Viral Load Analysis of Hepatitis C Virus in Huh7.5 Cell Culture System

authors:

avatar Roghayeh Teimourpour 1 , avatar Zahra Meshkat 1 , * , avatar Aida Gholoubi 2 , avatar Hosein Nomani 1 , avatar Sina Rostami 3

Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, IR Iran
Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
Department of Biology, Faculty of Sciences, Ferdowsi University of Mashhad, Mashhad, IR Iran

how to cite: Teimourpour R, Meshkat Z, Gholoubi A, Nomani H, Rostami S. Viral Load Analysis of Hepatitis C Virus in Huh7.5 Cell Culture System. Jundishapur J Microbiol. 2015;8(5):e19279. https://doi.org/10.5812/jjm.8(5)2015.19279.

Abstract

Background:

Previous studies using cell culture systems for the replication of hepatitis C virus have opened new research dimensions, and paved the ways for further and detailed studies of the virus in vitro.

Objectives:

The purpose of the present study was to cultivate hepatitis C virus in a cell culture system and evaluate viral amplification.

Materials and Methods:

In order to propagate hepatitis C virus, cloned whole genome of virus, JFH-1, was used. JFH-1 cDNA was introduced into strain JM109 of Escherichia coli and plasmid, containing the viral genome was purified from transformed bacteria. After XbaI digestion, RNA synthesis was induced using T7 RNA polymerase enzyme. Next, eukaryotic cell line Huh 7.5 was transfected by the purified RNA. Finally, Huh-7.5 cell line was infected with replicated virus and viral load was determined using real-time PCR (Polymerase Chain Reaction).

Results:

The amount of viral load, which was measured using real-time PCR was 17592 IU/mL.

Conclusions:

In the present study, using cell culture, a high titer (in acceptable range) of infectious hepatitis C virus was produced. This method could be used in future studies.

1. Background

Hepatitis C virus (HCV) is a single stranded RNA virus belonging to Flaviviridae family. HCV is a global health problem with an estimated of 3% infection of the world’s population. It was first discovered in 1989 and initially known as non-A, non-B hepatitis (NANBH). This virus primarily affects the liver and causes several severe diseases such as chronic hepatitis, cirrhosis, and hepatocellular carcinoma in human. The virus may transmit via intravenous drug usage, blood transfusion, organ transplantations, sexual contacts, and vertical transmission. The combination of pegylated IFN-α and ribavirin, with either boceprevir or telaprevir in some cases are administered to patients (1-5). Despite many studies related to this virus, no effective vaccine has been developed yet. This is partly due to the lack of suitable laboratory animal models and cell culture system that supports the viral replication. Hepatocytes and lymphocytes are permissive to HCV infection in vivo. However, in vitro condition, the virus is capable of primarily infecting these cells but cannot complete its propagation (6-8). Normally, this virus cannot replicate in cell culture systems. Only some limited HCV isolates such as H77 (genotype 1a), CG1b (genotype 1b), and JFH1 J6 (genotype 2a) are capable of replicating in Huh-7 cells.

JFH1 is a strain of HCV that was originally isolated from a Japanese patient with fulminant hepatitis. Transfection of the JFH1 genomic RNA into Huh-7 cell line leads to the production of HCV particles that are infectious in chimpanzee and can also replicate in vitro without adaptive mutations, thus producing infectious particles (8-11). Indeed, the establishment of an effective cell culture system for various genotypes and strains of HCV provides a valuable tool for studying the pathogenesis of HCV, and development of new antiviral agents and vaccines against this virus. In the present study, JFH1 isolate of HCV was cultured in Huh-7.5 cells. Subsequently, viral load of infectious viruses was evaluated by real-time PCR.

2. Objectives

The present study aimed to evaluate the multiplication of hepatitis C virus in a cell culture system and determination of its viral load.

3. Materials and Methods

3.1. Cells

Huh-7.5 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (Invitrogen, Canada), 0.2% sodium bicarbonate (Sigma, USA), 100 U/mL penicillin (Gibco, UK), and 100 µg/mL streptomycin (Gibco, UK) at 37°C in a 5% CO2 atmosphere.

