Open Access

Anti‑HBV effect of interferon‑thymosin α1 recombinant proteins in transgenic Dunaliella salina in vitro and in vivo

  • Authors:
    • Zhihao Zhang
    • Ping He
    • Yan Zhou
    • Xuhua Xie
    • Shuying Feng
    • Changyu Sun
  • View Affiliations

  • Published online on: May 29, 2018     https://doi.org/10.3892/etm.2018.6227
  • Pages: 517-522
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The aim of the present study was to investigate the anti‑hepatitis B virus (HBV) effect of interferon (IFN)‑thymosin α1 (TA1) in a transgenic Dunaliella salina (TDS) system in vitro and in vivo. The toxicity of TDS in the HepG2.2.15 cell line was assessed using an MTT assay. The effect of TDS on the secretion of HBV early antigen (HBeAg) and HBV surface antigen (HBsAg) in culture supernatants was measured using ELISA. In addition, HBV‑DNA was analyzed using quantitative polymerase chain reaction. Drug treatment experiments were performed in vivo on ducks congenitally infected with duck HBV (DHBV). The drug was administered once daily for 21 continuous days. Blood was drawn from all ducks prior to treatment, following treatment for 7, 14 and 21 days, and following drug withdrawal for 5 days. Serum DHBV‑DNA was determined using quantitative PCR. In addition, the histology of duck liver tissues was assessed using hematoxylin and eosin, and orcein staining. The results demonstrated that TDS suppressed cell viability and HBsAg and HBeAg secretion in HepG2.2.15 cells. Furthermore, the treatment index values for HBsAg and HBeAg following TDS treatment were 2.96 and 3.07 respectively, which were greater than those of the IFN‑α treated group. In addition, the DHBV‑infected duck model experiments indicated that serum DHBV‑DNA levels were significantly decreased in the group of TDS (20 g/kg) following treatment for 7, 14 and 21 days compared with the control group. Following withdrawal of the drug for 5 days, the levels of DHBV‑DNA did not relapse in the medium and high dose groups of TDS (10 and 20 g/kg, respectively). Histological analysis of duck liver also demonstrated that TDS and IFN‑α treatment alleviated inflammation and HBsAg signals in duck livers. In conclusion, TDS markedly suppresses HBV replication in vitro and in vivo and its anti‑HBV effect is greater than that of IFN‑α.

Introduction

Hepatitis B virus (HBV) is a hepatotropic enveloped DNA virus, which causes transient and chronic hepatitis B in humans (1). HBV infection has become a primary public health issue that results in liver cirrhosis and the development of hepatocellular carcinoma (2). In China, early in 2012, ~170 million people are chronically infected with HBV (3). However, effective therapy for patients with HBV is limited owing to a long disease course and easy relapse.

At present, two types of drug are utilized for HBV treatment, including nucleoside analogues and interferon (IFN)-α) and its derivatives (4). IFNs are cytokines that exhibit anti-proliferative, antiviral and immunomodulatory activities (5,6). Furthermore, IFN-α serves a key role in the inhibition of viral replication (7). IFN-γ mediates various critical functions by regulating pro-inflammatory, anti-viral and anti-tumor responses (8,9). However, IFNs possess a variety of shortcomings that limit their widespread application, including low response rates, liver decompensation, easy recurrence and numerous side effects, including fever and headache (10). Therefore, enhancing the efficacy of IFN is of vital importance.

The transgenic Dunaliella salina (TDS) system has been widely used as a novel bioreactor for expressing exogenous genes (1113) and has many advantages, including fast growth, low production cost, easy culture, easy transgenic manipulation and a large-scale production of exogenous proteins (1315). More importantly, the exogenous proteins may also be easily purified to meet the demands of safety and efficiency (9).

HepG2.2.15 cells are derived from the human hepatoblastoma cell line HepG2 and are characterized by exhibiting stable HBV expression and replication within the culture system (16). HepG2.2.15 has been frequently used as a cellular source capable of producing HBV in previous studies (17,18).

