Skip to main content
Erschienen in: BMC Infectious Diseases 1/2018

Open Access 01.12.2018 | Research article

Successful infection of BALB/c mice by a swine hepatitis E virus clone constructed with reverse genetics

verfasst von: Wenhai Yu, Chenchen Yang, Xianhui Hao, Tianwu Ma, Fen Huang

Erschienen in: BMC Infectious Diseases | Ausgabe 1/2018

Abstract

Background

Hepatitis E virus (HEV) is a leading cause of hepatitis worldwide. However, its infection biology and pathogenesis remain largely elusive. Furthermore, no proven medication is available for treating hepatitis E. Robust experimental models are urgently required to advance the research of HEV infection. Because of the lacking of a sophisticated small animal model, this study aimed to establish a mouse model of HEV infection.

Methods

We constructed a full-length swine HEV cDNA clone of genotype 4 (named as pGEM-HEV) by reverse genetics approach. And we inoculated with HEV RNA in BALB/c mice to establish small animal model for HEV infection and pathogenesis studies.

Results

The capped RNA transcripts of pGEM-HEV prepared in vitro were replication-competent in HepG2 cells. Importantly, BALB/c mice intravenously inoculated with RNA transcripts of pGEM-HEV developed an active infection as shown by shedding viruses in feces, detectable negative strand of HEV in the liver, spleen and kidney, and causing liver inflammation.

Conclusion

In this study, we successfully established of BALB/c mice-based small animal model for HEV provides an opportunity to further understand HEV pathogenesis and to develop effective antiviral medications.
Abkürzungen
A549 cells
Human lung carcinoma cell line
ACUC
Animal Care and Use Committee
ALP
Alkaline phosphatase
ALT
Alanine aminotransferase
AST
Aspartate aminotransferase
ATCC
American Type Culture Collection
DAPI
4′,6-diamidino-2-phenylindole
DEPC
Diethyl pyrocarbonate
DMEM
Dulbecco’s modified eagle medium
dpi
days post-inoculation
ELISA
Enzyme-linked immunosorbent assay
FBS
Fetal bovine serum
FITC
Fluorescein isothiocyanate
HepG2 cells
Human hepatocellular carcinoma cell line
HEV
Hepatitis E virus
IFA
Immunofluorescence assay
IgG
Immunoglobulin G
IgM
Immunoglobulin M
ORFs
Open reading frames
PBS
Phosphate buffered saline
qRT-PCR
quantitative real time PCR
RACE
Rapid Amplification of cDNA Ends
RT-nPCR
Reverse transcription nested PCR
S. D.
Standard deviation
SD rat
Sprague-Dawley rat
SDS-PAGE
Sodium dodecyl sulfate-polyacrylamide gel electrophoresis
SPF
Specific pathogen free

Background

Hepatitis E virus (HEV) is classified in the genus Orthohepevirus of the family Hepeviridae [1]. It is a non-enveloped, single-stranded, positive-sense RNA virus, with an approximately 7.3 kb genome. The viral genome consists of three open reading frames (ORFs) flanked by short 5′ and 3′ non-translated regions, ORF1 encodes a nonstructural protein, ORF2 encodes a capsid protein and ORF3 encodes a small multifunction protein that is essential for viral infection [25]. A unique feature as a hepatitis virus is that HEV has a zoonotic nature and can cross-species transmit in human, swine and deer [610].
HEV is considered the most common cause of hepatitis worldwide [11]. It causes both endemic and epidemic forms of hepatitis E in many developing countries. It is transmitted by the fecal-oral route and waterborne transmission is most often described. In developed countries, most documented cases of acute hepatitis E are sporadic and endemic cases attributed to food consumption [1113]. Although the infection is generally acute and self-limiting, up to about 25~30% mortality has been reported following HEV infection during pregnancy [14, 15]. However, the biology and pathogenesis of HEV infection remain largely elusive and no proven antiviral medication is available.
Robust experiment models are the most important tools for advancing fundamental and translational research of hepatitis E infection. Fortunately, several cell culture systems for propagating HEV have been recently developed [1618]. However, the development of animal models, in particular the use of small laboratory animals, has not been well-explored. Although swine and rabbit have been used to model HEV infection [19, 20], experimental infection in mouse model, the most commonly used laboratory species, has not been established.
We previously have attempted to establish BALB/c nude mice-based HEV model [21]. However, this strain lacks a thymus and is therefore unable to produce T-cells. The immunodeficient nature with a strict life condition and limited fertility has hampered the further application. To circumstance these bottlenecks, this study aimed to establish regular BALB/c mice-based HEV model. We first constructed an infectious cDNA clone of swine HEV with reverse genetics approach. We demonstrated its infectivity in cell culture and importantly also in BALB/c mice. Most interestingly, HEV provokes host response with production of anti-HEV antibodies and induction of liver inflammation, mimicking infection in human. Therefore, this model bears important implications for studying HEV infection and drug development.