3.2. Plasmid Preparation

JFH1 vector was transferred into Escherichia coli strain JM109 and the plasmid was purified from an overnight culture of recombinant E. coli by using QIAprep Miniprep Kit (Qiagen, USA). The quality and concentration of purified plasmid DNA were controlled using agarose gel electrophoresis together with measuring Optical Density (OD) of DNA at wave lengths of 260 nm and 280 nm.

3.3. DNA Digestion and RNA Synthesis

Double stranded linear DNA with blunt ends was used as a template for in vitro transcription. About 40 µg of JFH1plasmid was digested by Xba1 restriction enzyme (Fermentas, Germany). Mung bean nuclease was used to obtain blunt ends in digested plasmids. To eliminate the enzyme, phenol/chloroform protocol was applied. Briefly, 400 µL of a 1:1 phenol/chloroform mixture was added to the sample. After mixture, it was centrifuged at14000 rpm for 5 minutes. Aqueous phase was transferred to a new tube. Equal volume of chloroform was added, mixed and centrifuged at14000 rpm for 5 minutes. Then, aqueous phase was transferred into a new tube and equal volume of isopropanol was added. The tube was incubated for 30 minutes at 20°C. After incubation, it was centrifuged at14000 rpm for 45 minute at 4°C. Pellet was washed in 300 µL of 70% cold ethanol by centrifugation at 14000rpm for 10 minutes. Ethanol was discarded and pellet was air dried. The final pellet was dissolved in 20 μL of RNase-free water.

For RNA synthesis, transcription mixture consisted of 5 μL of Transcription buffer (5X), 5 μL of 10 mM ATP/GTP/CTP/UTP Mixture, 1 μg of linearized template DNA, 0.5 μL of (20U) RNase inhibitor, 1.5 μL of (30U) T7 Polymerase, and RNase-free water up to 25 μL. The mixture underwent overnight incubation at 37°C. Following incubation, to eliminate the template DNA, 2U of DNase I was added to the mixture and was incubated at 37°C for 15 minutes. To inactivate DNase enzyme, 2 μL of 0.5 M EDTA (pH 8.0) was added and the mixture was incubated at 65°C for 10 minutes. The final concentration of synthesized RNA was determined by gel electrophoresis and OD measurement.

3.4. RNA Transfection

One day before transfection, 3 × 105 Huh-7.5 cells in 2 mL of DMEM media were cultured in a 6-well plate and incubated at 37°C under 5% CO2 condition. After reaching 80% confluency, the growth medium was removed from cells and washed for one time with 2 mL of Opti-media (Invitrogen, Canada). In the next step, it was incubated at 37°C under 5% CO2 condition for 30 minute. The transfection mixture was prepared in DMRIE-C (Invitrogen, Canada) according to the manufacturer’s instructions. About 48 hours after transfection, culture media was collected for the following steps.

3.5. Infection of the Cells

One day before infection, 1 × 104 Huh-7.5 cells were cultured in a 6-well plate as described in the previous section. One milliliter of supernatant from transfected cells was used for the infection of the cells. After 1.5 hours incubation, the cells were kept at 37°C, under 5% CO2 condition for the virus to be adsorbed. Supernatant was discarded and cells were washed 3 times with phosphate buffer saline (PBS). Complete DMEM, containing 10% FBS was added and the cells were incubated for 48 h at 37°C under 5% CO2 condition. After incubation period, cells were washed 3 times for the second time with PBS. Next, fresh DMEM, containing 10% FBS was added and cells were incubated for more than 48 hours. Supernatant of infected cells was collected and analyzed by real-time PCR.

3.6. Viral Load Analysis by Real-Time PCR

The RNA copy number of hepatitis C virus (HCV) in supernatant of infected cells was determined by using HCV quantitative Real Time PCR Kit (Professional Biotech Pvt. Ltd, India). Briefly, viral RNA was isolated from culture media using QIAamp Viral RNA Mini Kit (Qiagen, USA). HCV RNA load was determined by Geno-Sen’s HCV real-time PCR Kit (Professional Biotech Pvt. Ltd, India) according to the manufacturer’s instructions using Rotor Gene (Corbett Research, Australia). The real-time PCR program comprised 15 minutes at 50°C (for cDNA synthesis), 10 minutes at 95°C (for initial denaturation) for 45 cycles, including denaturation of 95°C for 15 seconds, 20 seconds annealing at 55°C and an extension for 15 seconds at 72°C.

