Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Journal of Virology
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Accepted Manuscripts
    • COVID-19 Special Collection
    • Minireviews
    • JVI Classic Spotlights
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections, Retractions
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Publication Fees
    • Ethics Resources and Policies
  • About the Journal
    • About JVI
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Pathogenesis and Immunity

Novel Infectious cDNA Clones of Hepatitis C Virus Genotype 3a (Strain S52) and 4a (Strain ED43): Genetic Analyses and In Vivo Pathogenesis Studies

Judith M. Gottwein, Troels K. H. Scheel, Benoit Callendret, Yi-Ping Li, Heather B. Eccleston, Ronald E. Engle, Sugantha Govindarajan, William Satterfield, Robert H. Purcell, Christopher M. Walker, Jens Bukh
Judith M. Gottwein
1Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Copenhagen University Hospital, Hvidovre, and Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Troels K. H. Scheel
1Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Copenhagen University Hospital, Hvidovre, and Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Benoit Callendret
2The Center for Vaccines and Immunity, Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Yi-Ping Li
1Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Copenhagen University Hospital, Hvidovre, and Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Heather B. Eccleston
2The Center for Vaccines and Immunity, Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ronald E. Engle
3Hepatitis Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sugantha Govindarajan
4Liver Research Laboratory, Rancho Los Amigos Medical Center, Downey, California
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
William Satterfield
5Department of Veterinary Sciences, Michale E. Keeling Center for Comparative Medicine and Research, M. D. Anderson Cancer Center, Bastrop, Texas
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Robert H. Purcell
3Hepatitis Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Christopher M. Walker
2The Center for Vaccines and Immunity, Nationwide Children's Hospital, and Department of Pediatrics, The Ohio State University, Columbus, Ohio
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Jens Bukh
1Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases and Clinical Research Centre, Copenhagen University Hospital, Hvidovre, and Department of International Health, Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
3Hepatitis Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: jbukh@sund.ku.dk jbukh@niaid.nih.gov
DOI: 10.1128/JVI.02667-09
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

ABSTRACT

Previously, RNA transcripts of cDNA clones of hepatitis C virus (HCV) genotypes 1a (strains H77, HCV-1, and HC-TN), 1b (HC-J4, Con1, and HCV-N), and 2a (HC-J6 and JFH1) were found to be infectious in chimpanzees. However, only JFH1 was infectious in human hepatoma Huh7 cells. We performed genetic analysis of HCV genotype 3a (strain S52) and 4a (strain ED43) prototype strains and generated full-length consensus cDNA clones (pS52 and pED43). Transfection of Huh7.5 cells with RNA transcripts of these clones did not yield cells expressing HCV Core. However, intrahepatic transfection of chimpanzees resulted in robust infection with peak HCV RNA titers of ∼5.5 log10 international units (IU)/ml. Genomic consensus sequences recovered from serum at the times of peak viral titers were identical to the sequences of the parental plasmids. Both chimpanzees developed acute hepatitis with elevated liver enzymes and significant necroinflammatory liver changes coinciding with detection of gamma interferon-secreting, intrahepatic T cells. However, the onset and broadness of intrahepatic T-cell responses varied greatly in the two animals, with an early (week 4) multispecific response in the ED43-infected animal (3 weeks before the first evidence of viral control) and a late (week 11) response with limited breadth in the S52-infected animal (without evidence of viral control). Autologous serum neutralizing antibodies were not detected during the acute infection in either animal. Both animals became persistently infected. In conclusion, we generated fully functional infectious cDNA clones of HCV genotypes 3a and 4a. Proof of functionality of all genes might further the development of recombinant cell culture systems for these important genotypes.

Hepatitis C virus (HCV) is a small, enveloped virus with a single-stranded RNA genome, approximately 9.6 kb in length. The genome consists of 5′ and 3′ untranslated regions (UTRs) and a single open reading frame (ORF), encoding structural proteins (Core, E1, and E2), p7, and nonstructural proteins (NS2, NS3, NS4A, NS4B, NS5A, and NS5B) (22). Due to significant genetic heterogeneity, HCV was classified into 7 major genotypes and numerous subtypes, differing >30% and >20%, respectively, at the nucleotide level and at the amino acid level. Strains/isolates differ in 2 to 10% at the nucleotide/amino acid level, and quasispecies typically differ in up to 2% at the nucleotide/amino acid level (70). As a main cause of liver cirrhosis and hepatocellular carcinoma, chronic HCV infection poses a major public health burden. There is no vaccine available, and combination therapy with alpha interferon and ribavirin is characterized by many side effects and contraindications, as well as low efficacy (22). Research on the HCV life cycle and new therapeutics requires well-characterized experimental models and reagents representing the different virus variants.

Chimpanzees, the only animal model of HCV infection mirroring immunopathogenesis and viral persistence observed in human infections (4, 80), can be infected by intravenous inoculation with HCV particles and by intrahepatic transfection with RNA transcripts from full-length HCV cDNA clones. Molecular infectious clones of genotypes 1a (strains H77 [35, 83], HCV-1 [37], HC-TN [63]), 1b (HC-J4 [85], Con1 [6, 44], HCV-N [1]), and 2a (HC-J6 [84] and JFH1 [32, 79]) were developed. Such cDNA clones were used to initiate monoclonal infections in chimpanzees, to study the function of certain genome regions by reverse genetic studies, and to study HCV natural history and protective immunity (4). Furthermore, plasma pools from monoclonally infected chimpanzees were used for virus challenge in studies of vaccines and antivirals in chimpanzees (4) or SCID-uPA mice engrafted with human hepatocytes (49). However, only JFH1 (79, 89) and JFH1-based recombinants with Core-NS2 consensus sequences of prototype isolates of genotypes 1 to 7 (23, 24, 30, 42, 55, 65, 86) were able to induce productive infection of human hepatoma cells. In contrast to the intragenotypic recombinant J6/JFH (42), efficient growth of JFH1 depended on adaptive mutations in vitro and in vivo (32, 33, 62, 90). Also, most intergenotypic recombinants depended on adaptive mutations in vitro (24). Thus, testing of infectivity and pathogenesis of full-length authentic HCV clones depends on the chimpanzee model.

Because different HCV genotypes differ in their biology (69), as well as in their sensitivity to therapeutics (17) and neutralizing antibodies (24, 30, 43, 50, 65), it is of great importance to create research tools for all major genotypes. Thus, it is essential to have functional cDNA clones representing the major HCV genotypes and important subtypes. Genotype 3a is widespread worldwide; in some European countries, it is found in up to 50% of HCV-infected patients; further, it is highly prevalent in several countries in Asia and South America, as well as in Australia (22). Genotype 4a is highly prevalent in the Middle East and many African countries; in Egypt, up to 20% of the population is infected with HCV genotype 4a, making this country a potential site for clinical testing of HCV vaccine candidates (22).

In the present study, we performed a detailed genetic analysis and generated consensus cDNA clones of strains S52 (genotype 3a) and ED43 (genotype 4a). After observing lack of infectivity in Huh7.5 cells, we demonstrated that these cDNA clones were fully functional in chimpanzees. This permitted us to also analyze the host immune responses during the acute infection, including peripheral and intrahepatic T-cell responses.

MATERIALS AND METHODS

Source of HCV strains S52 and ED43.Genotype 3a strain S52 and genotype 4a strain ED43 were derived from challenge plasma pools from chimpanzees (5, 16) which had been experimentally infected with sera from chronically infected patients (8, 11).

Amplification, cloning, and sequence analysis.RNA was extracted from 200 μl of the S52 or ED43 plasma pool using high pure viral nucleic acid kit (Roche) or TRIzol reagent (Invitrogen). cDNA was synthesized with SuperScript II or III (Invitrogen) and random hexamers or specific reverse primers (TAG Copenhagen). After treatment of cDNA with RNase H (Invitrogen) and RNase T1 (Ambion), PCR was carried out with BD Advantage 2 polymerase mix (Clontech) (23); PCR of 3′UTR fragments was carried out with Amplitaq Gold DNA polymerase (Applied Biosystems) (83). Gel-purified amplicons were A tailed with Taq DNA polymerase (Invitrogen), cloned in pCR2.1-TOPO or pCR-XL-TOPO (Invitrogen), and transformed in Top10 chemically competent bacteria (Escherichia coli) (Invitrogen). 3′UTR amplicons were subcloned after restriction digestion as previously described (83). Sequence analysis and determination of consensus sequence were done using Sequencher (Gene Codes Corporation) and BioEdit (freeware). Polyprotein alignments and phylogenetic analysis were done using MEGA4.1 (freeware). HCV sequences used for alignments were from the European and American HCV databases (euHCVdb [http://euhcvdb.ibcp.fr/euHCVdb/ ] and LANL [http://hcv.lanl.gov/ ]). Standard molecular techniques, such as restriction digestion-based cloning and fusion PCR, were used for assembly of consensus sequences; fusion PCR was done with Pfu DNA polymerase (Stratagene).

Sequences of strain S52 were obtained by analysis of four amplicons: amplicon i (nucleotides [nt] 24 to 3396), amplicon ii (nt 3359 to 5186), amplicon iii (nt 5065 to 7596), and amplicon iv (nt 7530 to 9401). These amplicons covered the following amino acids: amplicon i, amino acids (aa) 1 to 1019; amplicon ii, aa 1008 to 1615; amplicon iii, aa 1576 to 2419; and amplicon iv, aa 2398 to 3020 on the polyprotein (all nucleotide and amino acid numbers refer to positions on pS52 with nt 1 being the 1st nucleotide of the 5′UTR and aa 1 being the 1st amino acid of the polyprotein; primer sequences were excluded). Another amplicon (amplicon v) contained the C-terminal NS5B sequence (starting from nt 9339) as well as the 3′UTR variable region, poly(U/UC) region, and the first 16 nucleotides of the conserved X region, and was obtained as previously described (83); this amplicon covered aa 3001 to 3021 of the polyprotein sequence. After subcloning, 5 clones of amplicons i, ii, and iv, 6 clones of amplicon iii, and 15 clones of amplicon v were sequenced to determine the consensus sequence. pS52 was constructed using clones derived from amplicons i to iv, a synthesized 3′UTR sequence (Genscript), and pGEM-9Zf-MOD. pGEM-9Zf-MOD was generated by replacement of the NotI/EcoRI fragment containing the HCV H77 sequence in pCV-H77C (83) by a convenient multiple cloning site (5′-GCG GCC GCA ACT TGT TTA AAC GCG CCT TAA TTA AGG ATT GGC GCG CCA ACC GGA ATT C −3′). In pS52, the NotI site is located immediately upstream of the T7 promoter sequence and the C-terminal XbaI site is located immediately upstream of the AscI site.

For strain ED43, 5′UTR and ORF sequences were obtained by two amplicons, amplicon i (nt 28 to 5631) and amplicon ii (nt 5476 to 9376), which covered aa 1 to 1763 and aa 1713 to 3008, respectively (all nucleotide and amino acid numbers refer to positions on pED43). Another amplicon (amplicon iii), spanning the C-terminal NS5B sequence (starting from nt 9301), the 3′UTR variable region, the poly(U/UC) region, and the first 16 nucleotides of the conserved X region, was obtained as previously described (83); this amplicon covered aa 2988 to 3008. After subcloning, 4 clones of amplicon i, 5 clones of amplicon ii, and 10 clones of amplicon iii were sequenced to determine the consensus sequence. pED43 was constructed by using clones derived from amplicons i to iii inserted into pCV-H77C (83) using NotI and NheI sites, thereby retaining the 3′-terminal sequence of pCV-H77C (83). Endotoxin-free maxipreparations (Qiagen) were prepared, and the HCV sequence was confirmed for pS52 and pED43.

The consensus sequence of the almost complete ORF of S52 or ED43 genomes recovered from sera of infected chimpanzees was determined by direct sequence analysis of PCR amplicons obtained in a reverse transcription (RT) nested PCR procedure (23). Primers used for RT, 1st- and 2nd-round PCR are given in Tables S1 and S2 in the supplemental material. To determine the 5′-terminal sequences of S52 and ED43 from challenge chimpanzee pools and transfected chimpanzees, we used the 5′RACE system for rapid amplification of cDNA ends (version 2.0; Invitrogen) with dC or dA tailing technology, according to the manufacturer's instruction, using S52- and ED43-specific reverse primers. The second PCR products were directly sequenced and/or TA cloned into pCR2.1-TOPO (Invitrogen) for subsequent sequencing.