Methods

Construction of a full-length cDNA clone of HEV

The full-length of swine HEV (genotype 4, KM01, GenBank No. KJ155502) was amplified with specific primers shown in Table 1 [22]. The collection of stool specimens was approved by the owner. Five overlapping fragments were amplified by PCR. The 3′ end and 5′ end of the virus were obtained using the RACE 5' or 3′ kit (Takara). The entire viral genome was ligated together at indicated restriction enzyme sites in each fragment (Fig. 1). A unique Xba I restriction enzyme site and a T7 RNA polymerase core promoter were introduced at the extreme 5′ terminus. Twenty-four adenosines (A) was engineered at the 3′ end of viral genome, followed by a Cla I restriction enzyme site for plasmid linearization (Fig. 1). PCR productions were purified and cloned into pMD-18 T vector, followed by sequencing with three clones of each fragment. The clone containing the consensus sequence was used for infectious clone assembly. One silent mutation at nucleotide (nt) 4120 (G → T) generated during the PCR amplification was retained as a genetic marker. The full-length genomic cDNA was introduced into the pGEM-7zf (+) vector (Promega) between Xba I and Cla I sites to produce a full-length HEV cDNA clone named pGEM-HEV.
Table 1
Primers sequence
Primer ID
Sequence (5′~ 3′)
Product
Length (bp)
Reference
S11
AGGCTCCTGGCRTYACTACTG
F1
1163 bp
2
S12
GCCYTGGCGAATGCTGTG
A11
GGCCRGGAATGTAATCACG
A12
GCGGCACTGGGCRTAAAACT
HEV-A1
AAAGGAATGAAGAGGCTGGAG
F2
1520 bp
This study
HEV-A3
GAAAAGTCTGGCCGTGATTAC
HEV-B2
TCCTCAGTAATAGTAAGGGC
HEV-B4
AGGTCGATGGTTACGTTCCC
HEV-C1
TGCCTGTTGGGCTGAGTTTTGATG
F3
1394 bp
This study
HEV-C3
GCCAGCCATAGCTTGGTTTGAAG
D1R
AAGGTCTTGCTCCACGCAGATATC
D3R
CTGGAAGAATGTTATACGAGACAC
D2F
CTTGTGGAGGCCATGGTGGAGAA
F4
1243 bp
This study
D4F
ATGGTGGAGAAAGGCCAGGAT
ER1
TCACGCCAAGCGGAGCCGAGT
ER2
GAAGGGGTTGGTTGGATGAAT
EF1
TTTCTGGGGTGACCGGGTTGATT
F5
1791 bp
This study
EF2
CTATATTCATCCAACCAACCCCT
HEV31
CAGGGAGCGCGGAACGCAGAAAAGA
HEV32
TCAATACTCCCGAGTTTTACCCACC
5' Race-A1
GCAGTGARTARAGYGCAAYCCCHGTCT
5' RACE
501 bp
2
5' Race-A2
CGRGCCATYGCCTCNGCRACATC
3' Race-S1
ACYACNACTGCTGCYACACGBTTYATGA
3' RACR
936 bp
2
3' Race-S2
CTYTGTTYAAYCTTGCTGAYACGCTKCTC
T7P1
GCCTAGCTAGCTAGTCTAGATAATACGACTCACTATA
5′ terminal
This study
PAP2
CGGTCGACCGATCGATTTTTTTTTTTTTTTTTTTTTTTTTCAGGGAGCGC
3′ terminal
This study
HEV1
AATTATGCC(T)CAGTAC(T)CGG(A)GTTG
Positive strand of HEV RNA
348 bp
[23]
HEV2
CCCTTA(G)TCC(T)TGCTGA(C)GCATTCTC
HEV3
GTT(A)ATGCTT(C)TGCATA(T)CATGGCT
HEV4
AGCCGACGAAATCAATTCTGTC
HEV6
AGCTCCTGTACCTGATGATGTTGACTC
Negative strand of HEV RNA
266 bp
[24]
HEV7
CTACAGAGCGCCAGCCTTGATTGC
HEV8
GCTCACGTCATCTGTCGCTGCTGG
HEV9
GGGCTGAACCAAAATCCTGACATC
WHO-F
GGTGGTTTCTGGGGTGAC
qRT-PCR
70 bp
[25]
WHO-R
AGGGGTTGGTTGGATGAA