4. Results

For viral load analysis, viral RNA was purified form collected supernatant of re-infected Huh-7.5 cells and was measured by real-time PCR. In the final sample, a viral load of 1.76 × 104 IU/mL (r2 = 0.997, Median 1.6 × 104 IU/mL) was detected (Figure 1) compared to the standard concentrations. Sample CT was in conformity with standard 2. This indicates the high accuracy of the test.

Standard Curve Used to Measure the Concentration of HCV RNA in Unknown Samples
Five standards from 2.5 × 103 to 2.5 × 107 IU/mL were used. Type (column 3): standards and sample arrangements; Ct (column 4): Ct value of standards and sample; Given Conc (IU/mL) (column 5): quantification standard concentrations; Calc Conc (IU/mL) (column 6): concentrations of the standards and sample that was measured by Rotor Gene.

5. Discussion

In order to set up a cell culture system, a specific cell line, able to sustain viral replication and a plasmid, containing the virus genome, which can multiply in this cell line are required. HCV can multiply in primary hepatocyte and lymphocyte cells but its replication is not permanent. Huh-7 cells were originated from heptocarcinoma, which originally obtained from a 57-year-old Japanese patient suffering from liver tumor in 1982 (9, 12-16). Although development of the Huh-7 cell line was a great breakthrough in designing a model for studying HCV, these cell lines have some restrictions. Among them, low virus yields and its limited spread are of significance (17).

Other derivatives of this cell line are up to 50 folds more permissive compared to classic Huh-7 cell line because of more spread of the infected viruses. Compared to Huh 7, Huh-7.5 cell line has a point mutation in RIG-1. This is responsible for the increased permissivity of Huh-7.5 to HCV. In fact, replicating form of ssRNA viral genome, dsRNA, of viruses such as HCV can activate RIG-1, which in turn leads to phosphorylation of IFN regulatory factor 3 followed by activation of innate immune responses against the virus. Deficiency in RIG-1 pathway confers resistance to viral multiplication. Also, recent studies showed that high expression levels of CD81 and SR-B or clladin-1 can improve viral multiplication and spread in a cell line (17-21). In a study in 2005, Kato and Wakita (15) reported 2.04 × 103 copies of HCV RNA units per mL. However, in the present study, we obtained HCV RNA levels of 1.76 × 104 IU/mL that is about 8.6 times more than that of reported viral loads in previous studies (9, 15). In other studies, HCV was cultured in vitro and its infectivity was confirmed by neutralization test using anti-CD81 and anti-E2. Also, its genome was detected by RT-PCR, its replication was blocked by interferon, and 50 nm particles were detected by immunoelectron microscopy and density gradient analysis.

Compared to other HCV strains, JFH1 could produce higher titers of virus particles and its production of core antigen was 50 times higher than that of other strains. In addition, JFH1 RNA titer was 1 to 2 logs higher than those of other strains and its infectivity titer was 102 pfu/mL (11, 17, 22). JFH1 causes a typical course of infection in animal models, but recent studies showed that it was attenuated and induced a transient infection in chimpanzee. Therefore, new chimeric strains may be useful for animal model studies. Designing chimeric genomes may provide a new way to increase viral production in cell cultures. However, there is genetic incompatibility between JFH1 and other genomic segments of other genotypes of HCV, which is replaced the same fragment in JFH1 genome. This obstacle can be resolved by using a serial passage of designed chimeras in cell cultures. This procedure leads to the accumulation of adaptive mutations, which increases virus yields (22, 23).