Generation of RNA transcripts and transfections.Plasmid DNA was linearized with XbaI (New England BioLabs), purified (Wizard SV gel and PCR clean-up system; Promega), and in vitro transcribed with T7 RNA polymerase (Promega) (23). Before generation of RNA transcripts for in vitro transfection, XbaI-digested pED43 with and without adaptive mutations was treated with mung bean nuclease (New England BioLabs) (65). The amount of RNA transcripts was estimated by standard agarose gel electrophoresis.

For in vitro transfections, Huh7.5 cells (gift from Charles M. Rice) were plated at 4 × 105 cells per well of a 6-well plate and maintained as previously described (23). After 12 to 24 h, lipofection complexes (2.5 μg of RNA transcripts and 5 μl of Lipofectamine 2000 [Invitrogen] in 500 μl of Opti-MEM [Invitrogen]) were added to cells for approximately 16 h.

For in vivo transfections, chimpanzees were housed in compliance with relevant guidelines and requirements (52) in a facility fully accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International. Chimpanzees CH5276 and CH5300 were inoculated intrahepatically by a percutaneous procedure (85) with RNA transcribed as described above from a total of 20 μg XbaI-digested and purified pS52 or pED43, respectively.

Monitoring of HCV infection in Huh7.5 cells.Huh7.5 cells were immunostained for HCV Core antigen using the primary mouse anti-HCV Core protein monoclonal antibody (B2) (Anogen, Yes Biotech Laboratories) at 1:200 in phosphate-buffered saline (PBS) with 5% bovine serum albumin and the secondary antibody Alexa Fluor 594-labeled goat anti-mouse IgG (Invitrogen) at 1:500 in PBS containing Tween; cell nuclei were counterstained with Hoechst 33342 (Invitrogen). The presence of HCV-positive cells was evaluated by fluorescence microscopy. The primary antibody was previously shown to recognize S52 and ED43 Core proteins (23, 65).

Monitoring of HCV infection in chimpanzees.Preinfection sera were obtained at weeks −39, −5, −1, and 0 for chimpanzee CH5276 and at weeks −16, −5, −1, and 0 for chimpanzee CH5300; preinfection liver biopsy samples were obtained at week −5 and −1 for both animals. For CH5276, serum and liver biopsy samples were collected weekly during weeks 1 to 32. For CH5300, serum and liver biopsy samples were taken weekly during week 1 to 18 and every 2 weeks during weeks 20 to 32. Thereafter, both animals were monitored monthly until week 54 to determine the final outcome of infection. Serum samples were monitored for HCV RNA (in-house TaqMan quantitative PCR [qPCR] [16] and Monitor test 2.0; Roche Diagnostics), HCV antibodies (enzyme-linked immunosorbent assay ELISA 2.0; Abbott), and alanine aminotransferase (ALT). Liver biopsy samples were examined for necroinflammatory changes (6) and steatosis.

Investigation of autologous HCV neutralizing serum antibodies in S52- and ED43-infected chimpanzees.Neutralization assays were described previously (30, 65). Briefly, heat-inactivated sera from chimpanzee CH5276 were preincubated with ∼25 focus-forming units (FFU) of S52/JFH1T27186,A4550C (24) and heat-inactivated sera from chimpanzee CH5300 were preincubated with ∼115 FFU ED43/JFH1A28196,A3269T (24) for 1 h at 37°C, followed by 3-h incubation on 6 × 103 Huh7.5 cells plated out the day before. After 48-h incubation, cultures were immunostained for HCV NS5A (23). The number of FFU was determined on an ImmunoSpot series 5 UV analyzer (CTL Europe GmbH) with customized software kindly provided to the Copenhagen Hepatitis C Program by Alexey Karulin, Wenji Zhang, and Paul Lehmann. The mean FFU count of 24 negative-control wells (∼5 FFU) was subtracted from FFU counts in experimental wells. Experimental counts obtained by this method were comparable to manual counts, and counts of up to 200 FFU/well were in the linear range of test dilution series. After subtraction of the mean FFU counts in negative-control wells, for chimpanzee CH5276, FFU counts ranged from 26 to 73 FFU/well; for chimpanzee CH5300, the counts ranged from 59 to 146 FFU/well. Percentages of neutralization were obtained by comparison with mean FFU counts from wells, in which the respective virus had been preincubated with serum samples from week −1 or from week 0. For a positive control for neutralization and to establish automated FFU counting, we carried out neutralization of genotype 6a (HK6a/JFH1T1389C,A1590C) virus with serum H06 from a persistently infected patient, patient H (2006, year 29 after infection, genotype 1a) (24) (see Fig. S1 in the supplemental material).

Investigation of cellular immune responses in S52- and ED43-infected chimpanzees.For in vitro stimulation of CD4+/CD8+ T cells, synthetic peptides 20 aa in length, overlapping by 10 residues, and spanning the entire HCV polyprotein were used. Genotype 3a (strain K3a/650; GenBank accession number D28917) peptides were obtained from the NIH Biodefense and Emerging Infections Research Resources Repository, NIAID, NIH. Information regarding these peptides is available at http://www.beiresources.org . The amino acid sequence of the peptide set differed at 177 amino acid positions (5.9%) compared to the sequence encoded by pS52. Nine peptide pools (41 to 72 peptides per pool) were assembled, roughly corresponding to HCV proteins Core/E1 (aa 1 to 383), E2 (aa 384 to 752), p7/NS2 (aa 753 to 1032), NS3-1 (aa 1033 to 1365), NS3-2 (aa 1349 to 1663), NS4A/B (aa 1664 to 1978), NS5A (aa 1979 to 2426), NS5B-1 (aa 2427 to 2752), and NS5B-2 (aa 2735 to 3021). Synthetic peptides spanning the entire polyprotein of HCV genotype 4a (strain ED43; GenBank accession number Y11604) were purchased from Genemed Synthesis. The patient sequence used to design this peptide set differed from the sequence encoded by pED43 by 67 aa (2.2%). Peptides were organized into 9 pools (30 to 40 peptides per pool) that represented Core/E1 (aa 1 to 400), E2 (aa 381 to 760), p7/NS2 (aa 741 to 1040), NS3-1 (aa 1021 to 1360), NS3-2 (aa 1341 to 1670), NS4A/B (aa 1651 to 1990), NS5A (aa 1971 to 2430), NS5B-1 (aa 2411 to 2720), and NS5B-2 (aa 2701 to 3008).

Direct ex vivo gamma interferon (IFN-γ) enzyme-linked immunospot (ELISPOT) assays were used to quantify HCV-specific IFN-γ-secreting cells in peripheral blood mononuclear cells (PBMC) according to the manufacturer's instructions (U-Cytech). PBMC (2 × 105) isolated on Ficoll gradients were plated in triplicate wells of an IFN-γ ELISPOT plate with peptide pools at a final concentration of 1 μg/ml of each peptide in Aim-V lymphocyte medium (Invitrogen) supplemented with 2% human AB serum (Gemini-BioScience) (AimV-2% HS). The plates were incubated at 37°C for 36 h and then developed according to the manufacturer's protocol. The cutoff for positive responses was set at 10 spots above the number of spots in negative-control wells or three times the number of spots in negative-control wells, whichever was higher. At no time did the number of IFN-γ spots in the negative-control wells exceed 20.

For evaluation of intrahepatic immune responses, liver biopsy samples were gently homogenized in PBS with 2% fetal calf serum (FCS). CD8+ T cells were positively isolated using Dynal magnetic beads (Invitrogen). Positive isolation of CD4+ T cells was performed from the remaining CD8+-depleted fraction by the same procedure. Enriched CD8+ and CD4+ T cells were separately stimulated with 6 × 106 freshly isolated autologous irradiated PBMC pulsed with the whole HCV peptide set (1 μg/ml final concentration of each peptide) in medium (RPMI 1640 supplemented with 10% FCS, 50 μg/ml penicillin-streptomycin, and 50 IU/ml of recombinant human interleukin 2). The cells were cultured at 37°C under 7% CO2, and half of the culture medium was replaced every 3 or 4 days with fresh medium. After 3 weeks, the cells were polyclonally stimulated using soluble anti-CD3 antibodies at a final concentration of 0.05 μg/ml and 3 × 106 irradiated human PBMC as feeder cells. Four to five weeks later, CD4+ or CD8+ T cells were plated in duplicate at 5 × 104 cells per well together with 1 × 105 autologous irradiated PBMC pulsed with peptide pools (1 μg/ml final concentration) as antigen-presenting cells in a 36-h IFN-γ ELISPOT assay. The cutoff for positive responses was set as described above for blood responses.

Nucleotide sequence accession numbers.The nucleotide sequences of the S52 consensus sequence, pS52 (infectious clone of S52), ED43 consensus sequence, and pED43 (infectious clone of ED43) have been deposited in the GenBank database with the accession numbers GU814263, GU814264, GU814265, and GU814266, respectively.

RESULTS

Genetic analysis of genotype 3a strain S52.The S52 HCV source was an acute-phase plasma pool from a chimpanzee, experimentally infected with serum from a chronically infected Italian patient (8). In this pool, the HCV RNA titer was 104.3 IU/ml, and the infectivity titer was 103 chimpanzee infectious doses (CID)/ml (5, 16).

The S52 consensus sequence was determined by clonal sequence analysis of five overlapping RT-PCR amplicons, spanning the complete ORF and partial UTRs as described in Materials and Methods. At each nucleotide position, 5 to 15 clones were analyzed. The S52 ORF consisted of 9,063 nucleotides (nt 340 to 9402; all nucleotide positions refer to pS52), encoding a 3,021-aa polyprotein, followed by a single stop codon. Genetic heterogeneity, with at least one of the analyzed clones being different from the S52 consensus sequence, was found at 199 nt (2.2%) and 67 aa (2.2%) positions (Table 1). Compared to the entire polyprotein, a high percentage of amino acid positions with genetic heterogeneity was found in E2, p7, and NS2 (Table 1). The amino acid sequence of E2 hypervariable region 1 (HVR1) was identical among the clones. As seen in Table S3 in the supplemental material, which shows amino acid positions with genetic heterogeneity, there was evidence for 2 different S52 quasispecies populations. For each sequenced clone, differences to the consensus sequence were found on average at 0.5% of positions at the nucleotide level (range, 0.1 to 1.1%), as well as on the amino acid level (range, 0.1 to 1.5%). At nucleotide position 5358, no distinct consensus could be determined, since 3/6 clones had T, and the other 3 clones had C, with T and C encoding the same amino acid.

View this table:
  • View inline
  • View popup
TABLE 1.

Genetic heterogeneity of the S52 virus population from chimpanzee acute-phase plasma pool

In the 5′UTR sequence initially determined for S52 (nt 24 to 339), genetic heterogeneity among the analyzed clones was found at three nucleotide positions (Table 1). In the S52 3′UTR, the first 23 nt of the variable region (nt 9403 to 9425) were identical in all 15 clones (see Fig. S2 in the supplemental material). This sequence was followed by a TTC motif (nt 9426 to 9428) that was present in 13/15 clones. Assuming a variable region of 26 nt (nt 9403 to 9428), the length of the poly(U/UC) region, which could be determined in 3/15 clones, was 108, 111, and 123 nt. The first 16 nucleotides of the 3′UTR X region were identical in all analyzed clones.

Partial sequences were previously determined for the S52 source patient; the partial 5′UTR and complete Core/E1 sequence (nt 58 to 1488) determined from HCV recovered from this patient (7-9) differed from the S52 consensus sequence obtained for the first passage chimpanzee pool at only one nucleotide/amino acid position (in E1). In contrast, the consensus S52 ORF sequence determined in the present study differed from 3 published genotype 3a isolates (55, 64, 81) by 4.8 to 6.5% and 3.6 to 5.8% at the nucleotide and amino acid level, respectively (Table 2). A phylogenetic analysis of the polyprotein of published HCV cDNA clones and representative HCV isolates showed that pS52 clustered with other genotype 3a isolates (Fig. 1).