In vitro transcription of capped full-length HEV genomic RNA

The full-length cDNA clones of pGEM-HEV was linearized by Cla I, then digested with proteinase K, and purified by phenol extraction followed by ethanol precipitation. Capped RNA transcripts from pGEM-HEV were synthesized in vitro with the T7 RiboMAX™ Express Large Scale RNA Production System (Promega, USA) according to the direction. The RNA transcripts from the cDNA clone was quantitated (1 μg /μL) and stored in − 80 °C until use.

Cell transfection and immunofluorescence assay

The human hepatoma HepG2 cells and human lung carcinoma A549 cells were incubated at 37 °C with 5% CO2 in Dulbecco’s modified eagle medium (DMEM) with 10% fetal bovine serum (FBS). To evaluate the infectivity of the pGEM-HEV clone, HepG2 (60~70% confluence) cells were transfected with the capped RNA transcripts as described previously [19]. Twenty-four hours post-transfection, the cells were fixed and stained with a HEV-specific antibody (Merck Millipore, MAB8003, German, 1:1000 dilution). Briefly, cells were fixed with 4% paraformaldehyde for 15 min at 37 °C and subsequently washed three times and incubated with HEV antibody for 60 min at 37 °C. Cells were washed three times with PBS (phosphate buffered saline) and incubated with FITC (fluorescein isothiocyanate) conjugated goat anti-mouse IgG (H + L) antibody (Promega, USA, 1:1000 dilution) for 45 min at 37 °C. Nuclei were counterstained with 4′,6-diamidino-2-phenylindole (DAPI). Cells were washed three times with PBS, and viewed under a fluorescence microscopy (Nikon Ti-E, Japan).

Inoculation of BALB/c mice with capped RNA transcripts

The protocol of animal experiments were approved by the Animal Care and Use Committee (ACUC) of Kunming University of Science and Technology. Six female, 8-week-old, SPF (specific pathogen free) BALB/c mouse purchased from SLAC laboratory animal (China), and maintained in a pathogen-free animal facility. Mice were negative to anti-HEV IgG and IgM antibodies, and HEV RNA. Six mice were randomly divided into 2 groups and each mouse was caged separately.
Group A containing Mouse 1, 2 and 3 were inoculated with RNA transcripts of the swine HEV clone pGEM-HEV (10 μg /mouse), and Group B containing mouse 4, 5 and 6 were inoculated with equal volume PBS. Feces and serum were collected every week from each mouse after inoculation. Mice were anaesthetized with pentobarbital (80 mg/kg) and euthanized by cervical dislocation at 28 days post-inoculation (dpi). Tissues, including liver, spleen, kidney and colon were collected. Animal carcasses were safely disposed by the ACUC of the Kunming University of Science and Technology (WH17037).

Detection of HEV RNA in feces, serum and tissues by nested RT-PCR and quantitative RT-PCR

Total RNA in feces, serum and tissues were extracted by Trizol (Invitrogen), according to the manufacturer’s instructions. The isolated RNA was used to synthesize first-strand cDNA. The cDNA was added as the template to perform the reverse transcription nested PCR (RT-nPCR). Both positive strand and (or) negative strands of HEV in feces, serum and tissues were detected with strand-specific primers [23, 24] as described previously and shown in Table 1.
The viral titer of HEV in the feces, serum and tissues were quantified using SYBR green-based quantitative RT-PCR (qRT-PCR) with HEV-specific primers [25]. qRT-PCR was performed using the BIO-RAD CFX Connect Real-Time System under the following conditions: 95 °C for 30 s, followed by 39 cycles of 95 °C for 5 s and 60 °C for 31 s.