Indeed, cell culture condition may affect viral production. Many studies showed that certain mutations mainly in structural proteins may be important for viral release and final virus production. In some studies, chimeric HCV were constructed based on JFH1 genomes. In these chimeric viruses, JFH1 core to NS2 regions were replaced with those genes of other genotypes. These chimeric viruses replicate efficiently in Huh-7 cells and produce infectious viral particles both in tissue cultures and animal models. There are different methods to quantify viral production. Plaque assays, TCID50, fluorescent focus assay, and protein assay are classic methods that are generally slow, time consuming, and with low sensitivity. Modern methods such as flow cytometry, quantitative polymerase chain reaction (qPCR), and enzyme-linked immunosorbent assay (ELISA) are now used with more sensitivity and less time-consuming. In the present study, we used real-time PCR for the quantification of viral load. This method cannot detect whole viral particles but can measure relatively low copy number of viral genomes. Therefore, real-time PCR is more sensitive in comparison to other methods for the measurement of viral loads (24-26).

In the present study, a cell culture system for HCV was developed and titer of the propagated virus was measured by real-time PCR. The result of real time PCR showed a high titer of the virus. Establishment of this cell culture system can support future studies on new antiviral agents and vaccines against HCV. In addition, it may help us in designing new chimeric viruses for our further studies.

References

  • 1.

    Mandell GL, Bennett JE, Dolin R. Mandell: Mandell, Douglas, and Bennett's Principles and Practice of Infectious Diseases. 7 ed. USA: Elsevier - Health Sciences Division; 2010. p. 2157-78.

  • 2.

    Brochot E, Duverlie G, Castelain S, Morel V, Wychowski C, Dubuisson J, et al. Effect of ribavirin on the hepatitis C virus (JFH-1) and its correlation with interferon sensitivity. Antivir Ther. 2007;12(5):805-13. [PubMed ID: 17713164].

  • 3.

    Casey LC, Lee WM. Hepatitis C virus therapy update 2013. Curr Opin Gastroenterol. 2013;29(3):243-9. [PubMed ID: 23563981]. https://doi.org/10.1097/MOG.0b013e32835ff972.

  • 4.

    Kim CW, Chang KM. Hepatitis C virus: virology and life cycle. Clin Mol Hepatol. 2013;19(1):17-25. [PubMed ID: 23593605]. https://doi.org/10.3350/cmh.2013.19.1.17.

  • 5.

    Alter MJ. Epidemiology of hepatitis C virus infection. World J Gastroenterol. 2007;13(17):2436-41. [PubMed ID: 17552026].

  • 6.

    Buck M. Direct infection and replication of naturally occurring hepatitis C virus genotypes 1, 2, 3 and 4 in normal human hepatocyte cultures. PLoS One. 2008;3(7). ee2660. [PubMed ID: 18628977]. https://doi.org/10.1371/journal.pone.0002660.

  • 7.

    Cribier B, Schmitt C, Bingen A, Kirn A, Keller F. In vitro infection of peripheral blood mononuclear cells by hepatitis C virus. J Gen Virol. 1995;76 ( Pt 10):2485-91. [PubMed ID: 7595352].

  • 8.

    Iacovacci S, Sargiacomo M, Parolini I, Ponzetto A, Peschle C, Carloni G. Replication and multiplication of hepatitis C virus genome in human foetal liver cells. Res Virol. 1993;144(4):275-9. [PubMed ID: 8210708].

  • 9.

    Wakita T. Isolation of JFH-1 strain and development of an HCV infection system. Methods Mol Biol. 2009;510:305-27. [PubMed ID: 19009271]. https://doi.org/10.1007/978-1-59745-394-3_23.

  • 10.

    Kato T, Matsumura T, Heller T, Saito S, Sapp RK, Murthy K, et al. Production of infectious hepatitis C virus of various genotypes in cell cultures. J Virol. 2007;81(9):4405-11. [PubMed ID: 17301131]. https://doi.org/10.1128/JVI.02334-06.

  • 11.

    Zhong J, Gastaminza P, Cheng G, Kapadia S, Kato T, Burton DR, et al. Robust hepatitis C virus infection in vitro. Proc Natl Acad Sci U S A. 2005;102(26):9294-9. [PubMed ID: 15939869]. https://doi.org/10.1073/pnas.0503596102.

  • 12.

    Bartenschlager R, Lohmann V. Novel cell culture systems for the hepatitis C virus. Antiviral Res. 2001;52(1):1-17. [PubMed ID: 11530183].