FIG. 1.
  • Open in new tab
  • Download powerpoint
FIG. 1.

Phylogenetic tree of pS52, pED43, in vivo infectious cDNA clones, and representative isolates of HCV genotypes 1 to 4. GenBank accession numbers are shown (the names of the isolates/clones are shown in parentheses). In vivo infectious cDNA clones are indicated by an asterisk. Multiple polyprotein sequence alignment and neighbor-joining tree analysis using amino acid p-distance model were performed using MEGA4.1. The scale shows the genetic distance in amino acid substitutions per site.

View this table:
  • View inline
  • View popup
TABLE 2.

Comparison of the ORF sequence of S52 with the ORFs of other genotype 3a isolates

Compared to 2 other genotype 3a 5′UTR sequences, GenBank accession numbers D17763 and D28917 (64, 81), the obtained S52 consensus sequence showed differences at one and three nucleotide positions, respectively. A higher degree of variability was seen in the short 3′UTR variable region; a comparison of this region to that of other genotype 3a isolates, D28917 (81), AF009075 (36), D17763 (64), and D85024 and D85025 (82), is shown in Fig. S2 in the supplemental material. A consensus ACACTCC motif (nt 9418 to 9424) was highly conserved. The TG dinucleotide preceding the beginning of the poly(U/UC region) for certain isolates (36, 78) was found in only 1 of 15 S52 clones analyzed. The lengths of the 3′UTR poly(U/UC) tracts were 84, 86, and 110 nt for D85024 (82), D85025 (82), and AF009075 (36), respectively. The S52 consensus sequence of the first 16 nt of the 3′UTR X region was identical to genotype 3a isolates AF009075, D85024, and D85025 (36, 82), as well as to the genotype 1a cDNA clone pCV-H77C (AF011751) (83).

Generation of consensus clone pS52.The consensus full-length cDNA clone pS52 was constructed as described in Materials and Methods. The S52 sequence contained the following three structural elements: (i) 5′UTR, (ii) ORF, and (iii) 3′UTR. The first structural element, the 5′UTR, consisted of 339 nt, in which nt 24 to 339 were the S52 consensus sequence, while nt 1 to 23 were deduced from published genotype 3a 5′UTR sequences (D17763 and D28917) (64, 81). For nt 1, at which G (D28917) and A (D17763) occurred, G was chosen to optimize in vitro transcription. The second structural element, the ORF, consisted of 9,063 nt (nt 340 to 9402) with two coding changes, G1037A and G1913A, in comparison to the S52 consensus sequence. However, at both positions, A encoded by pS52 was present in 2/5 clones analyzed. In addition, in clonal analysis previously performed for a S52 Core-E2 amplicon (50), A was found as consensus at both positions. Noncoding changes compared to the S52 consensus sequence were A639G, A915T, C1488T, G1575A, C1707T, C2655T, C2805T, C3069T, G3792A, T5187C, T7755A, T8469C, and G8745A. The A915T and T7755A changes were introduced to remove consensus XbaI sites at both positions, which were present in all 5 clones analyzed. All other noncoding changes in pS52 were found in at least 1 of the clones covering the respective position. The third structural element, the 3′UTR, consisted of 235 nt (nt 9403 to 9637) and contained three regions, a variable region of 26 nt (nt 9403 to 9428), which was identical to the S52 nucleotide consensus sequence, a poly(U/UC) region of 111 nt (nt 9429 to 9539), chosen from a representative clone, and a conserved X region of 98 nt (nt 9540 to 9637), determined by the sequence of pCV-H77C (83). The X region of 2 genotype 3a isolates (D85024 and D85025 [82]) was identical to that of pCV-H77C (83), whereas genotype 3a isolate AF009075 (36) differed at positions 9594 and 9635. An XbaI site was inserted immediately downstream of the HCV 3′UTR for generation of the HCV 3′ end (83).

Genetic analysis of genotype 4a strain ED43.The ED43 HCV source was an acute-phase challenge plasma pool from a chimpanzee, experimentally infected with serum from a chronically infected Egyptian patient. This plasma pool had an HCV RNA titer of 105.6 IU/ml and an infectivity titer of 105 CID/ml (5, 16). Previously, the complete ORF of the source patient's virus was sequenced by Chamberlain et al. (11) (GenBank accession number Y11604), and the complete 3′ UTR of the patient virus was sequenced by Kolykhalov et al. (43E; Peter Simmonds, personal communication) (36).

In the present study, the consensus sequence of ED43 from the chimpanzee plasma pool was determined by clonal sequence analysis of three overlapping RT-PCR amplicons spanning the complete ORF and partial UTRs as described in Materials and Methods. In agreement with the patient virus sequence (11), the ED43 ORF was found to consist of 9,024 nt (nt 341 to 9364), coding for 3,008 aa, and terminated by two stop codons. Genetic heterogeneity, with at least one of the analyzed clones deviating from the ED43 consensus sequence, was found at 145 nucleotide (1.6%) and 64 amino acid (2.1%) positions (Table 3). Compared to the average for the entire polyprotein, heterogeneity was relatively high in Core, E1, p7, NS2, and NS4A. The nucleotide and amino acid sequences of HVR1 were identical among the clones. Amino acid positions, at which individual clones differed from the ED43 consensus sequence, are shown in Table S4 in the supplemental material. For each sequenced clone, differences to the consensus sequence were found on average at 0.3% (range, 0.1 to 0.5%) at the nucleotide level and 0.4% (range, 0.1 to 0.8%) at the amino acid level. No distinct consensus could be determined at the noncoding positions 1966 (G/A), 1999 (C/T), 3751 (A/G), and 3871 (C/T) where 2/4 clones represented each sequence.

View this table:
  • View inline
  • View popup
TABLE 3.

Genetic heterogeneity of the ED43 virus population from chimpanzee acute-phase plasma pool

In the determined ED43 5′UTR sequences (nt 28 to 340), genetic heterogeneity among 4 clones was found at 6 positions (Table 3). In the ED43 3′UTR, the 36-nt variable region (nt 9365 to 9400) was identical in the 10 clones analyzed (see Fig. S3 in the supplemental material). The exact length of the poly(U/UC) region could be determined in all 10 clones and ranged from 72 to 86 nt. The first 16 nt of the 3′UTR X region were identical in all clones analyzed.

For ED43 derived from the source patient (GenBank accession number Y11604) (11), nucleotides 62 to 340 of the 5′UTR were determined; this sequence differed from the ED43 consensus sequence derived from the chimpanzee plasma pool at 2 positions. The ED43 consensus ORF sequence, determined in the present study, differed at 125 nucleotide (1.4%) and 67 amino acid (2.2%) positions from the patient sequence (Tables 4 and 5). Differences of at least 2.2% at the amino acid level were detected in NS2, NS4B, NS5A, and NS5B. Differences of less than 1% were detected in E1, E2, and NS5A, and notably, the HVR1 sequence of both isolates was identical at the nucleotide and amino acid levels. At aa 2011 (position 39 of NS5A) of the ED43 polyprotein, C was found; C at this position was previously shown to be critical for replication (71). However, in the source patient, W was reported to be present at this position (11). The 3′UTR variable region of the ED43 consensus sequence determined in the present study was identical to the equivalent sequence of the source patient determined by Kolykhalov et al. (GenBank accession number AF009077) (36). In this sequence, the poly(U/UC) region of 46 nt was shorter than we observed for ED43. The consensus sequence of the first 16 nt of the ED43 X region (nt 9482 to 9497) was identical to the equivalent sequence of AF009077.

View this table:
  • View inline
  • View popup
TABLE 4.

Comparison of the ORF sequence of ED43 derived from chimpanzee plasma pool with the ORFs of other genotype 4a isolates at the nucleotide level

View this table:
  • View inline
  • View popup
TABLE 5.

Comparison of the polyprotein sequence of ED43 derived from chimpanzee plasma pool with the polyproteins of other genotype 4a isolates at the amino acid level

From 7 other genotype 4a isolates with reported ORF consensus sequence (19, 76), the ED43 consensus sequence differed at 8.8 to 9.5% and 5.5 to 6.7% at the nucleotide and amino acid levels, respectively (Tables 4 and 5). Phylogenetic analysis showed that the ED43 consensus sequence determined in this study clustered with other genotype 4a isolate sequences (Fig. 1). The obtained ED43 5′UTR consensus sequence differed from a published genotype 4a 5′UTR sequence (GenBank accession number D45193) (20) at only 1 position. High homology was found between the 3′UTR variable region of ED43 and that of several other genotype 4a sequences (20, 77) (see Fig. S3 in the supplemental material). The ED43 variable region was terminated by a TG dinucleotide as reported for other isolates (36, 78). A similar length of the poly(U/UC) region of 82 nt was reported for another genotype 4a isolate (GenBank accession number AF009076). In addition, the first 16 nt of the ED43 X region were identical to the equivalent sequence of this genotype 4a isolate (36), as well as to pCV-H77C (83).

Generation of consensus clone pED43.The consensus full-length cDNA clone pED43 was constructed with the following three structural elements: (i) 5′UTR, (ii) ORF, and (iii) 3′UTR. The first structural element, the 5′UTR, consisted of 340 nt, with nt 28 to 340 having the ED43 consensus sequence, while nt 1 to 27 were derived from D45193 (20); a G was inserted immediately upstream of the 5′UTR to facilitate in vitro transcription. The second structural element, the ORF, consisted of 9024 nt (nt 341 to 9364), encoding the ED43 amino acid consensus sequence. Compared to the ED43 consensus sequence, noncoding changes were A2458G, A2593G, C3988T, A4459C, C4915T, and T5428C; each of these changes was present in 1/4 clones analyzed. For determination of the pED43 sequence at nt 1966 and 1999, at which no distinct consensus was obtained, we used a previously determined consensus sequence (50). The third structural element, the 3′UTR, consisted of 215 nt (nt 9365 to 9579) with a variable region of 36 nt (nt 9365 to 9400) identical to the ED43 consensus sequence, with a representative poly(U/UC) region of 81 nt (nt 9401 to 9481) and with a conserved X region of 98 nt (nt 9482 to 9579) determined by the sequence of pCV-H77C (83). The sequence of the X region in pED43 is identical to that of another genotype 4a isolate (AF009076) (36). An XbaI site was introduced immediately downstream of the HCV 3′ UTR (83).

RNA transcripts of pS52 and pED43 were nonviable in Huh7.5 hepatoma cells.Previously, Huh7.5 cells were shown to be permissive to infection with strain JFH1 (79, 89) and JFH1-based intra- and intergenotypic recombinants, including recombinants with Core-NS2 of S52 and ED43 (23, 24, 30, 42, 55, 65, 86). Therefore, we tested whether full-length S52 and ED43 replicated or led to productive infection of Huh7.5 cultures. Thus, replicate cultures were transfected with RNA transcripts from pS52, pED43, and positive-control pJ6/JFH1 (42). For J6/JFH1, HCV-Core antigen-positive cells were detected after transfection, and viral spread to almost the complete Huh7.5 culture occurred in 4 to 10 days. In contrast, we detected no HCV-Core-positive cells in S52 and ED43 cultures, stained two or three times per week, and monitored for 4 weeks. In total, four independent transfections with RNA transcripts from pS52 and two transfections with pED43 transcripts were analyzed.

We next tested whether selected adaptive mutations, leading to efficient growth of intergenotypic recombinants S52/JFH1 (23) and H77/JFH1 (65, 86) and/or JFH1 (33) in hepatoma cell lines could confer replication capability to the full-length S52. Therefore, we constructed pS52 with single-nucleotide exchanges in different proteins as follows: in p7, T2717G (identified in S52/JFH1 [23]); in NS3, A4097T (identified in H77/JFH1 [65, 86]) or A4549C (identified in S52/JFH1 [23]); and in NS5A, G7171C (identified in S52/JFH1 [23]) or G7621C (identified in JFH1 [33]). Similarly, we introduced two NS2 mutations in pED43 (A2819G and A3269T) that have been shown to confer cell culture viability to ED43/JFH1 (65). However, after transfection of Huh7.5 cells with the respective RNA transcripts, no HCV-Core-positive cells were observed; the ED43A2819G,A3269T culture was monitored for 1 week, and all other cultures were monitored for 4 weeks. Thus, cDNA clones pS52 and pED43, with or without putative adaptive mutations, were apparently not replication competent in Huh7.5 cells, and long-term cultures did not lead to adaptation that yielded infectious particles.