Determination of HEV antibodies by ELISA

The HEV IgG and IgM antibodies in serum were tested by ELISA (KHB, China) according to the manufacturer’s instructions. The cutoff values for the IgG and IgM assay were determined based on 0.22 (or 0.24 for IgM) plus the mean OD450/630 values of serum from uninfected mouse (± standard deviation, S. D.).

Profile liver biochemistry in serum

The activities of alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) in serum were measured with an automated biochemistry analyzer (Olympus 2700, Japan).

Detection of HEV ORF2 protein in tissues by Western blotting

The livers, spleens, kidneys and colons were collected and HEV capsid protein were detected in these tissues by Western blotting. Briefly, the tissues were broken in liquid nitrogen by grinding, and were lysed in RIPA buffer. Equal volumes of tissue lysates from each condition were resolved by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Analysis of protein was visualized by direct Western blotting using antibodies directed against the indicated antigens.

Infection of A549 cells with viruses recovered from mouse inoculated with capped RNA transcripts of swine HEV clones

The mouse feces were suspended into 0.1% DEPC (diethyl pyrocarbonate)-PBS. The supernatant was collected by centrifuge and microorganisms were removed by filter. A549 cells were infected with the viruses recovered from mice experimentally infected with feces supernatant. Cells were harvested at 72 h post-infection, and the HEV ORF2 protein was be detected by Western blotting.

Histopathologic examination

The liver tissues were fixed in 10% neutral buffered formalin, sectioned at a thickness of 3 μm, and stained with hematoxylin and eosin. All sections were examined using a Nikon Ti-E microscope (Japan).

Statistical analysis

Prism software (GraphPad Software) was used for statistical analysis. Data were presented as mean ± S. D..

Results

A full-length cDNA clone of swine HEV is infectious

To assess the infectivity of the pGEM-HEV cDNA clone, capped RNA transcripts were transfected into HepG2 cells. Twenty-four hours post-transfection, HEV antigens were detected by immunofluorescence assay (IFA) with an anti-HEV ORF2 monoclonal antibody. The intense fluorescent signals of HEV antigens indicating that the transfected viral RNA was replication-competent in HepG2 cells (Fig. 2). The fluorescent signal was detected in HepG2 cells transfected with the capped RNA transcripts, while no signal in non-tranfected control cells.