The multifunctional cytokine, thymosin α1 (TA1) is a polypeptide hormone with multiple bioactivities that is being clinically trialed for the treatment of HBV and hepatitis C virus (19,20). The present study designed a novel fusion interferon (IFN-TA1) combining IFN-α/IFN-γ with TA1 in a TDS system. The aim was to assess the anti-HBV effect of IFN-TA1 in TDS in vitro and in vivo. The HepG2.2.15 cell line and DHBV-infected duck model were utilized to evaluate the anti-HBV activity of IFN-TA1 in TDS.

Materials and methods

Human tissues

Human liver tissues from 13 patients (6 male, 7 female; age, 31–46) with liver hemangioma receiving liver surgery were obtained from September 2011 to May 2014 in the First Affiliated Hospital of Zhengzhou University (Zhengzhou, China). Patients were included in the present study if they exhibited normal biochemical indexes and had no history of hypertension, diabetes, fatty liver disease and other chronic diseases, including liver cirrhosis. Patients were excluded if they received hepatotoxic drugs, smoked and consumed alcohol in the first 3 months prior to surgery. The use of human tissue was approved by the ethics committee of the First Affiliated Hospital of Zhengzhou University (Zhengzhou, China) and all patients gave informed consent prior to enrollment in the present study.

Transformation of IFN-α/IFN-γ fusion gene and TA1 in TDS

The D. salina strain. UTEX-1644, was obtained from the Algae Culture Collection at the University of Texas (Austin, USA) and were grown in modified PKS medium (NaCl 87.7 g/l; MgS04 with 7H20, 1.2 g/l; CaCl2, 0.022 g/l; KN03 1.0 g/l; KH2P04, 0.054 g/l, ferric salt solution 2.0 ml/l including Na2; EDTA with 2H2 0 0.74 g/l; FeCl3, 6H20 0.216 g/l) with a 12-h light-dark cycle under a light intensity of 50 mmol photon m−2s−1 at 26°C for 24 h (21). To amplify the IFN-α, IFN-γ and TA1 gene, the total RNA of human liver tissues was isolated using Trizol (Sigma, St. Louis, MO, USA). The IFN-α/IFN-γ fusion gene was generated using splicing by overlap-polymerase chain reaction (PCR) (22). The IFN-α/IFN-γ fusion gene and TA1 gene were then inserted into pUΩ-GUS using restriction enzyme HaeIII (New England BioLabs, Inc., Ipswich, MA, USA) and T4 DNA ligase (New England BioLabs, Inc., Ipswich, MA, USA) to generate pUΩ-IFN-α/IFN-γ and pUΩ-TA1 novel vectors, respectively. Subsequently, these vectors were co-transformed into D. salina cells using the glass bead method (21). The individual positive TDS colonies were selected using 3 mg/l of phosphinothricin (Hoechst-Roussel AG, Frankfurt, Germany) at 26°C for 5 days and used for further study after 24 h.

Cell culture

HepG2.2.15 cells purchased from American Type Culture Collection (ATCC, Manassas, VA, USA) were incubated in Dulbecco's modified Eagle's medium (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 100 IU/ml penicillin and streptomycin, 380 mg/l antibiotic G-418 sulfate (Promega Corportation, Madison, WI, USA) and 1% L-glutamine at 37°C in 5% CO2.

Cell viability assay

Cells were incubated in a 96-well plate at a density of 5×104 cells per 100 µl at 37°C for 24 h. The cells were then treated with 1,000 IU/ml IFN-α or various concentrations of TDS (0.1, 0.2, 0.4, 0.8, 1.6 and 3.2 mg/ml) at 37°C for 5 days. Untreated cells were utilized as a control. Following treatment, cell viability was measured using an MTT assay as described previously (23). Based on the cell cytotoxicity detected by the MTT assay, the concentrations of TDS (0.4, 0.8 and 1.6 mg/ml) were selected for the following experiments in considerations of the lower toxicity. Furthermore, a treatment index (TI) was used to assess the clinical application prospect of the drug (24): TI <1, toxic, ineffective; TI=1-2, effective, with some toxicity; TI >2, greater effectiveness, with low toxicity.

HBV surface antigen (HBsAg) and HBV early antigen (HBeAg) assay

Viral proteins in the culture medium, HBsAg and HBeAg, from the cells treated with different concentrations of TDS (0.1, 0.2, 0.4, 0.8, 1.6 and 3.2 mg/ml) were measured by using HBeAg (cat. no. KA3288) and HBsAg (cat. no. KA0286) ELISA kits (Abnova, Taipei City, Taiwan) according to the manufacturers' protocols.