Finally, for comparison, we also showed that previously developed in vivo infectious cDNA clones of genotype 1a (pCV-H77C) (83), 1b (pCV-J4L6S) (85), and 2a (pJ6CF) (84) were nonviable in Huh7.5 cells. After transfection with the respective RNA transcripts, these cultures were monitored for 2 weeks by immunostaining for HCV-Core.

RNA transcripts of pS52 were infectious in vivo.After intrahepatic transfection of chimpanzee CH5276 with pS52 RNA transcripts, the HCV RNA titers increased to peak levels of 105 to 105.5 IU/ml during weeks 6 to 14 (Fig. 2A). The animal became persistently infected with relatively high virus titers at the end of follow-up (week 54). Furthermore, CH5276 developed acute hepatitis with elevated liver enzyme levels. ALT levels of ∼100 U/liter coincided with significant necroinflammatory liver changes, detected during weeks 19 to 32 (Fig. 2A). Analysis of liver biopsy samples showed no evidence of steatosis, which has been reported to be correlated with genotype 3a infection (61).

FIG. 2.
  • Open in new tab
  • Download powerpoint
FIG. 2.

Course of HCV infection and host immune responses in chimpanzee CH5276 following intrahepatic transfection with RNA transcripts of pS52 (genotype 3a). (A) Course of infection. Serum HCV RNA was monitored by in-house TaqMan assay (detection limit of 10 IU/ml) and/or by Roche Monitor test 2.0 (detection limit of 600 IU/ml). At the top of the panel, HCV RNA test results, anti-HCV antibody results, and liver histology scores are shown. Serum samples that were positive by TaqMan and/or by Monitor test 2.0 (solid black rectangles) and serum samples negative by TaqMan (white rectangle) are indicated in the top row. Anti-HCV antibodies were detected by 2nd generation ELISA: +, positive; −, negative. Necroinflammatory liver changes were scored as follows: 0, healthy or normal; 1, mild; 2, mild-moderate; 3, moderate-severe; 4, severe. HCV Monitor titers are indicated by the solid black circles in the graph; samples below the detection limit are shown as not detected (ND). The gray shaded area in the graph shows serum ALT levels (U/liter). The time points of direct sequence analysis of recovered viral genomes are indicated by the two white arrows. (B) Serum neutralizing antibodies. Percent neutralization of JFH1-based intergenotypic recombinants expressing S52 envelope proteins. The serum samples were diluted 1:20; >50% neutralization was considered significant. Values represent the means of three neutralizations; the standard error of the mean (SEM) range was 3 to 13%. Negative values are shown as 0%. For 1:80 serum dilutions, neutralization was <20% at all time points. (C and D) Peripheral and intrahepatic CD4+/CD8+ T-cell responses. The number of IFN-γ-secreting cells after stimulation with a panel of overlapping peptides specific for genotype 3a (strain K3a/650) and spanning the entire HCV polyprotein was determined in ELISPOT assays. PBMC were used directly. Intrahepatic CD4+ and CD8+ T cells were expanded from liver biopsy samples as described in Materials and Methods. The bars represent the total numbers of IFN-γ-secreting CD4+ and CD8+ T cells following stimulation with the different pools, after background subtraction. Cutoff points (dotted lines) were determined for individual experiments as described in Materials and Methods. Results below the cutoff are indicated by bars up to the dotted line. ND, not detectable.

The consensus sequence (nt 296 to 9387) of viral genomes recovered from serum at peak HCV RNA titers (weeks 7 and 10) was identical to pS52. Using the 5′RACE procedure, the 5′-terminal sequence (nt 1 to 350) was determined for week 10 viruses; the consensus sequence was identical to pS52. However, at nt 1, we observed a quasispecies G/A (pS52 had G). Subsequent analysis of the 5′-terminal sequence of S52 from the acute-phase source plasma pool, not previously analyzed, showed that S52 had consensus A at position 1. Thus, it is possible that the heterogeneity at nucleotide position 1 in the transfected animal represented a reversion to the original sequence of strain S52.

The chimpanzee became anti-HCV positive in a commercial enzyme-linked immunosorbent assay (ELISA) test at week 19 postinfection (Fig. 2A). However, chimpanzee CH5276 did not develop significant levels of autologous neutralizing antibodies during the acute infection, since preincubation of S52/JFH1 viral particles (24) with 1:20 and 1:80 dilutions of week 2 to 32 sera did not lead to >50% neutralization of S52/JFH1 infectivity in Huh7.5 cells compared to preincubation with preinfection sera (Fig. 2B).

To further examine the immunopathogenesis of S52 infection, we monitored the occurrence of genotype 3a-specific IFN-γ-secreting CD4+/CD8+ T cells in peripheral blood (Fig. 2C) and liver biopsy (Fig. 2D) samples. Throughout follow-up, PBMC collected from chimpanzee CH5276 did not have any detectable IFN-γ secretion above the background level in ELISPOT assays, after stimulation with 9 different genotype 3a peptide pools (Fig. 2C and Table 6). Intrahepatic IFN-γ-secreting CD4+/CD8+ T cells were studied similarly after in vitro expansion and were detected from week 9 (Fig. 2D and Table 6). An increase in the percentage of IFN-γ-secreting intrahepatic T cells was detected during weeks 11 to 32, several weeks before the occurrence of peak ALT levels; it also preceded the most pronounced necroinflammatory liver changes, which were observed during weeks 19 to 32. Intrahepatic T-cell responses were detected at 14/23 time points tested, with the strongest response at week 22 (Fig. 2D and Table 6). Intrahepatic T-cell responses were characterized by limited breadth; the highest diversity was recorded at week 18, with responses to 5/9 peptide pools (Core/E1, p7/NS2, NS3-2, NS4A/B, and NS5B-1). Stimulation with the NS3-2 peptide pool resulted in the highest response rate (14/23 time points tested) and the highest number of IFN-γ-secreting cells (Table 6).

View this table:
  • View inline
  • View popup
TABLE 6.

Peripheral and intrahepatic CD4+/CD8+ T-cell responses in chimpanzee CH5276

RNA transcripts from pED43 were infectious in vivo.After intrahepatic transfection of chimpanzee CH5300 with pED43 transcripts, HCV RNA titers increased to peak levels of 104.5 to 105.5 IU/ml during weeks 1 to 8 posttransfection (Fig. 3A). During weeks 9 to 12, there was a 100-fold decrease in HCV RNA titers, and the virus titers remained at relatively low levels (102.5 to 104 IU/ml) throughout 54 weeks of follow-up. The course of infection was characterized by a fast onset of acute hepatitis with peak serum ALT levels of 100 to 200 U/liter during weeks 5 to 10 (Fig. 3A). Peak ALT levels coincided with significant necroinflammatory liver changes during weeks 5 to 13. Following week 10, ALT levels decreased to 20 to 70 U/liter, and subsequently, we observed a decrease in the score of liver necroinflammatory changes. There was no evidence of steatosis in liver biopsy samples.

FIG. 3.
  • Open in new tab
  • Download powerpoint
FIG. 3.

Course of HCV infection and host immune responses in chimpanzee CH5300 following intrahepatic transfection with RNA transcripts of pED43 (genotype 4a). (A) See the legend to Fig. 2A. (B) JFH1-based intergenotypic recombinants expressing ED43 envelope proteins were used as described in the legend to Fig. 2B; the SEM range was 1 to 16%. For 1:80 serum dilutions, neutralization was <20% at all time points. (C and D) Peptides specific for genotype 4a (strain ED43) were used as described in the legend to Fig. 2C and D.

The consensus sequence (nt 297 to 9359) of viral genomes recovered from week 1 and 6 serum of chimpanzee CH5300 did not show any changes compared to the pED43 sequence. Using the 5′RACE procedure, the 5′-terminal sequence (nt 1 to 351) was determined for week 6 viruses from transfected chimpanzee CH5300 and from the original chimpanzee plasma pool source sample; both consensus sequences were identical to pED43 with A at position 1. However, the G added before the 5′-terminal A in pED43 to enhance in vitro transcription was not present in viruses recovered from CH5300.

In chimpanzee CH5300, the ELISA test became positive from week 6 posttransfection (Fig. 3A). However, CH5300 did not develop significant levels of autologous neutralizing antibodies during the acute infection (Fig. 3B), since preincubation of ED43/JFH1 particles (65) with 1:20 and 1:80 dilutions of week 2 to 32 sera did not lead to significant neutralization of ED43/JFH1 infectivity in Huh7.5 cells (Fig. 3B).

Intrahepatic CD4+/CD8+ T cells, secreting IFN-γ upon stimulation with HCV genotype 4a peptide pools, were detected at week 4 posttransfection (Fig. 3D), as ALT levels increased and the first evidence of necroinflammatory changes was seen in the liver (Fig. 3A). The strongest intrahepatic T-cell response was observed during weeks 7 to 11, coinciding with significant necroinflammatory liver changes. However, intrahepatic T cells reactive to genotype 4a peptides were detected throughout follow-up. CH5300 mounted a multispecific T-cell response. At 12/21 time points with a T-cell response above the threshold level, more than 4/9 peptide pools induced IFN-γ secretion by intrahepatic T cells (Table 7). The greatest diversity was observed at weeks 4 and 7 (response to 8/9 pools) and at weeks 8, 11, and 15 (response to 7/9 pools). The highest stimulation rates were achieved with the NS5B-2, Core-E1, NS3-2, and NS5A peptide pools (response at 20, 16, 16, and 15 of 25 time points tested, respectively) (Table 7).

View this table:
  • View inline
  • View popup
TABLE 7.

Peripheral and intrahepatic CD4+/CD8+ T cell responses in chimpanezee CH5300

In chimpanzee CH5300, detection of HCV-specific intrahepatic T cells was followed by occurrence of HCV genotype 4a-reactive PBMC at several time points during week 7 to 15 (Fig. 3C and D). At 7 of 24 time points tested, IFN-γ secretion was observed upon stimulation with NS3-2 peptide pools. In addition, reactivity to the NS3-1 pool was seen at weeks 7 and 8 (Table 7).

DISCUSSION

We generated and characterized infectious cDNA clones of important HCV genotypes, genotypes 3a (pS52) and 4a (pED43). Transfection of Huh7.5 cells with RNA transcripts of these clones, with and without adaptive mutations identified in JFH1 (33) and JFH1-based intergenotypic recombinants (23, 65, 86), did not lead to productive HCV infection. In contrast, intrahepatic transfection of two chimpanzees with RNA transcripts of pS52 and pED43 led to robust infection. Even though both animals became persistently infected, the course of infection and of cellular immune responses differed. Infection with S52 was characterized by the late onset of overt hepatitis coinciding with T-cell responses of limited breadth, not able to control viremia. In contrast, infection with ED43 led to early onset of hepatitis and multispecific T-cell responses, resulting in transient control of viremia. Overall, our data showed that the constructed consensus cDNA clones of S52 and ED43 were fully functional in vivo.

To determine the S52 and ED43 consensus sequences, we studied the quasispecies distribution in standardized acute-phase plasma pools from experimentally infected chimpanzees (5). A relatively high degree of genetic heterogeneity was found in genome regions that are generally characterized by significant genetic diversity, such as E1, E2, p7, and NS2 (Tables 1 and 3) (69). In addition, relatively high genetic heterogeneity was found in ED43 Core and NS3 genes, which in general show less genetic diversity (69). In contrast, E2 hypervariable region 1 was not variable among S52 or ED43 clones, respectively. The genetic heterogeneity of S52 in the chimpanzee plasma pool was greater than that of ED43 (Tables 1 and 3), partly due to the occurrence of two different S52 quasispecies subpopulations (see Table S3 in the supplemental material). Different quasispecies subpopulations were previously found in plasma pools of genotype 1b strain J4 (85) and genotype 2a strain J6 (84).