Capped RNA transcripts of pGEM-HEV was infectious in BALB/c mice

To further investigate whether this clone can be used to establish infection in mice, we explored the infection in BALB/c mouse model. Mice in groups A and B were intravenously injected with full-length capped RNA transcripts from pGEM-HEV (HEV RNA transcripts) and PBS (Control), respectively. HEV RNA was first detected in feces at 3 dpi in group A inoculated with RNA transcripts, and it was all detected in feces from three mice feces after 7 dpi, and lasted to the end of the experiment (Table 2). The viral titer of HEV in the feces of mice inoculated with HEV RNA transcripts was detected from 3 to 28 dpi (Fig. 3a). In the serum, HEV RNA positive-strands were all detected in mice inoculated with RNA transcripts from 7 to 28 dpi (Fig. 3b). The HEV negative-strand RNA was detected in one mouse at 7 dpi and two mice at 14 dpi in group A inoculated with RNA transcript, and all detected at 21 dpi. However, mice injected with PBS were negative to both positive and negative strands during the whole experiment.
Table 2
Detection of HEV RNA by RT-nPCR in feces, serum and tissues of mouse inoculated with capped full-length RNA transcripts of pGEM-HEV or PBS
Sample
Strand
HEV RNA transcripts
Control
Feces
0 dpi
Positive
3 dpi
Positive
+(1/3)
+(0/3)
4 dpi
Positive
+(2/3)
+(0/3)
7 dpi
Positive
+(3/3)
+(0/3)
14 dpi
Positive
+(3/3)
+(0/3)
21 dpi
Positive
+(3/3)
+(0/3)
28 dpi
Positive
+(3/3)
+(0/3)
Serum
0 dpi
Positive
+(0/3)
+(0/3)
Negative
+(0/3)
+(0/3)
7 dpi
Positive
+(3/3)
+(0/3)
Negative
+(1/3)
+(0/3)
14 dpi
Positive
+(3/3)
+(0/3)
Negative
+(2/3)
+(0/3)
21 dpi
Positive
+(3/3)
+(0/3)
Negative
+(3/3)
+(0/3)
28 dpi
Positive
+(3/3)
+(0/3)
Negative
+(2/3)
+(0/3)
Tissues
Liver
Positive
+(3/3)
+(0/3)
Negative
+(3/3)
+(0/3)
Spleen
Positive
+(3/3)
+(0/3)
Negative
+(3/3)
+(0/3)
Kidney
Positive
+(2/3)
+(0/3)
Negative
+(2/3)
+(0/3)
Colon
Positive
+(0/3)
+(0/3)
Negative
+(1/3)
+(0/3)
Replication of HEV in multiple tissues, including liver, spleen, kidney and colon, has been reported in both pigs and nude mice when inoculated with HEV [21, 26]. In order to further analyze the replication of HEV in different tissues, HEV genome RNA and capsid protein (ORF2) were analyzed by RT-nPCR, qRT-PCR and Western blotting, respectively. In the present study, HEV RNA was detected with a high viral titer in the liver (6.2 × 105 ± 5.8 × 104 copies/g), spleen (1.3 × 104 ± 3.7 × 104 copies/g), kidney (7.1 × 104 ± 3.1 × 104 copies/g) and colon (1.9 × 104 ± 5.8 × 103 copies/g) of mice inoculated with HEV RNA transcripts at 28 dpi (Fig. 3c). HEV RNA (both positive and negative strands) were detected in the liver and spleen in mice inoculated with HEV RNA transcripts at 28 dpi (Table 2). Although colon has been reported as a replicate site of HEV [21], only one mouse inoculated with HEV RNA transcripts at 28 dpi was found to be positive (Table 2). Meanwhile, the capsid protein of HEV (ORF2) were detected in the liver, spleen and kidney of mice inoculated with HEV RNA transcripts (Fig. 3d-g) by Western blotting. Shedding viruses in the feces, detected HEV RNA antigens in the serum and tissues indicated that HEV RNA transcripts was infectious in BALB/c mice.

BALB/c mice produce infectious HEV viruses upon inoculation of capped RNA transcripts of the swine HEV clone

To confirm whether BALB/c mice can produce infectious HEV viruses, we attempted to infect A549 cells with the recovered viruses from mice inoculated with capped RNA transcripts of the swine HEV clone. A549 cells were harvested 72 h after infection. HEV capsid protein was detected by Western blotting (Fig. 4) in cells infected with the feces of BALB/c mice inoculated with HEV RNA transcripts. This result confirmed that the inoculated BALB/c mice can produce infectious HEV, which can replicate in A549 cells.

Capped RNA transcripts of pGEM-HEV provokes humoral response in BALB/c mice

In response to HEV infection, human body develops antibodies to HEV (IgM and IgG). To investigate whether BALB/c mice provoke similar response to RNA transcripts of HEV infection, we measured serum anti-HEV IgM and IgG. An increased anti-HEV IgM and IgG levels were observed in all the mice inoculated with HEV RNA transcripts compared with the control group (Fig. 5). In the RNA transcripts inoculated group, the increased IgM level reached the pink at 21 dpi. IgG began to increase at 21 dpi, and continued to increase until the end of the experiment. These results demonstrated that infection of the swine HEV infectious clone stimulated humoral response in BALB/c mice.

Infection of capped HEV RNA transcripts induces liver inflammation in BALB/c mice

Hepatitis is swelling and inflammation of the liver, and most commonly caused by hepatitis virus infections. To determine whether infection of capped HEV RNA transcripts can also cause liver injury/inflammation in BALB/c mice, we first tested the widely used liver enzymes for detecting liver damage. The levels/activities were characterized using an automated biochemistry analyzer. To our surprise, the elevated ALT, AST and ALP levels/activities in BALB/c inoculated with HEV RNA transcripts was very similar to the clinical feature of HEV infected patients (Fig. 6a, b and c).
Furthermore, histopathologic examination showed swelling of liver cells and liver capsules were filled with inflammatory exudates and liver hemorrhage in mice inoculated with RNA transcripts of HEV (Fig. 6d). Increased infiltrating lymphocytes and macrophages were observed in these mice. In contrast, no damage was observed in any of the control tissues. These results indicate that infection of swine HEV RNA transcripts triggered liver inflammation in BALB/c mice.