Quantification of HBV DNA

HBV DNA was detected in HepG2.2.15 cells treated with IFN-α or TDS (0.8, 1.6 and 3.2 mg/ml) using quantitative PCR. Total DNA was extracted from the cell supernatant using the TIANamp Virus DNA/RNA kit (Tiangen Biotech Co., Ltd., Beijing, China) and Wizard® Genomic DNA Purification kit (Promega Corporation). The quantification of HBV DNA copies was performed using the SYBR green premix reagent (Takara Biotechnology Co., Ltd., Dalian, China). Total DNA (2 µg) was used as the template for each quantitative PCR assay. PCR was performed using an ABI 7900 real-time PCR detector (Applied Biosystems; Thermo Fisher Scientific, Inc.). The thermocycling conditions were as follows: 93°C for 2 min, 10 cycles at 93°C for 45 sec and 55°C for 60 sec, and 30 cycles at 93°C for 30 sec and 55°C for 45 sec. GAPDH served as a control gene. The primers utilized for HBV DNA fragment amplification were as follows: Forward primer, 5′-CCTCTTCATCCTGCTGCT-3′ and reverse primer, 5′-AACTGAAAGCCAAACAGTG-3′. GAPDH primers: Forward primer, 5′-CGGAGTCAACGGATTTGGTCGTAT-3′ and reverse primer, 5′-AGCCTTCTCCATGGTGGTGAAGAC-3′. Assays were repeated in triplicate and mean quantification cycle values were used to calculate the levels of HBV-DNA using the 2−ΔΔCq method (25). The inhibitory rate was calculated using the formula: Inhibitory rate (%)=[DNA copy (C) control-C sample]/C control ×100.

Animals and drug treatment

A total of 3 day-old ducklings (weight, 40–50 g) were purchased from Zhejiang Academy of Agricultural Sciences (Zhejiang, China). The sex of ducklings was not distinguished as prior studies have demonstrated that this does not affect results (26,27). All ducks received ad libitum access to standard diet and water, and housed under controlled conditions (temperature, 28–30°C; humidity 56–70%; and a 24-h light cycle) (26,27). After adaptive maintenance for 3 days, 0.5 ml of blood was obtained from each duck for analysis. Only DHBV-positive ducklings were used for the subsequent experiments. Animal handling protocols were approved by the Animal Ethics Committees of The First Affiliated Hospital of Zhengzhou University (Zhengzhou, China).

Ducks infected with congenital DHBV were randomly divided into 5 groups (each n=6): An IFN-α group treated with IFN-α (4,000 IU/injections, 500 µl/day; Sigma); 3 groups which received TDS via gastric perfusion at different concentrations (5, 10 and 20 g/kg TDS, respectively); and a control group treated with normal saline. The respective treatments were administered once daily for 21 consecutive days. Blood was drawn from all ducks prior to treatment (T0), following 7 (T7), 14 (T14) and 21 (T21) days of treatment and following withdrawal of the drug after 5 days (P5). The serum samples were separated using centrifugation at 7,000 × g for 15 min at 4°C and stored at −80°C. The levels of DHBV DNA in the serum were detected by quantitative PCR using the SYBR Green real-time PCR Master Mix (Takara Biotechnology Co., Ltd.) with the following primers: Forward primer, 5′-GATACTGGAGCCCAAACC-3′ and reverse primer 5′-GGCAGAGGAGGAAGTCAT-3′. GAPDH served as a control gene, forward primer, 5′-CACAGCCACACACGAAGACA-3′ and reverse primer, 5′-CCTTAGCCAGCCCCAGTAGA-3′. The thermocycling conditions were as follows: 95°C for 1 min, 40 cycles including 95°C for 5 sec, 56°C for 5 sec and 72°C for 25 sec and 40°C for 10 sec. The level of HBV-DNA was calculated using the 2−ΔΔCq method (25).