The chimpanzee ED43 consensus sequence determined in this study and the previously determined consensus sequence of the source patient (GenBank accession number Y11604) (11) differed by 2.2% at the amino acid level (Table 5). Interestingly, relatively great variation was found not only in NS2 and NS5A but also in NS4B and NS5B, which are usually less diverse (69). In contrast, E1 and E2 were relatively similar (Table 5). Like other genotype 4a isolates (19, 76), ED43 and Y11604 have a putative interferon sensitivity-determining region (ISDR; aa 2210 to 2245) in NS5A, which is 4 aa shorter than observed for most other genotypes. However, the putative ISDR in ED43 and Y11604 differed in 4 aa (11%) at the N terminus of this region. Studies of the impact of sequence variations in putative ISDR on interferon sensitivity will be facilitated by replicon/cell culture systems with genotype-specific NS5A.

Variation between the polyprotein consensus sequence of S52, originating from a patient in Sardinia, Italy, and genotype 3a isolates from patients in New Zealand (isolate NZL1) (64), Switzerland (isolate 452) (55), and Japan (isolate K3a/650) (81) was found at 3.6 to 5.8% of the amino acid positions (Table 2). Greater variation was observed between sequences of ED43, which originated from Egypt, and several genotype 4a isolates from the Boston area (Massachusetts) (76) as well as one isolate from Spain (19) (5.5 to 6.7% at the amino acid level) (Table 5). In contrast, ED43 was more similar to genotype 4a isolates obtained from other Egyptian patients, based on comparison of Core/E1 and NS5B sequences (58). A high degree of variation between S52 and other genotype 3a isolates, as well as between ED43 and other genotype 4a isolates, was found in genome regions, for which in general relatively great diversity was reported (69); these regions included E1, E2 (especially HVR1), p7, NS2, and NS5A (Tables 2, 4, and 5).

The polyprotein encoded by pS52 had two nonconsensus residues, which were, however, naturally occurring in the S52 pool. The polyprotein encoded by pED43 reflected the consensus sequence. Both clones had a number of noncoding changes compared to the respective consensus ORF sequence; these apparently did not influence the course of infection in chimpanzees, since the course of viremia in these animals was similar to the course of infection previously observed in animals infected with virus from the source pools (5). It should be noted that nt 1 (G) in the 5′UTR of pS52 was nonconsensus (S52 had A) and that there was some evidence that the virus recovered from the transfected animal showed reversion to the consensus A at this position. As previously reported for development of a genotype 1b cDNA clone (85), the conserved 3′ X regions in pS52 and pED43 were derived from pCV-H77C (83) but showed close homology to X regions of other published genotype 3a and 4a sequences. Compared to previously developed cDNA clones of genotypes 1a, 1b, and 2a (1, 6, 32, 35, 37, 63, 79, 83-85) as well as consensus sequences of selected isolates of various genotypes and subtypes, the pS52 sequence formed a distinct cluster with previously published genotype 3a isolate sequences (55, 64, 81), while the pED43 sequence clustered with described genotype 4a isolate sequences (19, 76) (Fig. 1).

Huh7 human hepatoma cells and cell lines derived from Huh7 cells could not be productively infected by transfection of RNA transcripts of previously developed infectious cDNA clones of genotype 1a (pCV-H77C and pHC-TN [63]), genotype 1b (pCV-J4L6S and pFK-Con1 [6]), and genotype 2a (pJ6CF). However, in these cell lines, low levels of virus production were recorded after transfection of RNA transcripts of genotype 1a clone pH77-S (88) and genotype 1b clone pFK-Con1 (56) with specific adaptive mutations. Even though transcripts from pJFH1 led to productive infection of Huh7 cells (79, 89), efficient growth characteristics also depended on acquisition of adaptive mutations (33, 62, 90). In this study, pS52 and pED43 were not viable in Huh7.5 cell culture. Cell culture adaptive mutations identified in JFH1 (33) and JFH1-based intergenotypic recombinants (23, 65, 86) did not lead to cell culture adaptation of pS52 and pED43. Such mutations might adapt the respective protein to cell culture, but they might also mediate interaction of HCV proteins derived from different genotypes (18, 31, 45, 87). Proof of functionality of pS52 and pED43 in vivo implied proof of functionality of the individual proteins. This knowledge will further the development of intergenotypic recombinant cell culture systems containing yet undefined, minimal JFH1 elements that are critical for cell culture viability. Transfection of RNA transcripts of a genotype 1b cDNA clone into SCID-uPA mice engrafted with human hepatocytes was recently reported (34). In future studies, this model system might offer an alternative for studies of infectivity of cDNA clones in vivo. However, it will not allow studies of immune responses, pathogenesis, and persistence.

In chimpanzees, a comparable course of infection following inoculation with viral particles and intrahepatic transfection with RNA transcripts of cDNA clones was reported (4, 67, 80). Intrahepatic transfection of chimpanzees CH5276 and CH5300 with RNA transcripts of pS52 and pED43 led to robust infection, comparable to infection seen after transfection with previously described fully functional cDNA clones (1, 14, 28, 35, 37, 46, 47, 63, 72, 74, 83-85). At time points with peak HCV RNA titers, the consensus ORF sequences of recovered viruses were identical to that of pS52 and pED43, indicating full functionality of the constructed sequence. This was in contrast to JFH1, which had already acquired adaptive mutations 2 weeks after intrahepatic transfection (32). Even though chimpanzees CH5276 and CH5300 became persistently infected, significant differences were observed regarding the course of viremia, hepatitis, and cellular immune responses. As previously described for H77-infected chimpanzees (14, 46), in CH5300, the initial increase in viral RNA (weeks 1 to 6) showed a biphasic pattern with a primary rapid and secondary slower slope, separated by a transient decline (at week 2) (Fig. 3A). It has been suggested that this decrease in viral replication results from activation of innate antiviral defense mechanisms and especially the type I IFN system (46), because no intrahepatic HCV-reactive T cells but elevated intrahepatic 2′,5′-oligoadenylate synthetase 1 mRNA levels were found during the first weeks of HCV infection (46, 72). Interestingly, for CH5276, the decline in HCV RNA observed at week 2 was far more pronounced than for CH5300. Also, the second increase in HCV RNA was more delayed in CH5276 than in CH5300 and acutely infected chimpanzees previously studied (Fig. 2A). In human patients, HCV is highly sensitive to treatment with alpha interferon during the acute phase of infection (29), and in chronically infected individuals, genotype 3a is more sensitive to interferon treatment than genotypes 1 and 4 are (22, 48). Thus, genotype 3a might also be more sensitive to endogenous interferon production during the acute phase of infection. In line with this, higher spontaneous clearance rates have been reported for genotype 3a in one study (41), but not other studies (51). In addition, even though genotype 2a is supposed to be relatively sensitive to IFN (48), after transfection of a chimpanzee with RNA transcripts from genotype 2a cDNA clone pJ6CF, the decline in RNA titers was not as pronounced as seen for S52 (84). In order to draw conclusions about the dependence of early kinetics of HCV infection on the genotype, more studies with different isolates and including monitoring correlates of innate immunity and other host factors will be needed.

In several studies, different patterns of viremia were observed during the course of acute HCV infection (2, 15, 67). In chimpanzee CH5300, a plateau with peak HCV RNA titers (weeks 5 and 6) was followed by a rapid 100-fold decrease of HCV RNA, presumably mediated by the onset of the adaptive immune response and associated with liver damage (Fig. 3). This pattern was typically observed in animals that cleared HCV, but it has also been observed in animals that subsequently developed persistent infection (15, 27, 57, 67); it has not been entirely clarified which immunological and/or viral features are determining such different outcomes. In other animals with persistent infection, as was observed for CH5276, HCV RNA titers remained relatively stable during the acute phase of infection (15, 27, 57, 67).

Chimpanzees with acute S52 and ED43 infections did not develop neutralizing antibodies. While neutralizing antibodies have commonly been found in the chronic phase of infection, they have frequently been absent during the acute phase (10, 15, 22, 53, 63). Even though in patients, the occurrence of neutralizing antibodies in the acute phase was associated with viral clearance (38, 54), neutralizing antibodies were not a prerequisite for infection control, since they were reported to be absent during resolving infections (10, 15, 63). In this study, we implemented a new method to evaluate neutralization of HCV. ELISPOT software was customized for counting focus-forming units, consisting of groups of Huh7.5 cells expressing HCV NS5A, visualized with a horseradish peroxidase-catalyzed reaction. Compared to manual counting, this method greatly facilitates evaluation of biological assays, such as analysis of antibody-mediated neutralization as well as analysis of receptor blocking and the effect of antiviral compounds.

Closely spaced sampling points provided a unique opportunity to compare the kinetics and breadth of T-cell immunity in liver and blood throughout the acute and chronic phases of infection. In one animal (CH5300), cellular immunity developed early, targeted multiple HCV proteins, and was associated with transient control of viral replication, while in the other animal (CH5276), a narrow response that was delayed for months provided no apparent control of viremia. While these divergent patterns of cellular immunity and virus replication have been reported (2, 15, 27, 57, 67, 75), two unique aspects of acute infection are highlighted by the present study. First, CD4+ and CD8+ T cells targeting multiple structural and nonstructural HCV proteins were isolated from the liver of chimpanzee CH5300 after 4 weeks of infection. Nevertheless, control of viremia and increased transaminase activity were not evident until 3 weeks later when cellular immune responses were first detected in blood (Fig. 3A and Table 7). Transient anergy of HCV-specific T cells that circulate in the blood of humans (25, 40, 73) and chimpanzees (68) has been reported, but whether this phenomenon can explain the long delay between the infiltration of effector T cells into the liver and initial suppression of viremia in chimpanzee CH5300 is not known. Mutational escape of major histocompatibility complex (MHC) class I-restricted epitopes is common in chimpanzees and humans that develop chronic infections (3). It is possible that an early ineffective T-cell response that is detectable only in the liver drives mutational escape of dominant epitopes that cannot be recognized when fully functional effector cells emerge several weeks later in blood (2). Whether this apparent delay between the initial T-cell infiltration of the liver and control of viremia is shorter in animals and humans that successfully eradicate the infection merits further study using closely spaced sampling points like those described here. The onset of this adaptive immune response was associated with ALT elevation and temporary suppression of viremia. Viral persistence despite such a primarily successful immune response has been reported previously (72).

Early, strong, multispecific, and sustained CD4+ and CD8+ T-cell responses have been associated with viral clearance in humans and chimpanzees (2, 15, 27, 57, 60, 67). In chimpanzees, the occurrence of intrahepatic HCV-reactive IFN-γ-secreting CD4+ and CD8+ T cells correlated with an increase in ALT and with at least temporary resolution of viremia (12, 72). Also, the occurrence of HCV-reactive PBMC, usually present at low frequency, was associated with viral clearance (68, 72). In general, T-cell responses to HCV occur with a 4- to 8-week delay after infection, even during a successful adaptive immune response (2, 15, 27, 57, 67).

During the acute phase of HCV infection in some patients, CD4+/CD8+ T-cell populations were found to decrease before rebound of viremia (13, 21, 26, 39); several mechanisms might contribute to such impairment of the cellular immune response (15, 59). Similarly, in chimpanzee CH5300, a decrease in the number of HCV-reactive T cells was observed before rebound of viremia (Fig. 3). At week 12, no HCV-reactive CD4+ cells were found, and at weeks 10 and 11, no HCV-reactive CD8+ cells were found (data not shown). In addition, HCV-reactive T cells observed in CH5300 were mostly CD4+. Thus, before the rebound of viremia, CD8+ T cells with reactivity limited to NS3-2 and NS5B-2 peptide pools were found only during weeks 7 to 9 (data not shown).

The inadequate T-cell response mounted by chimpanzee CH5276 was characterized by the late occurrence of intrahepatic T cells with limited breadth of reactivity and the absence of HCV-reactive PBMC (Fig. 2). In contrast to previous reports, there was no correlation between the occurrence of HCV-specific CD8+ T cells in the liver and blood (66). High numbers of intrahepatic CD8+ T cells were detected at several time points between weeks 11 and 26 (Fig. 2D and data not shown), but no peripheral HCV-reactive PBMC were found throughout the course of infection (Fig. 2C). Similar results were reported by Thimme et al. (72).