Discussions

Lessons from the research of hepatitis B and C viruses have taught us the importance of developing robust experimental models for understanding their infection biology, pathogenesis, as well as antiviral drug development. However, the restricted tropism of host and cell type for hepatitis virus infection has always challenged the development of experimental models. Despite the recent establishment of several cell culture systems of HEV [27], animal models are urgently need for studying HEV pathogenicity and antiviral drug development.
Non-human primates have been successfully employed for HEV research in early days [28, 29], but non-human primates are not available for research anymore, because of the ethics. Although swine is the mainly reservoir of HEV, it is not suitable as an experimental model for HEV because of its large body size. In contrast, mice are the most commonly used small animal for research purpose. This study, we reported the successful establishment of HEV infection in BALB/c mice with an infectious swine HEV clone generated by reverse genetics approach.
In fact, we have previously attempted to establish HEV mouse model using BALB/c nude mice [21]. Because nude mice lack the thymus and are therefore unable to produce T-cells. We thought that the immunodeficient nature of these mice could be an advantage of permitting HEV infection. Indeed, we have succeeded the infection in BALB/c nude mice with genotype 4 swine HEV strain [21]. However, the immunodeficient property in turn hampered the study of HEV pathogenesis. Because hepatitis is an inflammatory process mainly mediated by host immune response. Furthermore, these mice have a strict life condition and limited fertility, which also hampers the further application. To circumstance these limitations, we now have succeeded with infecting the regular BALB/c mice, which have a competent immune system. In the present study, the RNA transcripts of infectious cDNA clone of swine HEV was successfully infected BALB/c mice. HEV RNA was detected at 3 dpi in feces, which is similar to our genotype 4 swine HEV strain inoculated into BALB/c nude mice [21], but earlier than that inoculated pigs with genotype 4 swine HEV RNA transcripts (7 dpi) [20] and Sprague-Dawley rat (SD rat) inoculated with genotype 4 swine HEV RNA transcripts [30]. HEV produces an intermediate negative-strand RNA when it replicates. The negative-strand of HEV was detectable at 7 dpi in serum of all mice inoculated with RNA transcripts. Furthermore, the negative-strand was found in the HEV replicated sites, including liver (3/3), spleen (3/3), kidney (2/3) and colon (1/3). Moreover, HEV RNA was detected in the feces, serum and tissues (liver, spleen, kidney and colon). Shedding HEV in feces, and detection of negative-strand RNA in liver, spleen and kidney firmly demonstrated the infectivity of the genotype 4 swine HEV pGEM-HEV cDNA clone is infectious in BALB/c mice, although previous study reported that C57BL/6 mice is not permissive for HEV infection [31]. More interestingly, we observed humoral response and liver injury in infected BALB/c mice, indicating that this model is promising for studying HEV pathogenesis.
Although HEV infects BALB/c mice has been successfully established, key parameters should be determined in the future, such as identification of infected cell populations, observation of inflammatory manifestation, and then explicit comparisons with HEV infected patients to assess potential for clinical relevance. The establishment of HEV infection based on BALB/c mice is benefit to screen anti-HEV drugs and develop vaccines in vivo.

Conclusion

In this study, we successfully established BALB/c mice-based animal model with an infectious cDNA clone of genotype 4 swine HEV constructed by reverse genetics approach. This swine HEV clone strain is capable of modeling HEV infection, and studying host response and pathogenesis. Furthermore, this model also bears importantly implications for future anti-HEV drug development.

Acknowledgements

Authors thank Xiancheng Zhao for her contribution on HEV full-length genome amplification, and Ping Yan for his contribution on sample collection.