Histological analysis

Following 21 days of drug treatment, ducks were immediately anesthetized with sodium pentobarbital (Sigma; intraperitoneal injection; 150 mg/kg) and subsequently sacrificed by exsanguination. Liver specimens were collected from the ducks and separated into two sections (1×1 cm). One was fixed with 4% buffered formalin and embedded in paraffin for 24 h at room temperature. The samples were then stained with hematoxylin and eosin (H&E) for 15 min at room temperature and examined under an optical microscope.

The second section was also fixed with 4% buffered formalin for 24 h at room temperature and embedded in paraffin for 24 h at room temperature. Then the samples were stained with orcein for 30 min at 37°C and examined under an optical microscope to detect HBsAg.

Statistical analysis

Data are presented as the mean ± standard deviation and each experiment was performed in triplicate. Data were analyzed using SPSS 16.0 (SPSS, Inc., Chicago, IL, USA). Statistically significant differences between two groups were detected using Student's t-test and the comparison of multiple groups was performed using one-way analysis of variance followed by a post-hoc Tukey's test. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of TDS treatment on cell viability and HBV antigen secretion in HepG2.2.15 cells in vitro

The effect of the drug treatment on cell viability and HBV antigen secretion in HepG2.215 cells in vitro was assessed. As presented in Fig. 1A, TDS had a marked inhibitory effect on cell viability, suggesting that TDS produced a cytotoxic effect on HepG2.2.15 cells. In addition, treatment with TDS (0.4, 0.8, 1.6 and 3.2 mg/ml) resulted in a significant reduction of HBsAg (Fig. 1B) and HBeAg secretion (Fig. 1C). The TI values of TDS for HBsAg and HBeAg were 2.96 and 3.07 respectively in HepG2.2.15 cells, indicating that TDS was effective and exhibited low toxicity (Fig. 1D). Furthermore, the TI values of TDS for HBsAg and HBeAg were significantly higher than that of IFN-α, which suggests that TDS may be more effective in clinical application.

Effect of TDS treatment on HBV-DNA load in HepG2.2.15 cell culture medium

To further confirm the anti-HBV activity of TDS, the levels of HBV-DNA in the cell supernatant were assessed. The results indicated that, compared with control group, treatment of TDS (0.8 and 1.6 mg/ml) significantly decreased HBV-DNA levels in the culture medium, with inhibition ratios of 49.3 and 65.7%, respectively (Fig. 2A and B). However, no significant difference was observed in the IFN-α group when compared with the control group. The results revealed that TDS treatment suppresses HBV-DNA replication in HepG2.2.15 cells.

Effect of TDS treatment on duck HBV (DHBV)-DNA levels, inflammation and HBsAg in duck livers

Subsequently, the present study assessed the anti-HBV effect of TDS in vivo. As presented in Fig. 3A, duck serum DHBV-DNA levels were significantly decreased in the TDS group (20 g/kg) following treatment for 7, 14 and 21 days compared with that of the control group. Following treatment for 21 days and withdrawal of the drug for 5 days, the levels of duck serum DHBV-DNA in the TDS groups (10 and 20 g/kg) were significantly reduced when compared with the control group. Following drug withdrawal for 5 days, the levels of DHBV-DNA did not relapse in the TDS groups (10 and 20 g/kg). However, relapse following cessation of TDS was observed in the IFN-α and TDS (5 g/kg) groups.

Histological observation was then performed using H&E and orcein staining. The degree of swelling in liver cells was notable and the expression of infiltrating lymphocytes was positive in the control group. However, these results were alleviated following IFN-α and TDS treatment. In addition, HBsAg was observed as brown granules in the control group. However, a decrease in the signals for HBsAg was observed in IFN-α and TDS groups compared with the control group (Fig. 3B). These results demonstrated that TDS and IFN-α treatment alleviated inflammation and HBsAg in duck livers.

Discussion

IFNs are considered to serve a key role in the control of viral infections (28). A previous study has indicated that interferons suppress HBV replication in transgenic mice that produce a high level of HBV (29). IFN-α has previously been used to treat HBV infections; however, IFN-α treatment generates sustained virological response in a small quantity of patients (30). The incorporation of TA1 into the fusion gene of IFN-α/IFN-γ may be a promising strategy for the development of anti-HBV drugs (31). The present study demonstrated that IFN-TA1 in a TDS model efficiently reduced HBsAg and HBeAg secretion and HBV-DNA replication in HepG2.2.15 cells in vitro. Furthermore, TDS treatment suppressed DHBV-DNA levels, inflammation and HBsAg signals in duck livers in vivo.