In both chimpanzees, chronic infection was marked by a decrease in magnitude and breadth of T-cell responses as described previously (67). However, a certain T-cell population persisted, potentially due to continuous antigen stimulation. As seen for chimpanzee CH5300, this population partially controlled but did not eradicate the infection. Viral persistence might be due to different viral evasion strategies, one of them being viral escape by mutation of epitopes targeted by the adaptive immune system (2, 15, 57, 67). In the current study, we did not investigate potential escape mutants.

In conclusion, we generated fully functional cDNA clones of genotypes 3a (pS52) and 4a (pED43). pS52 and pED43 can be used to study monoclonal genotype 3a and 4a infection in chimpanzees. In this study, infection with S52 was characterized by late onset of overt hepatitis, coinciding with T-cell responses of limited breadth, not able to control viremia. In contrast, infection with ED43 led to early onset of hepatitis and multispecific T-cell responses, resulting in transient control of viremia. Proof of functionality of all genes of pS52 and pED43 is important for the development of research tools for genotypes 3a and 4a, such as recombinant cell culture systems and enzymatic assays.

ACKNOWLEDGMENTS

We thank the animal technical staff at the Michale E. Keeling Center for Comparative Medicine and Research for animal care. We also thank Marisa St. Claire (NIH) for expert assistance during the intrahepatic transfection procedures. We are grateful to CTL, especially Alexey Karulin, Wenji Zhang, and Paul Lehmann, for providing customized software for automated counting of FFU on an ImmunoSpot series 5 UV analyzer (CTL Europe GmbH). We thank Jens Ole Nielsen and Ove Andersen (Copenhagen University Hospital, Hvidovre, Denmark) for their support of the project.

This work was supported by grants from the Copenhagen University Hospital, Hvidovre, Denmark (J.M.G. and T.K.H.S.), the Lundbeck Foundation (J.B.), the Novo Nordisk Foundation (J.M.G. and J.B.), the Danish Medical Research Council (J.B.), the A.P. Møller and the Chastine Mc-Kinney Møller Foundation (J.M.G., T.K.H.S., and J.B.), The Danish Cancer Society (J.M.G. and J.B.), and the U.S. Public Health Service (R37 AI47367 and U19 AI48231; C.M.W.). This study was also supported by the NIH/NCRR U-42 Chimpanzee Biomedical Resource Grant (W.S.), the Intramural Research Program of the NIAID, NIH, and by NIAID contract NO1-AO-62713. T.K.H.S is the recipient of a Ph.D. stipend from the Faculty of Health Sciences, University of Copenhagen, B.C. is the recipient of a Postdoctoral Research Fellowship from the American Liver Foundation, and J.B. is the recipient of a professorship at the University of Copenhagen with external funding from the Lundbeck Foundation.

FOOTNOTES

    • Received 18 December 2009.
    • Accepted 19 February 2010.
  • Copyright © 2010 American Society for Microbiology