Funding

This study was supported by National Natural Science Foundation of China (grant number 31360619 and 81660338 to Fen Huang), Natural Science Foundation of Yunnan province (grant number 2017FA036 to Fen Huang and 2018FB132 to Wenhai Yu), Fundamental Research Funds for the Central Universities (grant number 2016ZX310179) and PUMC Youth Fund (grant number 2017310038) to Wenhai Yu. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Ethics approval

The animal protocol was approved by the Animal Care and Use Committee (ACUC) of the Kunming University of Science and Technology.
Not applicable

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Smith DB, Simmonds P, Jameel S, Emerson SU, Harrison TJ, Meng XJ, Okamoto H, Van der Poel WH, Purdy MA. Consensus proposals for classification of the family Hepeviridae. J Gen Virol. 2014;95(Pt 10):2223–32.CrossRef Smith DB, Simmonds P, Jameel S, Emerson SU, Harrison TJ, Meng XJ, Okamoto H, Van der Poel WH, Purdy MA. Consensus proposals for classification of the family Hepeviridae. J Gen Virol. 2014;95(Pt 10):2223–32.CrossRef
2.
Zurück zum Zitat Billam P, Sun ZF, Meng XJ. Analysis of the complete genomic sequence of an apparently avirulent strain of avian hepatitis E virus (avian HEV) identified major genetic differences compared with the prototype pathogenic strain of avian HEV. J Gen Virol. 2007;88(Pt 5):1538–44.CrossRef Billam P, Sun ZF, Meng XJ. Analysis of the complete genomic sequence of an apparently avirulent strain of avian hepatitis E virus (avian HEV) identified major genetic differences compared with the prototype pathogenic strain of avian HEV. J Gen Virol. 2007;88(Pt 5):1538–44.CrossRef
3.
Zurück zum Zitat Panda SK, Nanda SK, Zafrullah M, Ansari IH, Ozdener MH, Jameel S. An Indian strain of hepatitis E virus (HEV): cloning, sequence, and expression of structural region and antibody responses in sera from individuals from an area of high-level HEV endemicity. J Clin Microbiol. 1995;33(10):2653–9.PubMedPubMedCentral Panda SK, Nanda SK, Zafrullah M, Ansari IH, Ozdener MH, Jameel S. An Indian strain of hepatitis E virus (HEV): cloning, sequence, and expression of structural region and antibody responses in sera from individuals from an area of high-level HEV endemicity. J Clin Microbiol. 1995;33(10):2653–9.PubMedPubMedCentral
4.
Zurück zum Zitat Huang YW, Opriessnig T, Halbur PG, Meng XJ. Initiation at the third in-frame AUG codon of open reading frame 3 of the hepatitis E virus is essential for viral infectivity in vivo. J Virol. 2007;81(6):3018–26.CrossRef Huang YW, Opriessnig T, Halbur PG, Meng XJ. Initiation at the third in-frame AUG codon of open reading frame 3 of the hepatitis E virus is essential for viral infectivity in vivo. J Virol. 2007;81(6):3018–26.CrossRef
5.
Zurück zum Zitat Tam AW, Smith MM, Guerra ME, Huang CC, Bradley DW, Fry KE, Reyes GR. Hepatitis E virus (HEV): molecular cloning and sequencing of the full-length viral genome. Virology. 1991;185(1):120–31.CrossRef Tam AW, Smith MM, Guerra ME, Huang CC, Bradley DW, Fry KE, Reyes GR. Hepatitis E virus (HEV): molecular cloning and sequencing of the full-length viral genome. Virology. 1991;185(1):120–31.CrossRef
6.
Zurück zum Zitat Meng XJ, Purcell RH, Halbur PG, Lehman JR, Webb DM, Tsareva TS, Haynes JS, Thacker BJ, Emerson SU. A novel virus in swine is closely related to the human hepatitis E virus. Proc Natl Acad Sci U S A. 1997;94(18):9860–5.CrossRef Meng XJ, Purcell RH, Halbur PG, Lehman JR, Webb DM, Tsareva TS, Haynes JS, Thacker BJ, Emerson SU. A novel virus in swine is closely related to the human hepatitis E virus. Proc Natl Acad Sci U S A. 1997;94(18):9860–5.CrossRef