  • 13.

    Irshad M, Khushboo I, Singh S, Singh S. Hepatitis C virus (HCV): a review of immunological aspects. Int Rev Immunol. 2008;27(6):497-517. [PubMed ID: 19065353]. https://doi.org/10.1080/08830180802432178.

  • 14.

    Tan SL. Hepatitis C Viruses: Genomes and Molecular BiologyLilly Research Laboratories, Indianapolis. USA: Taylor & Francis; 2006.

  • 15.

    Kato T, Wakita T. [Production of infectious hepatitis C virus in cell culture]. Uirusu. 2005;55(2):287-95. [PubMed ID: 16557015].

  • 16.

    Kato N, Ikeda M, Mizutani T, Sugiyama K, Noguchi M, Hirohashi S, et al. Replication of hepatitis C virus in cultured non-neoplastic human hepatocytes. Jpn J Cancer Res. 1996;87(8):787-92. [PubMed ID: 8797883].

  • 17.

    Liu S, Xiao L, Nelson C, Hagedorn CH. A cell culture adapted HCV JFH1 variant that increases viral titers and permits the production of high titer infectious chimeric reporter viruses. PLoS One. 2012;7(9). ee44965. [PubMed ID: 23028707]. https://doi.org/10.1371/journal.pone.0044965.

  • 18.

    Bartenschlager R, Pietschmann T. Efficient hepatitis C virus cell culture system: what a difference the host cell makes. Proc Natl Acad Sci U S A. 2005;102(28):9739-40. [PubMed ID: 15998731]. https://doi.org/10.1073/pnas.0504296102.

  • 19.

    Pietschmann T. Full-length infectious HCV chimeras. Methods Mol Biol. 2009;510:347-59. [PubMed ID: 19009274]. https://doi.org/10.1007/978-1-59745-394-3_26.

  • 20.

    Komorowski M, Kolakowska A, Godzik P, Madalinski K. [HCV replication in Huh-7.5 cell line]. Med Dosw Mikrobiol. 2012;64(3):239-44. [PubMed ID: 23285778].

  • 21.

    Murayama A, Sugiyama N, Yoshimura S, Ishihara-Sugano M, Masaki T, Kim S, et al. A subclone of HuH-7 with enhanced intracellular hepatitis C virus production and evasion of virus related-cell cycle arrest. PLoS One. 2012;7(12). ee52697. [PubMed ID: 23285155]. https://doi.org/10.1371/journal.pone.0052697.

  • 22.

    Kaul A, Woerz I, Meuleman P, Leroux-Roels G, Bartenschlager R. Cell culture adaptation of hepatitis C virus and in vivo viability of an adapted variant. J Virol. 2007;81(23):13168-79. [PubMed ID: 17881454]. https://doi.org/10.1128/JVI.01362-07.

  • 23.

    Bungyoku Y, Shoji I, Makine T, Adachi T, Hayashida K, Nagano-Fujii M, et al. Efficient production of infectious hepatitis C virus with adaptive mutations in cultured hepatoma cells. J Gen Virol. 2009;90(Pt 7):1681-91. [PubMed ID: 19282429]. https://doi.org/10.1099/vir.0.010983-0.

  • 24.

    Flint SJ, Enquist LW, Racaniello VR, Skalka AM. Principles of Virology. 3 ed. USA: ASMscience; 2009. https://doi.org/10.1128/9781555814434.

  • 25.

    Liu X, He N, Fu Z, Duan S, Gao M, Zhang ZF. Plasma Hepatitis C Virus Viral Load Among Hepatitis C Virus Mono-Infected and HCV/HIV Co-Infected Individuals in Yunnan Province,China. Hepat Mon. 2012;12(7):453-9. [PubMed ID: 23008726]. https://doi.org/10.5812/hepatmon.6160.

  • 26.

    Behzadi MA, Ziyaeyan M. Hepatitis C Virus Load in Seropositive Liver and Kidney Transplant Recipients by Quantitative Real-Time PCR Before and After Transplantation. Jundishapur J Microbiol. 2013;6(8). ee7365. https://doi.org/10.5812/jjm.7365.