REFERENCES

  1. 1.↵
    Beard, M. R., G. Abell, M. Honda, A. Carroll, M. Gartland, B. Clarke, K. Suzuki, R. Lanford, D. V. Sangar, and S. M. Lemon. 1999. An infectious molecular clone of a Japanese genotype 1b hepatitis C virus. Hepatology30:316-324.
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    Bowen, D. G., and C. M. Walker. 2005. Adaptive immune responses in acute and chronic hepatitis C virus infection. Nature436:946-952.
    OpenUrlCrossRefPubMedWeb of Science
  3. 3.↵
    Bowen, D. G., and C. M. Walker. 2005. Mutational escape from CD8+ T cell immunity: HCV evolution, from chimpanzees to man. J. Exp. Med.201:1709-1714.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    Bukh, J. 2004. A critical role for the chimpanzee model in the study of hepatitis C. Hepatology39:1469-1475.
    OpenUrlCrossRefPubMedWeb of Science
  5. 5.↵
    Bukh, J., P. Meuleman, R. Tellier, R. Engle, S. M. Feinstone, G. Eder, W. Satterfield, S. Govindarajan, K. Krawczynski, R. H. Miller, G. Leroux-Roels, and R. H. Purcell. 30 March 2010, posting date. Challenge pools of hepatitis C virus genotype 1-6 prototype strains: replication fitness and pathogenicity in chimpanzees and human liver-chimeric mouse models. J. Infect. Dis. doi:10.1086/651579.
    OpenUrlCrossRef
  6. 6.↵
    Bukh, J., T. Pietschmann, V. Lohmann, N. Krieger, K. Faulk, R. E. Engle, S. Govindarajan, M. Shapiro, M. St. Claire, and R. Bartenschlager. 2002. Mutations that permit efficient replication of hepatitis C virus RNA in Huh-7 cells prevent productive replication in chimpanzees. Proc. Natl. Acad. Sci. U. S. A.99:14416-14421.
    OpenUrlAbstract/FREE Full Text
  7. 7.↵
    Bukh, J., R. H. Purcell, and R. H. Miller. 1992. Sequence analysis of the 5′ noncoding region of hepatitis C virus. Proc. Natl. Acad. Sci. U. S. A.89:4942-4946.
    OpenUrlAbstract/FREE Full Text
  8. 8.↵
    Bukh, J., R. H. Purcell, and R. H. Miller. 1993. At least 12 genotypes of hepatitis C virus predicted by sequence analysis of the putative E1 gene of isolates collected worldwide. Proc. Natl. Acad. Sci. U. S. A.90:8234-8238.
    OpenUrlAbstract/FREE Full Text
  9. 9.↵
    Bukh, J., R. H. Purcell, and R. H. Miller. 1994. Sequence analysis of the core gene of 14 hepatitis C virus genotypes. Proc. Natl. Acad. Sci. U. S. A.91:8239-8243.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    Bukh, J., R. Thimme, J. C. Meunier, K. Faulk, H. C. Spangenberg, K. M. Chang, W. Satterfield, F. V. Chisari, and R. H. Purcell. 2008. Previously infected chimpanzees are not consistently protected against reinfection or persistent infection after reexposure to the identical hepatitis C virus strain. J. Virol.82:8183-8195.
    OpenUrlAbstract/FREE Full Text
  11. 11.↵
    Chamberlain, R. W., N. Adams, A. A. Saeed, P. Simmonds, and R. M. Elliott. 1997. Complete nucleotide sequence of a type 4 hepatitis C virus variant, the predominant genotype in the Middle East. J. Gen. Virol.78:1341-1347.
    OpenUrlCrossRefPubMedWeb of Science
  12. 12.↵
    Cooper, S., A. L. Erickson, E. J. Adams, J. Kansopon, A. J. Weiner, D. Y. Chien, M. Houghton, P. Parham, and C. M. Walker. 1999. Analysis of a successful immune response against hepatitis C virus. Immunity10:439-449.
    OpenUrlCrossRefPubMedWeb of Science
  13. 13.↵
    Cox, A. L., T. Mosbruger, G. M. Lauer, D. Pardoll, D. L. Thomas, and S. C. Ray. 2005. Comprehensive analyses of CD8+ T cell responses during longitudinal study of acute human hepatitis C. Hepatology42:104-112.
    OpenUrlCrossRefPubMedWeb of Science
  14. 14.↵
    Dahari, H., M. Major, X. Zhang, K. Mihalik, C. M. Rice, A. S. Perelson, S. M. Feinstone, and A. U. Neumann. 2005. Mathematical modeling of primary hepatitis C infection: noncytolytic clearance and early blockage of virion production. Gastroenterology128:1056-1066.
    OpenUrlCrossRefPubMedWeb of Science
  15. 15.↵
    Dustin, L. B., and C. M. Rice. 2007. Flying under the radar: the immunobiology of hepatitis C. Annu. Rev. Immunol.25:71-99.
    OpenUrlCrossRefPubMedWeb of Science
  16. 16.↵
    Engle, R. E., R. S. Russell, R. H. Purcell, and J. Bukh. 2008. Development of a TaqMan assay for the six major genotypes of hepatitis C virus: comparison with commercial assays. J. Med. Virol.80:72-79.
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.↵
    Feld, J. J., and J. H. Hoofnagle. 2005. Mechanism of action of interferon and ribavirin in treatment of hepatitis C. Nature436:967-972.
    OpenUrlCrossRefPubMedWeb of Science
  18. 18.↵
    Franck, N., S. J. Le, C. Guguen-Guillouzo, and L. Erdtmann. 2005. Hepatitis C virus NS2 protein is phosphorylated by the protein kinase CK2 and targeted for degradation to the proteasome. J. Virol.79:2700-2708.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    Franco, S., C. Tural, B. Clotet, and M. A. Martinez. 2007. Complete nucleotide sequence of genotype 4 hepatitis C viruses isolated from patients co-infected with human immunodeficiency virus type 1. Virus Res.123:161-169.
    OpenUrlCrossRefPubMedWeb of Science
  20. 20.↵
    Fujimura, Y., S. Ishimoto, T. Shimoyama, N. Narita, Y. Kuze, A. Yoshioka, H. Fukui, T. Tanaka, F. Tsuda, H. Okamoto, Y. Miyakawa, and M. Mayumi. 1996. Genotypes and multiple infections with hepatitis C virus in patients with haemophilia A in Japan. J. Viral Hepat.3:79-84.
    OpenUrlPubMedWeb of Science
  21. 21.↵
    Gerlach, J. T., H. M. Diepolder, M. C. Jung, N. H. Gruener, W. W. Schraut, R. Zachoval, R. Hoffmann, C. A. Schirren, T. Santantonio, and G. R. Pape. 1999. Recurrence of hepatitis C virus after loss of virus-specific CD4(+) T-cell response in acute hepatitis C. Gastroenterology117:933-941.
    OpenUrlCrossRefPubMedWeb of Science
  22. 22.↵
    Gottwein, J. M., and J. Bukh. 2008. Cutting the Gordian knot—development and biological relevance of hepatitis C virus cell culture systems. Adv. Virus Res.71:51-133.
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.↵
    Gottwein, J. M., T. K. Scheel, A. M. Hoegh, J. B. Lademann, J. Eugen-Olsen, G. Lisby, and J. Bukh. 2007. Robust hepatitis C genotype 3a cell culture releasing adapted intergenotypic 3a/2a (S52/JFH1) viruses. Gastroenterology133:1614-1626.
    OpenUrlCrossRefPubMedWeb of Science
  24. 24.↵
    Gottwein, J. M., T. K. Scheel, T. B. Jensen, J. B. Lademann, J. C. Prentoe, M. L. Knudsen, A. M. Hoegh, and J. Bukh. 2009. Development and characterization of hepatitis C virus genotype 1-7 cell culture systems: role of CD81 and scavenger receptor class B type I and effect of antiviral drugs. Hepatology49:364-377.
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    Gruener, N. H., F. Lechner, M. C. Jung, H. Diepolder, T. Gerlach, G. Lauer, B. Walker, J. Sullivan, R. Phillips, G. R. Pape, and P. Klenerman. 2001. Sustained dysfunction of antiviral CD8+ T lymphocytes after infection with hepatitis C virus. J. Virol.75:5550-5558.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    Gruner, N. H., T. J. Gerlach, M. C. Jung, H. M. Diepolder, C. A. Schirren, W. W. Schraut, R. Hoffmann, R. Zachoval, T. Santantonio, M. Cucchiarini, A. Cerny, and G. R. Pape. 2000. Association of hepatitis C virus-specific CD8+ T cells with viral clearance in acute hepatitis C. J. Infect. Dis.181:1528-1536.
    OpenUrlCrossRefPubMedWeb of Science
  27. 27.↵
    Guidotti, L. G., and F. V. Chisari. 2006. Immunobiology and pathogenesis of viral hepatitis. Annu. Rev. Pathol.1:23-61.
    OpenUrlCrossRefPubMedWeb of Science
  28. 28.↵
    Hong, Z., M. Beaudet-Miller, R. E. Lanford, B. Guerra, J. Wright-Minogue, A. Skelton, B. M. Baroudy, G. R. Reyes, and J. Y. Lau. 1999. Generation of transmissible hepatitis C virions from a molecular clone in chimpanzees. Virology256:36-44.
    OpenUrlCrossRefPubMedWeb of Science
  29. 29.↵
    Jaeckel, E., M. Cornberg, H. Wedemeyer, T. Santantonio, J. Mayer, M. Zankel, G. Pastore, M. Dietrich, C. Trautwein, and M. P. Manns. 2001. Treatment of acute hepatitis C with interferon alfa-2b. N. Engl. J. Med.345:1452-1457.
    OpenUrlCrossRefPubMedWeb of Science
  30. 30.↵
    Jensen, T. B., J. M. Gottwein, T. K. Scheel, A. M. Hoegh, J. Eugen-Olsen, and J. Bukh. 2008. Highly efficient JFH1-based cell-culture system for hepatitis C virus genotype 5a: failure of homologous neutralizing-antibody treatment to control infection. J. Infect. Dis.198:1756-1765.
    OpenUrlCrossRefPubMedWeb of Science
  31. 31.↵
    Jirasko, V., R. Montserret, N. Appel, A. Janvier, L. Eustachi, C. Brohm, E. Steinmann, T. Pietschmann, F. Penin, and R. Bartenschlager. 2008. Structural and functional characterization of nonstructural protein 2 for its role in hepatitis C virus assembly. J. Biol. Chem.283:28546-28562.
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    Kato, T., Y. Choi, G. Elmowalid, R. K. Sapp, H. Barth, A. Furusaka, S. Mishiro, T. Wakita, K. Krawczynski, and T. J. Liang. 2008. Hepatitis C virus JFH-1 strain infection in chimpanzees is associated with low pathogenicity and emergence of an adaptive mutation. Hepatology48:732-740.
    OpenUrlCrossRefPubMed
  33. 33.↵
    Kaul, A., I. Woerz, P. Meuleman, G. Leroux-Roels, and R. Bartenschlager. 2007. Cell culture adaptation of hepatitis C virus and in vivo viability of an adapted variant. J. Virol.81:13168-13179.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    Kimura, T., M. Imamura, N. Hiraga, T. Hatakeyama, D. Miki, C. Noguchi, N. Mori, M. Tsuge, S. Takahashi, Y. Fujimoto, E. Iwao, H. Ochi, H. Abe, T. Maekawa, K. Arataki, C. Tateno, K. Yoshizato, T. Wakita, T. Okamoto, Y. Matsuura, and K. Chayama. 2008. Establishment of an infectious genotype 1b hepatitis C virus clone in human hepatocyte chimeric mice. J. Gen. Virol.89:2108-2113.
    OpenUrlCrossRefPubMedWeb of Science
  35. 35.↵
    Kolykhalov, A. A., E. V. Agapov, K. J. Blight, K. Mihalik, S. M. Feinstone, and C. M. Rice. 1997. Transmission of hepatitis C by intrahepatic inoculation with transcribed RNA. Science277:570-574.
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    Kolykhalov, A. A., S. M. Feinstone, and C. M. Rice. 1996. Identification of a highly conserved sequence element at the 3′ terminus of hepatitis C virus genome RNA. J. Virol.70:3363-3371.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    Lanford, R. E., H. Lee, D. Chavez, B. Guerra, and K. M. Brasky. 2001. Infectious cDNA clone of the hepatitis C virus genotype 1 prototype sequence. J. Gen. Virol.82:1291-1297.
    OpenUrlCrossRefPubMedWeb of Science
  38. 38.↵
    Lavillette, D., Y. Morice, G. Germanidis, P. Donot, A. Soulier, E. Pagkalos, G. Sakellariou, L. Intrator, B. Bartosch, J. M. Pawlotsky, and F. L. Cosset. 2005. Human serum facilitates hepatitis C virus infection, and neutralizing responses inversely correlate with viral replication kinetics at the acute phase of hepatitis C virus infection. J. Virol.79:6023-6034.
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    Lechner, F., N. H. Gruener, S. Urbani, J. Uggeri, T. Santantonio, A. R. Kammer, A. Cerny, R. Phillips, C. Ferrari, G. R. Pape, and P. Klenerman. 2000. CD8+ T lymphocyte responses are induced during acute hepatitis C virus infection but are not sustained. Eur. J. Immunol.30:2479-2487.
    OpenUrlCrossRefPubMedWeb of Science
  40. 40.↵
    Lechner, F., D. K. Wong, P. R. Dunbar, R. Chapman, R. T. Chung, P. Dohrenwend, G. Robbins, R. Phillips, P. Klenerman, and B. D. Walker. 2000. Analysis of successful immune responses in persons infected with hepatitis C virus. J. Exp. Med.191:1499-1512.
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    Lehmann, M., M. F. Meyer, M. Monazahian, H. L. Tillmann, M. P. Manns, and H. Wedemeyer. 2004. High rate of spontaneous clearance of acute hepatitis C virus genotype 3 infection. J. Med. Virol.73:387-391.
    OpenUrlCrossRefPubMedWeb of Science
  42. 42.↵
    Lindenbach, B. D., M. J. Evans, A. J. Syder, B. Wolk, T. L. Tellinghuisen, C. C. Liu, T. Maruyama, R. O. Hynes, D. R. Burton, J. A. McKeating, and C. M. Rice. 2005. Complete replication of hepatitis C virus in cell culture. Science309:623-626.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    Logvinoff, C., M. E. Major, D. Oldach, S. Heyward, A. Talal, P. Balfe, S. M. Feinstone, H. Alter, C. M. Rice, and J. A. McKeating. 2004. Neutralizing antibody response during acute and chronic hepatitis C virus infection. Proc. Natl. Acad. Sci. U. S. A.101:10149-10154.
    OpenUrlAbstract/FREE Full Text
  44. 44.↵
    Lohmann, V., F. Korner, J. Koch, U. Herian, L. Theilmann, and R. Bartenschlager. 1999. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science285:110-113.
    OpenUrlAbstract/FREE Full Text
  45. 45.↵
    Ma, Y., J. Yates, Y. Liang, S. M. Lemon, and M. Yi. 2008. NS3 helicase domains involved in infectious intracellular hepatitis C virus particle assembly. J. Virol.82:7624-7639.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    Major, M. E., H. Dahari, K. Mihalik, M. Puig, C. M. Rice, A. U. Neumann, and S. M. Feinstone. 2004. Hepatitis C virus kinetics and host responses associated with disease and outcome of infection in chimpanzees. Hepatology39:1709-1720.
    OpenUrlCrossRefPubMedWeb of Science
  47. 47.↵
    Major, M. E., K. Mihalik, J. Fernandez, J. Seidman, D. Kleiner, A. A. Kolykhalov, C. M. Rice, and S. M. Feinstone. 1999. Long-term follow-up of chimpanzees inoculated with the first infectious clone for hepatitis C virus. J. Virol.73:3317-3325.
    OpenUrlAbstract/FREE Full Text
  48. 48.↵
    Manns, M. P., J. G. McHutchison, S. C. Gordon, V. K. Rustgi, M. Shiffman, R. Reindollar, Z. D. Goodman, K. Koury, M. Ling, and J. K. Albrecht. 2001. Peginterferon alfa-2b plus ribavirin compared with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a randomised trial. Lancet358:958-965.
    OpenUrlCrossRefPubMedWeb of Science
  49. 49.↵
    Meuleman, P., L. Libbrecht, R. De Vos, B. de Hemptinne, K. Gevaert, J. Vandekerckhove, T. Roskams, and G. Leroux-Roels. 2005. Morphological and biochemical characterization of a human liver in a uPA-SCID mouse chimera. Hepatology41:847-856.
    OpenUrlCrossRefPubMedWeb of Science
  50. 50.↵
    Meunier, J. C., R. E. Engle, K. Faulk, M. Zhao, B. Bartosch, H. Alter, S. U. Emerson, F. L. Cosset, R. H. Purcell, and J. Bukh. 2005. Evidence for cross-genotype neutralization of hepatitis C virus pseudo-particles and enhancement of infectivity by apolipoprotein C1. Proc. Natl. Acad. Sci. U. S. A.102:4560-4565.
    OpenUrlAbstract/FREE Full Text
  51. 51.↵
    Micallef, J. M., J. M. Kaldor, and G. J. Dore. 2006. Spontaneous viral clearance following acute hepatitis C infection: a systematic review of longitudinal studies. J. Viral Hepat.13:34-41.
    OpenUrlCrossRefPubMedWeb of Science
  52. 52.↵
    National Research Council. 1996. Guide for the care and use of laboratory animals. National Academy Press, Washington, DC.
  53. 53.↵
    Netski, D. M., T. Mosbruger, E. Depla, G. Maertens, S. C. Ray, R. G. Hamilton, S. Roundtree, D. L. Thomas, J. McKeating, and A. Cox. 2005. Humoral immune response in acute hepatitis C virus infection. Clin. Infect. Dis.41:667-675.
    OpenUrlCrossRefPubMedWeb of Science
  54. 54.↵
    Pestka, J. M., M. B. Zeisel, E. Blaser, P. Schurmann, B. Bartosch, F. L. Cosset, A. H. Patel, H. Meisel, J. Baumert, S. Viazov, K. Rispeter, H. E. Blum, M. Roggendorf, and T. F. Baumert. 2007. Rapid induction of virus-neutralizing antibodies and viral clearance in a single-source outbreak of hepatitis C. Proc. Natl. Acad. Sci. U. S. A.104:6025-6030.
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    Pietschmann, T., A. Kaul, G. Koutsoudakis, A. Shavinskaya, S. Kallis, E. Steinmann, K. Abid, F. Negro, M. Dreux, F. L. Cosset, and R. Bartenschlager. 2006. Construction and characterization of infectious intragenotypic and intergenotypic hepatitis C virus chimeras. Proc. Natl. Acad. Sci. U. S. A.103:7408-7413.
    OpenUrlAbstract/FREE Full Text
  56. 56.↵
    Pietschmann, T., M. Zayas, P. Meuleman, G. Long, N. Appel, G. Koutsoudakis, S. Kallis, G. Leroux-Roels, V. Lohmann, and R. Bartenschlager. 2009. Production of infectious genotype 1b virus particles in cell culture and impairment by replication enhancing mutations. PLoS Pathog.5:e1000475.
    OpenUrlCrossRefPubMed
  57. 57.↵
    Post, J., S. Ratnarajah, and A. R. Lloyd. 2009. Immunological determinants of the outcomes from primary hepatitis C infection. Cell. Mol. Life Sci.66:733-756.
    OpenUrlCrossRefPubMed
  58. 58.↵
    Ray, S. C., R. R. Arthur, A. Carella, J. Bukh, and D. L. Thomas. 2000. Genetic epidemiology of hepatitis C virus throughout Egypt. J. Infect. Dis.182:698-707.
    OpenUrlCrossRefPubMedWeb of Science
  59. 59.↵
    Rehermann, B. 2007. Chronic infections with hepatotropic viruses: mechanisms of impairment of cellular immune responses. Semin. Liver Dis.27:152-160.
    OpenUrlCrossRefPubMedWeb of Science
  60. 60.↵
    Rehermann, B., and M. Nascimbeni. 2005. Immunology of hepatitis B virus and hepatitis C virus infection. Nat. Rev. Immunol.5:215-229.
    OpenUrlCrossRefPubMedWeb of Science
  61. 61.↵
    Rubbia-Brandt, L., R. Quadri, K. Abid, E. Giostra, P. J. Male, G. Mentha, L. Spahr, J. P. Zarski, B. Borisch, A. Hadengue, and F. Negro. 2000. Hepatocyte steatosis is a cytopathic effect of hepatitis C virus genotype 3. J. Hepatol.33:106-115.
    OpenUrlCrossRefPubMedWeb of Science
  62. 62.↵
    Russell, R. S., J. C. Meunier, S. Takikawa, K. Faulk, R. E. Engle, J. Bukh, R. H. Purcell, and S. U. Emerson. 2008. Advantages of a single-cycle production assay to study cell culture-adaptive mutations of hepatitis C virus. Proc. Natl. Acad. Sci. U. S. A.105:4370-4375.
    OpenUrlAbstract/FREE Full Text
  63. 63.↵
    Sakai, A., S. Takikawa, R. Thimme, J. C. Meunier, H. C. Spangenberg, S. Govindarajan, P. Farci, S. U. Emerson, F. V. Chisari, R. H. Purcell, and J. Bukh. 2007. In vivo study of the HC-TN strain of hepatitis C virus recovered from a patient with fulminant hepatitis: RNA transcripts of a molecular clone (pHC-TN) are infectious in chimpanzees but not in Huh7.5 cells. J. Virol.81:7208-7219.
    OpenUrlAbstract/FREE Full Text
  64. 64.↵
    Sakamoto, M., Y. Akahane, F. Tsuda, T. Tanaka, D. G. Woodfield, and H. Okamoto. 1994. Entire nucleotide sequence and characterization of a hepatitis C virus of genotype V/3a. J. Gen. Virol.75:1761-1768.
    OpenUrlCrossRefPubMedWeb of Science
  65. 65.↵
    Scheel, T. K., J. M. Gottwein, T. B. Jensen, J. C. Prentoe, A. M. Hoegh, H. J. Alter, J. Eugen-Olsen, and J. Bukh. 2008. Development of JFH1-based cell culture systems for hepatitis C virus genotype 4a and evidence for cross-genotype neutralization. Proc. Natl. Acad. Sci. U. S. A.105:997-1002.
    OpenUrlAbstract/FREE Full Text
  66. 66.↵
    Shin, E. C., S. Capone, R. Cortese, S. Colloca, A. Nicosia, A. Folgori, and B. Rehermann. 2008. The kinetics of hepatitis C virus-specific CD8 T-cell responses in the blood mirror those in the liver in acute hepatitis C virus infection. J. Virol.82:9782-9788.
    OpenUrlAbstract/FREE Full Text
  67. 67.↵
    Shoukry, N. H., A. G. Cawthon, and C. M. Walker. 2004. Cell-mediated immunity and the outcome of hepatitis C virus infection. Annu. Rev. Microbiol.58:391-424.
    OpenUrlCrossRefPubMedWeb of Science
  68. 68.↵
    Shoukry, N. H., A. Grakoui, M. Houghton, D. Y. Chien, J. Ghrayeb, K. A. Reimann, and C. M. Walker. 2003. Memory CD8+ T cells are required for protection from persistent hepatitis C virus infection. J. Exp. Med.197:1645-1655.
    OpenUrlAbstract/FREE Full Text
  69. 69.↵
    Simmonds, P. 2004. Genetic diversity and evolution of hepatitis C virus - 15 years on. J. Gen. Virol.85:3173-3188.
    OpenUrlCrossRefPubMedWeb of Science
  70. 70.↵
    Simmonds, P., J. Bukh, C. Combet, G. Deleage, N. Enomoto, S. Feinstone, P. Halfon, G. Inchauspe, C. Kuiken, G. Maertens, M. Mizokami, D. G. Murphy, H. Okamoto, J. M. Pawlotsky, F. Penin, E. Sablon, I. T. Shin, L. J. Stuyver, H. J. Thiel, S. Viazov, A. J. Weiner, and A. Widell. 2005. Consensus proposals for a unified system of nomenclature of hepatitis C virus genotypes. Hepatology42:962-973.
    OpenUrlCrossRefPubMedWeb of Science
  71. 71.↵
    Tellinghuisen, T. L., J. Marcotrigiano, A. E. Gorbalenya, and C. M. Rice. 2004. The NS5A protein of hepatitis C virus is a zinc metalloprotein. J. Biol. Chem.279:48576-48587.
    OpenUrlAbstract/FREE Full Text
  72. 72.↵
    Thimme, R., J. Bukh, H. C. Spangenberg, S. Wieland, J. Pemberton, C. Steiger, S. Govindarajan, R. H. Purcell, and F. V. Chisari. 2002. Viral and immunological determinants of hepatitis C virus clearance, persistence, and disease. Proc. Natl. Acad. Sci. U. S. A.99:15661-15668.
    OpenUrlAbstract/FREE Full Text
  73. 73.↵
    Thimme, R., D. Oldach, K. M. Chang, C. Steiger, S. C. Ray, and F. V. Chisari. 2001. Determinants of viral clearance and persistence during acute hepatitis C virus infection. J. Exp. Med.194:1395-1406.
    OpenUrlAbstract/FREE Full Text
  74. 74.↵
    Thomson, M., M. Nascimbeni, S. Gonzales, K. K. Murthy, B. Rehermann, and T. J. Liang. 2001. Emergence of a distinct pattern of viral mutations in chimpanzees infected with a homogeneous inoculum of hepatitis C virus. Gastroenterology121:1226-1233.
    OpenUrlCrossRefPubMed
  75. 75.↵
    Thomson, M., M. Nascimbeni, M. B. Havert, M. Major, S. Gonzales, H. Alter, S. M. Feinstone, K. K. Murthy, B. Rehermann, and T. J. Liang. 2003. The clearance of hepatitis C virus infection in chimpanzees may not necessarily correlate with the appearance of acquired immunity. J. Virol.77:862-870.
    OpenUrlAbstract/FREE Full Text
  76. 76.↵
    Timm, J., M. Neukamm, T. Kuntzen, A. Y. Kim, R. T. Chung, C. Brander, G. M. Lauer, B. D. Walker, and T. M. Allen. 2007. Characterization of full-length hepatitis C virus genotype 4 sequences. J. Viral Hepat.14:330-337.
    OpenUrlCrossRefPubMed
  77. 77.↵
    Tokita, H., H. Okamoto, H. Iizuka, J. Kishimoto, F. Tsuda, Y. Miyakawa, and M. Mayumi. 1998. The entire nucleotide sequences of three hepatitis C virus isolates in genetic groups 7-9 and comparison with those in the other eight genetic groups. J. Gen. Virol.79:1847-1857.
    OpenUrlPubMedWeb of Science
  78. 78.↵
    Tokita, H., S. M. Shrestha, H. Okamoto, M. Sakamoto, M. Horikita, H. Iizuka, S. Shrestha, Y. Miyakawa, and M. Mayumi. 1994. Hepatitis C virus variants from Nepal with novel genotypes and their classification into the third major group. J. Gen. Virol.75:931-936.
    OpenUrlCrossRefPubMed
  79. 79.↵
    Wakita, T., T. Pietschmann, T. Kato, T. Date, M. Miyamoto, Z. Zhao, K. Murthy, A. Habermann, H. G. Krausslich, M. Mizokami, R. Bartenschlager, and T. J. Liang. 2005. Production of infectious hepatitis C virus in tissue culture from a cloned viral genome. Nat. Med.11:791-796.
    OpenUrlCrossRefPubMedWeb of Science
  80. 80.↵
    Walker, C. M. 1997. Comparative features of hepatitis C virus infection in humans and chimpanzees. Springer Semin. Immunopathol.19:85-98.
    OpenUrlCrossRefPubMedWeb of Science
  81. 81.↵
    Yamada, N., K. Tanihara, M. Mizokami, K. Ohba, A. Takada, M. Tsutsumi, and T. Date. 1994. Full-length sequence of the genome of hepatitis C virus type 3a: comparative study with different genotypes. J. Gen. Virol.75:3279-3284.
    OpenUrlCrossRefPubMedWeb of Science
  82. 82.↵
    Yamada, N., K. Tanihara, A. Takada, T. Yorihuzi, M. Tsutsumi, H. Shimomura, T. Tsuji, and T. Date. 1996. Genetic organization and diversity of the 3′ noncoding region of the hepatitis C virus genome. Virology223:255-261.
    OpenUrlCrossRefPubMed
  83. 83.↵
    Yanagi, M., R. H. Purcell, S. U. Emerson, and J. Bukh. 1997. Transcripts from a single full-length cDNA clone of hepatitis C virus are infectious when directly transfected into the liver of a chimpanzee. Proc. Natl. Acad. Sci. U. S. A.94:8738-8743.
    OpenUrlAbstract/FREE Full Text
  84. 84.↵
    Yanagi, M., R. H. Purcell, S. U. Emerson, and J. Bukh. 1999. Hepatitis C virus: an infectious molecular clone of a second major genotype (2a) and lack of viability of intertypic 1a and 2a chimeras. Virology262:250-263.
    OpenUrlCrossRefPubMedWeb of Science
  85. 85.↵
    Yanagi, M., M. St. Claire, M. Shapiro, S. U. Emerson, R. H. Purcell, and J. Bukh. 1998. Transcripts of a chimeric cDNA clone of hepatitis C virus genotype 1b are infectious in vivo. Virology244:161-172.
    OpenUrlCrossRefPubMedWeb of Science
  86. 86.↵
    Yi, M., Y. Ma, J. Yates, and S. M. Lemon. 2007. Compensatory mutations in E1, p7, NS2, and NS3 enhance yields of cell culture-infectious intergenotypic chimeric hepatitis C virus. J. Virol.81:629-638.
    OpenUrlAbstract/FREE Full Text
  87. 87.↵
    Yi, M., Y. Ma, J. Yates, and S. M. Lemon. 2009. Trans-complementation of an NS2 defect in a late step in hepatitis C virus (HCV) particle assembly and maturation. PLoS Pathog.5:e1000403.
    OpenUrlCrossRefPubMed
  88. 88.↵
    Yi, M., R. A. Villanueva, D. L. Thomas, T. Wakita, and S. M. Lemon. 2006. Production of infectious genotype 1a hepatitis C virus (Hutchinson strain) in cultured human hepatoma cells. Proc. Natl. Acad. Sci. U. S. A.103:2310-2315.
    OpenUrlAbstract/FREE Full Text
  89. 89.↵
    Zhong, J., P. Gastaminza, G. Cheng, S. Kapadia, T. Kato, D. R. Burton, S. F. Wieland, S. L. Uprichard, T. Wakita, and F. V. Chisari. 2005. Robust hepatitis C virus infection in vitro. Proc. Natl. Acad. Sci. U. S. A.102:9294-9299.
    OpenUrlAbstract/FREE Full Text
  90. 90.↵
    Zhong, J., P. Gastaminza, J. Chung, Z. Stamataki, M. Isogawa, G. Cheng, J. A. McKeating, and F. V. Chisari. 2006. Persistent hepatitis C virus infection in vitro: coevolution of virus and host. J. Virol.80:11082-11093.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Download PDF
Citation Tools
Novel Infectious cDNA Clones of Hepatitis C Virus Genotype 3a (Strain S52) and 4a (Strain ED43): Genetic Analyses and In Vivo Pathogenesis Studies
Judith M. Gottwein, Troels K. H. Scheel, Benoit Callendret, Yi-Ping Li, Heather B. Eccleston, Ronald E. Engle, Sugantha Govindarajan, William Satterfield, Robert H. Purcell, Christopher M. Walker, Jens Bukh
Journal of Virology Apr 2010, 84 (10) 5277-5293; DOI: 10.1128/JVI.02667-09