7.
Zurück zum Zitat Hsieh SY, Meng XJ, Wu YH, Liu ST, Tam AW, Lin DY, Liaw YF. Identity of a novel swine hepatitis E virus in Taiwan forming a monophyletic group with Taiwan isolates of human hepatitis E virus. J Clin Microbiol. 1999;37(12):3828–34.PubMedPubMedCentral Hsieh SY, Meng XJ, Wu YH, Liu ST, Tam AW, Lin DY, Liaw YF. Identity of a novel swine hepatitis E virus in Taiwan forming a monophyletic group with Taiwan isolates of human hepatitis E virus. J Clin Microbiol. 1999;37(12):3828–34.PubMedPubMedCentral
8.
Zurück zum Zitat Feagins AR, Opriessnig T, Huang YW, Halbur PG, Meng XJ. Cross-species infection of specific-pathogen-free pigs by a genotype 4 strain of human hepatitis E virus. J Med Virol. 2008;80(8):1379–86.CrossRef Feagins AR, Opriessnig T, Huang YW, Halbur PG, Meng XJ. Cross-species infection of specific-pathogen-free pigs by a genotype 4 strain of human hepatitis E virus. J Med Virol. 2008;80(8):1379–86.CrossRef
9.
Zurück zum Zitat Feagins AR, Opriessnig T, Guenette DK, Halbur PG, Meng XJ. Detection and characterization of infectious hepatitis E virus from commercial pig livers sold in local grocery stores in the USA. J Gen Virol. 2007;88(Pt 3):912–7.CrossRef Feagins AR, Opriessnig T, Guenette DK, Halbur PG, Meng XJ. Detection and characterization of infectious hepatitis E virus from commercial pig livers sold in local grocery stores in the USA. J Gen Virol. 2007;88(Pt 3):912–7.CrossRef
10.
Zurück zum Zitat Tei S, Kitajima N, Takahashi K, Mishiro S. Zoonotic transmission of hepatitis E virus from deer to human beings. Lancet. 2003;362(9381):371–3.CrossRef Tei S, Kitajima N, Takahashi K, Mishiro S. Zoonotic transmission of hepatitis E virus from deer to human beings. Lancet. 2003;362(9381):371–3.CrossRef
11.
Zurück zum Zitat Rein DB, Stevens GA, Theaker J, Wittenborn JS, Wiersma ST. The global burden of hepatitis E virus genotypes 1 and 2 in 2005. Hepatology. 2012;55(4):988–97.CrossRef Rein DB, Stevens GA, Theaker J, Wittenborn JS, Wiersma ST. The global burden of hepatitis E virus genotypes 1 and 2 in 2005. Hepatology. 2012;55(4):988–97.CrossRef
12.
Zurück zum Zitat Dalton HR, Bendall R, Ijaz S, Banks M. Hepatitis E: an emerging infection in developed countries. Lancet Infect Dis. 2008;8(11):698–709.CrossRef Dalton HR, Bendall R, Ijaz S, Banks M. Hepatitis E: an emerging infection in developed countries. Lancet Infect Dis. 2008;8(11):698–709.CrossRef
13.
Zurück zum Zitat Aggarwal R, Krawczynski K. Hepatitis E: an overview and recent advances in clinical and laboratory research. J Gastroenterol Hepatol. 2000;15(1):9–20.CrossRef Aggarwal R, Krawczynski K. Hepatitis E: an overview and recent advances in clinical and laboratory research. J Gastroenterol Hepatol. 2000;15(1):9–20.CrossRef
14.
Zurück zum Zitat Khuroo MS, Kamili S. Aetiology, clinical course and outcome of sporadic acute viral hepatitis in pregnancy. J Viral Hepat. 2003;10(1):61–9.CrossRef Khuroo MS, Kamili S. Aetiology, clinical course and outcome of sporadic acute viral hepatitis in pregnancy. J Viral Hepat. 2003;10(1):61–9.CrossRef
15.
Zurück zum Zitat Kumar A, Beniwal M, Kar P, Sharma JB, Murthy NS. Hepatitis E in pregnancy. Int J Gynaecol Obstet. 2004;85(3):240–4.CrossRef Kumar A, Beniwal M, Kar P, Sharma JB, Murthy NS. Hepatitis E in pregnancy. Int J Gynaecol Obstet. 2004;85(3):240–4.CrossRef
16.
Zurück zum Zitat Shukla P, Nguyen HT, Torian U, