in Huh7.5.1 cells cotransfected with Flag2B-core and HIF-1α siRNA were significantly reduced compared with the Flag2B-core plus NC siRNA-transfected cells. The ELISA results in Fig. 3C (389.2±29.6 vs 768.8±47.3 pg/ml) were in line with the results in Fig. 3A and B.

Discussion

HCV infections are associated with the development of HCC, however, the underlying mechanisms by which HCV induces HCC are not well understood. Indeed, although accumulating studies have implicated the specific roles of HCV proteins in the modulation of cell proliferation and pathogenesis, it is unclear which viral gene products are crucial for the establishment of HCC (19,20). It has been demonstrated that the core protein of HCV can induce HCC in transgenic mice by the modulation of cellular gene products, which has brought the core protein to the attention of researchers (21).

The core protein is located at the N-terminal portion of the HCV polyprotein and is highly conserved among various HCV subtypes. Apart from functioning as the building block of the viral nucleocapsid, which is involved in binding and packaging the viral RNA genome, the core protein exhibits pleiotropic roles in numerous activities, including gene transcription, cell proliferation and cell death through interference with the normal functions of an extensive list of cellular proteins (21). In this regard, the present study was undertaken to investigate whether HCV core gene expression is able to trigger angiogenesis, which is pivotal in tumor formation and maintenance. Our results demonstrated that the induction of HCV core protein expression enhances the transcriptional level and amount of HIF-1α as well as VEGF in Huh7.5.1 cells. HIF-1α and VEGF are regulators of angiogenesis and are important in wound healing, the regeneration of new vessels and reproductive functions. Therefore, these results indicated that the HCV core protein is able to stimulate angiogenesis.

The first study of HIF-1α overexpression in human cancer was ~10 years ago. Since then, a large amount of data has been collected demonstrating that HIF-1α overexpression is associated with tumor angiogenesis and increased mortality in cancer of the brain, breast, oropharynx, esophagus, colon, ovary and uterine cervix (2225). Notably, proteins encoded by transforming viruses that cause tumors in humans, including EBV latent membrane protein 1, hepatitis B virus × protein, human papillomavirus E6/E7 proteins and human T-cell leukemia virus Tat protein, also induce HIF-1α activity (2629). Therefore, it is evident that HIF-1α activity represents a fundamental common pathway in cancer pathogenesis.

What are the mechanisms by which the HCV core protein activates HIF-1α? In various types of human cancer, the increased expression of HIF-1α is induced either by intratumoral hypoxia or by genetic alterations affecting key oncogenes and tumor suppressor genes. For example, ras signaling has been demonstrated to be instrumental in hypoxia-induced stabilization of HIF-1α and inactivation of p53 in tumor cells, which contributes to the activation of the angiogenic switch via amplification of normal HIF-1 dependent responses to hypoxia (30). Regarding HCV-induced tumors, the HCV core protein is able to co-operate with the ras oncogene in the transformation of rodent fibroblasts under certain conditions and is able to exert transcriptional repression of the p53 promoter (31). However, we were unable to draw a conclusion regarding the mechanism of activation of HIF-1α by the HCV core protein as there is not enough evidence that HCV can directly activate any oncogene or deactivate any tumor suppressor genes at present. As the complete cell culture systems of HCV are now available, it may be useful to study more aspects of the HCV core protein in order to elucidate the exact mechanisms underlying HCV core protein induction of HIF-1α.

Our results confirmed that the core protein activates HIF-1α, which, in turn, increases the expression of VEGF. At present, VEGF inhibitors are undergoing clinical testing as a strategy for the prevention and treatment of certain malignancies. These findings may prompt worldwide study into the inhibition of HIF-1α, which may be a novel approach to cancer therapy.

In conclusion, the role of HCV in inducing oncogenesis is complicated and awaits further detailed investigation. In the case of the prevention and control of the virus induced tumors, the cellular response factors activated by viral infection warrant further studies. Thus, a mixture of antibodies or inhibitors may be required that target the virus itself and such cellular factors.

Acknowledgements

This study was supported by research grants from the Major State Basic Research Development Program (973 Program; 2012CB518900), the National Clinical Key Subject (no. 2010305), the National Science Foundation of China (no. 81101485 to Zhu CL, no. 31270206 to Wu KL), the Open Research Program of the State Key Laboratory of Virology of China (no. 2011009, 2012007, 2013004) and the China Postdoctoral Foundation (no. 201104485).

References

1 

Raimondi S, Bruno S, Mondelli MU and Maisonneuve P: Hepatitis C virus genotype 1b as a risk factor for hepatocellular carcinoma development: a meta-analysis. J Hepatol. 50:1142–1154. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Yotsuyanagi H, Koike K, Yasuda K, et al: Hepatitis C virus genotypes and development of hepatocellular carcinoma. Cancer. 76:1352–1355. 1995. View Article : Google Scholar : PubMed/NCBI

3 

De Francesco R: Molecular virology of the hepatitis C virus. J Hepatol. 31(Suppl 1): 47–53. 1999.

4 

Rosenberg S: Recent advances in the molecular biology of hepatitis C virus. J Mol Biol. 313:451–464. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Suzuki R, Suzuki T, Ishii K, Matsuura Y and Miyamura T: Processing and functions of Hepatitis C virus proteins. Intervirology. 42:145–152. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Duong FH, Filipowicz M, Tripodi M, La Monica N and Heim MH: Hepatitis C virus inhibits interferon signaling through up-regulation of protein phosphatase 2A. Gastroenterology. 126:263–277. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Jones DM, Patel AH, Targett-Adams P and McLauchlan J: The hepatitis C virus NS4B protein can trans-complement viral RNA replication and modulates production of infectious virus. J Virol. 83:2163–2177. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Park CY, Jun HJ, Wakita T, Cheong JH and Hwang SB: Hepatitis C virus nonstructural 4B protein modulates sterol regulatory element-binding protein signaling via the AKT pathway. J Biol Chem. 284:9237–9246. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Wagoner J, Austin M, Green J, et al: Regulation of CXCL-8 (interleukin-8) induction by double-stranded RNA signaling pathways during hepatitis C virus infection. J Virol. 81:309–318. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Yi M, Ma Y, Yates J and Lemon SM: Trans-complementation of an NS2 defect in a late step in hepatitis C virus (HCV) particle assembly and maturation. PLoS Pathog. 5:e10004032009. View Article : Google Scholar : PubMed/NCBI

11 

Semenza GL: Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology (Bethesda). 24:97–106. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Roos-Mattjus P and Sistonen L: The ubiquitin-proteasome pathway. Ann Med. 36:285–295. 2004. View Article : Google Scholar

13 

Shemirani B and Crowe DL: Hypoxic induction of HIF-1alpha and VEGF expression in head and neck squamous cell carcinoma lines is mediated by stress activated protein kinases. Oral Oncol. 38:251–257. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Shi YH and Fang WG: Hypoxia-inducible factor-1 in tumour angiogenesis. World J Gastroenterol. 10:1082–1087. 2004.PubMed/NCBI

15 

Nasimuzzaman M, Waris G, Mikolon D, Stupack DG and Siddiqui A: Hepatitis C virus stabilizes hypoxia-inducible factor 1alpha and stimulates the synthesis of vascular endothelial growth factor. J Virol. 81:10249–10257. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Gillespie DL, Flynn JR, Ragel BT, et al: Silencing of HIF-1alpha by RNA interference in human glioma cells in vitro and in vivo. Methods Mol Biol. 487:283–301. 2009.PubMed/NCBI

17 

Katoh R, Miyagi E, Kawaoi A, et al: Expression of vascular endothelial growth factor (VEGF) in human thyroid neoplasms. Hum Pathol. 30:891–897. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Machein MR and Plate KH: VEGF in brain tumors. J Neurooncol. 50:109–120. 2000. View Article : Google Scholar

19 

Banerjee A, Ray RB and Ray R: Oncogenic potential of hepatitis C virus proteins. Viruses. 2:2108–2133. 2010. View Article : Google Scholar : PubMed/NCBI