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Journal of Virology article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Novel Infectious cDNA Clones of Hepatitis C Virus Genotype 3a (Strain S52) and 4a (Strain ED43): Genetic Analyses and In Vivo Pathogenesis Studies
(Your Name) has forwarded a page to you from Journal of Virology
(Your Name) thought you would be interested in this article in Journal of Virology.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Novel Infectious cDNA Clones of Hepatitis C Virus Genotype 3a (Strain S52) and 4a (Strain ED43): Genetic Analyses and In Vivo Pathogenesis Studies
Judith M. Gottwein, Troels K. H. Scheel, Benoit Callendret, Yi-Ping Li, Heather B. Eccleston, Ronald E. Engle, Sugantha Govindarajan, William Satterfield, Robert H. Purcell, Christopher M. Walker, Jens Bukh
Journal of Virology Apr 2010, 84 (10) 5277-5293; DOI: 10.1128/JVI.02667-09
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • ABSTRACT
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • ACKNOWLEDGMENTS
    • FOOTNOTES
    • REFERENCES
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

DNA, Complementary
hepacivirus
Hepatitis C
Hepatocytes
Monkey Diseases
RNA, Viral

Related Articles

Cited By...

About

  • About JVI
  • Editor in Chief
  • Editorial Board
  • Policies
  • For Reviewers
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Article Types
  • Ethics
  • Contact Us

Follow #Jvirology

@ASMicrobiology

       

 

JVI in collaboration with

American Society for Virology

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0022-538X; Online ISSN: 1098-5514