The HBV marker is an essential tool for the assessment of HBV infection (32). Following infection, viral replication occurs inside hepatocytes and consequently HBV DNA, and viral proteins, including HBeAg and HBsAg can be easily detected in serum. The levels of these clinical markers are commonly used to evaluate the disease stage of patients (33,34). Therefore, the inhibition of HBV replication may inevitably reduce the secretion of HBeAg and HBsAg (35). In the present study, the HepG2.2.15 cell line, which contains multiple copies of the HBV genome and is capable of secreting HBV virions into the supernatant, was used as in vitro model (16). The results demonstrated that TDS markedly inhibited the levels of HBV DNA and HBsAg and HBeAg in the culture medium of HepG2.2.15 cells. Additionally, TI values for HBsAg and HBeAg were higher in the TDS group than those of the IFN-α group, indicating that TDS treatment enhances the effect of treatment in vitro.

DHBV is closely associated with human HBV in regard to its mode of replication, genomic organization and hepatotropism (36). DHBV in its natural host, the duck, has been used as an animal model in a preclinical study of drugs designed for the treatment of HBV (37). The results of the present study demonstrated that TDS was a potent inhibitor of DHBV replication in ducks congenitally infected with DHBV. In addition, DHBV-DNA levels were markedly reduced in the high dosage TDS group (20 g/kg) following treatment for 7, 14 and 21 days and withdrawal of the drug for 5 days compared with that in the control group. The levels of DHBV-DNA did not relapse in the high and medium dosage groups of TDS (20 and 10 g/kg, respectively) following drug withdrawal for 5 days. However, relapse following cessation of TDS was observed in the low dosage TDS (5 g/kg) and IFN-α groups. Additionally, the histological analysis of duck liver confirmed that TDS and IFN-α treatment alleviated inflammatory and HBsAg signals in duck livers. Therefore, these results revealed that TDS strengthens the anti-DHBV effects in vivo.

In conclusion, the present study demonstrated that TDS, which produces IFN-TA1 recombinant proteins, effectively inhibited HBsAg and HBeAg secretion and HBV-DNA replication in vitro and suppressed DHBV replication and inflammation in vivo. The present results indicate that D. salina may be used as a bioreactor for the production of IFN-TA1 recombinant proteins. In addition, the anti-HBV effect of TDS is greater than that of IFN-α, which may be an effective antiviral medicine in future treatment of HBV.

Acknowledgements

Not applicable.

Funding

The authors are thankful for the financial support from the Key Project of Science and Technology Department in Henan Province (grant no. 152102310045).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

ZZ wrote the paper and performed the research; PH and YZ performed the research; XX analyzed the data; SF and CS designed the research. All authors have read and approved this manuscript.

Ethics approval and consent to participate

Animal handling protocols were approved by the Animal Ethics Committees of The First Affiliated Hospital of Zhengzhou University (Zhengzhou, China).

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Seeger C and Mason WS: Hepatitis B virus biology. Microbiol Mol Biol Rev. 64:51–68. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Choi JG, Chung YH, Kim JA, Jin YJ, Park WH, Lee D, Shim JH, Lee YS, Seo DD, Jang MK, et al: High HBV-DNA titer in surrounding liver rather than in hepatocellular carcinoma tissue predisposes to recurrence after curative surgical resection. J Clin Gastroenterol. 46:413–419. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Yang G, Han M, Chen F, Xu Y, Chen E, Wang X, Liu Y, Sun J, Hou J, Ning Q and Wang Z: Hepatitis B virus genotype B and mutations in basal core promoter and pre-core/core genes associated with acute-on-chronic liver failure: A multicenter cross-sectional study in China. Hepatol Int. 8:508–516. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Mangano C, Squadrito G, Cacciola I, Carpentieri M, Foti G and Raimondo G: Effectiveness of add-on pegylated interferon alfa-2a therapy in a lamivudine-treated patient with chronic hepatitis B. Ann Hepatol. 10:84–87. 2011.PubMed/NCBI