20 

McGivern DR and Lemon SM: Virus-specific mechanisms of carcinogenesis in hepatitis C virus associated liver cancer. Oncogene. 30:1969–1983. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Moriya K, Fujie H, Shintani Y, et al: The core protein of hepatitis C virus induces hepatocellular carcinoma in transgenic mice. Nat Med. 4:1065–1067. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Chun SY, Johnson C, Washburn JG, Cruz-Correa MR, Dang DT and Dang LH: Oncogenic KRAS modulates mitochondrial metabolism in human colon cancer cells by inducing HIF-1alpha and HIF-2alpha target genes. Mol Cancer. 9:2932010. View Article : Google Scholar : PubMed/NCBI

23 

Doronkin S, Djagaeva I, Nagle ME, Reiter LT and Seagroves TN: Dose-dependent modulation of HIF-1alpha/sima controls the rate of cell migration and invasion in Drosophila ovary border cells. Oncogene. 29:1123–1134. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Talks KL, Turley H, Gatter KC, et al: The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol. 157:411–421. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Wong C, Wellman TL and Lounsbury KM: VEGF and HIF-1alpha expression are increased in advanced stages of epithelial ovarian cancer. Gynecol Oncol. 91:513–517. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Moon EJ, Jeong CH, Jeong JW, et al: Hepatitis B virus × protein induces angiogenesis by stabilizing hypoxia-inducible factor-1alpha. FASEB J. 18:382–384. 2004.

27 

Tomita M, Semenza GL, Michiels C, et al: Activation of hypoxia-inducible factor 1 in human T-cell leukaemia virus type 1-infected cell lines and primary adult T-cell leukaemia cells. Biochem J. 406:317–323. 2007. View Article : Google Scholar

28 

Wakisaka N, Kondo S, Yoshizaki T, Murono S, Furukawa M and Pagano JS: Epstein-Barr virus latent membrane protein 1 induces synthesis of hypoxia-inducible factor 1 alpha. Mol Cell Biol. 24:5223–5234. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Zhang EY and Tang XD: Human papillomavirus type 16/18 oncoproteins: potential therapeutic targets in non-smoking associated lung cancer. Asian Pac J Cancer Prev. 13:5363–5369. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Markert EK, Levine AJ and Vazquez A: Proliferation and tissue remodeling in cancer: the hallmarks revisited. Cell Death Dis. 3:e3972012. View Article : Google Scholar : PubMed/NCBI

31 

Smirnova IS, Aksenov ND, Kashuba EV, et al: Hepatitis C virus core protein transforms murine fibroblasts by promoting genomic instability. Cell Oncol. 28:177–190. 2006.PubMed/NCBI

Related Articles

Journal Cover

May-2014
Volume 9 Issue 5

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
Zhu C, Liu X, Wang S, Yan X, Tang Z, Wu K, Li Y and Liu F: Hepatitis C virus core protein induces hypoxia‑inducible factor 1α-mediated vascular endothelial growth factor expression in Huh7.5.1 cells. Mol Med Rep 9: 2010-2014, 2014
APA
Zhu, C., Liu, X., Wang, S., Yan, X., Tang, Z., Wu, K. ... Liu, F. (2014). Hepatitis C virus core protein induces hypoxia‑inducible factor 1α-mediated vascular endothelial growth factor expression in Huh7.5.1 cells. Molecular Medicine Reports, 9, 2010-2014. https://doi.org/10.3892/mmr.2014.2039
MLA
Zhu, C., Liu, X., Wang, S., Yan, X., Tang, Z., Wu, K., Li, Y., Liu, F."Hepatitis C virus core protein induces hypoxia‑inducible factor 1α-mediated vascular endothelial growth factor expression in Huh7.5.1 cells". Molecular Medicine Reports 9.5 (2014): 2010-2014.
Chicago
Zhu, C., Liu, X., Wang, S., Yan, X., Tang, Z., Wu, K., Li, Y., Liu, F."Hepatitis C virus core protein induces hypoxia‑inducible factor 1α-mediated vascular endothelial growth factor expression in Huh7.5.1 cells". Molecular Medicine Reports 9, no. 5 (2014): 2010-2014. https://doi.org/10.3892/mmr.2014.2039