5 

Cho H and Kelsall BL: The role of type I interferons in intestinal infection, homeostasis, and inflammation. Immunol Rev. 260:145–167. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Bandurska K, Król I and Myga-Nowak M: Interferons: Between structure and function. Postepy Hig Med Dosw (Online). 68:428–440. 2014.(In Polish). View Article : Google Scholar : PubMed/NCBI

7 

Pestka S, Krause CD and Walter MR: Interferons, interferon-like cytokines, and their receptors. Immunol Rev. 202:8–32. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Horras CJ, Lamb CL and Mitchell KA: Regulation of hepatocyte fate by interferon-γ. Cytokine Growth Factor Rev. 22:35–43. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Gao B, Wang H, Lafdil F and Feng D: STAT proteins-key regulators of anti-viral responses, inflammation, and tumorigenesis in the liver. J Hepatol. 57:430–441. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Koumbi L: Current and future antiviral drug therapies of hepatitis B chronic infection. World J Hepatol. 7:1030–1040. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Geng D, Wang Y, Wang P, Li W and Sun Y: Stable expression of hepatitis B surface antigen gene in Dunaliella salina (Chlorophyta). J Appl Phycol. 15:451–456. 2003. View Article : Google Scholar

12 

Sun Y, Yang Z, Gao X, Li Q, Zhang Q and Xu Z: Expression of foreign genes in Dunaliella by electroporation. Mol Biotechnol. 30:185–192. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Feng S, Li X, Xu Z and Qi J: Dunaliella salina as a novel host for the production of recombinant proteins. Appl Microbiol Biotechnol. 98:4293–4300. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Wang HH, Yin WB and Hu ZM: Advances in chloroplast engineering. J Genet Genomics. 36:387–398. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Barzegari A, Hejazi MA, Hosseinzadeh N, Eslami S, Mehdizadeh Aghdam E and Hejazi MS: Dunaliella as an attractive candidate for molecular farming. Mol Biol Rep. 37:3427–3430. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Sells MA, Chen ML and Acs G: Production of hepatitis B virus particles in Hep G2 cells transfected with cloned hepatitis B virus DNA. Proc Natl Acad Sci USA. 84:1005–1009. 1987. View Article : Google Scholar : PubMed/NCBI

17 

Zhu X, Xie C, Li YM, Huang ZL, Zhao QY, Hu ZX, Wang PP, Gu YR, Gao ZL and Peng L: TMEM2 inhibits hepatitis B virus infection in HepG2 and HepG2.2.15 cells by activating the JAK-STAT signaling pathway. Cell Death Dis. 7:e22392016. View Article : Google Scholar : PubMed/NCBI

18 

Li L, Lei QS, Zhang SJ, Kong LN and Qin B: Suppression of USP18 potentiates the anti-HBV activity of interferon alpha in HepG2.2.15 cells via JAK/STAT signaling. PLoS One. 11:e01564962016. View Article : Google Scholar : PubMed/NCBI

19 

Romani L, Bistoni F, Montagnoli C, Gaziano R, Bozza S, Bonifazi P, Zelante T, Moretti S, Rasi G, Garaci E and Puccetti P: Thymosin alpha1: An endogenous regulator of inflammation, immunity, and tolerance. Ann N Y Acad Sci. 1112:326–338. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Billich A: Thymosin alpha1. SciClone Pharmaceuticals. Curr Opin Invest Drugs. 3:698–707. 2002.

21 

Feng SY, Xue LX, Liu HT and Lu PJ: Improvement of efficiency of genetic transformation for Dunaliella salina by glass beads method. Mol Biol Rep. 36:1433–1439. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Meng XP, Yang JY, Qiao XL, Liang LX and Shou-Min XI: Optimization of construction of fusion gene-BTRCP-CypA by using SOE-PCR technique. Biotechnology. 23:51–54. 2013.

23 

Pant K, Gupta P, Damania P, Yadav AK, Gupta A, Ashraf A and Venugopal SK: Mineral pitch induces apoptosis and inhibits proliferation via modulating reactive oxygen species in hepatic cancer cells. BMC Complement Altern Med. 16:1482016. View Article : Google Scholar : PubMed/NCBI

24 

Mi S: A research for screening anti-hepatitis B virus drugs with the 2.2.15 cell line. Zhonghua Yi Xue Za Zhi. 72:612–615, 640. 1992.(In Chinese). PubMed/NCBI

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Sari M: Effects of production system and gender on liveweight and body measurements in pekin ducks. Atatürk Üniversitesi Vet Bil Derg. 8:112–121. 2013.

27 

Low HC: Molecular analysis of acute and chronic duck hepatitis B virus (DHBV) infections in ducks. (unpublished PhD thesis). University of Adelaide, School of Molecular and Biomedical Science; Adelaide, South Australia; 2012

28 

Tian Y, Chen WL and Ou JH: Effects of interferon-α/β on HBV replication determined by viral load. PLoS Pathog. 7:e10021592011. View Article : Google Scholar : PubMed/NCBI

29 

Guidotti LG, Morris A, Mendez H, Koch R, Silverman RH, Williams BR and Chisari FV: Interferon-regulated pathways that control hepatitis B virus replication in transgenic mice. J Virol. 76:2617–2621. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Hansen BE, Buster EH, Steyerberg EW, Lesaffre E and Janssen HL: Prediction of the response to peg-interferon-alfa in patients with HBeAg positive chronic hepatitis B using decline of HBV DNA during treatment. J Med Virol. 82:1135–1142. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Lu NF, Huang AL, Zheng RQ, Zhu YB, Xia ZF, Tang N, Yan G, Gao XL and Wu Y: Anti-HBV effect of fusion protein (TA1-IFN) in vitro. Zhonghua Gan Zang Bing Za Zhi. 13:252–254. 2005.(In Chinese). PubMed/NCBI

32 

Cho HJ, Kim JK, Nam JS, Wang HJ, Lee JH, Kim BW, Kim SS, Noh CK, Shin SJ, Lee KM, et al: High circulating microRNA-122 expression is a poor prognostic marker in patients with hepatitis B virus-related hepatocellular carcinoma who undergo radiofrequency ablation. Clin Biochem. 48:1073–1078. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Ganem D and Prince AM: Hepatitis B virus infection-natural history and clinical consequences. N Engl J Med. 350:1118–1129. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Rehermann B and Nascimbeni M: Immunology of hepatitis B virus and hepatitis C virus infection. Nat Rev Immunol. 5:215–229. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Xu WS, Zhao KK, Miao XH, Ni W, Cai X, Zhang RQ and Wang JX: Effect of oxymatrine on the replication cycle of hepatitis B virus in vitro. World J Gastroenterol. 16:2028–2037. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Mason WS, Seal G and Summers J: Virus of Pekin ducks with structural and biological relatedness to human hepatitis B virus. J Virol. 36:829–836. 1980.PubMed/NCBI

37 

Liu Q, Jia R, Wang M, Huang J, Zhu D, Chen S, Yin Z, Wang Y, Chen X and Cheng A: Cloning, expression and purification of duck hepatitis B virus (DHBV) core protein and its use in the development of an indirect ELISA for serologic detection of DHBV infection. Arch Virol. 159:897–904. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 16 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Z, He P, Zhou Y, Xie X, Feng S and Sun C: Anti‑HBV effect of interferon‑thymosin α1 recombinant proteins in transgenic Dunaliella salina in vitro and in vivo. Exp Ther Med 16: 517-522, 2018
APA
Zhang, Z., He, P., Zhou, Y., Xie, X., Feng, S., & Sun, C. (2018). Anti‑HBV effect of interferon‑thymosin α1 recombinant proteins in transgenic Dunaliella salina in vitro and in vivo. Experimental and Therapeutic Medicine, 16, 517-522. https://doi.org/10.3892/etm.2018.6227
MLA
Zhang, Z., He, P., Zhou, Y., Xie, X., Feng, S., Sun, C."Anti‑HBV effect of interferon‑thymosin α1 recombinant proteins in transgenic Dunaliella salina in vitro and in vivo". Experimental and Therapeutic Medicine 16.2 (2018): 517-522.
Chicago
Zhang, Z., He, P., Zhou, Y., Xie, X., Feng, S., Sun, C."Anti‑HBV effect of interferon‑thymosin α1 recombinant proteins in transgenic Dunaliella salina in vitro and in vivo". Experimental and Therapeutic Medicine 16, no. 2 (2018): 517-522. https://doi.org/10.3892/etm.2018.6227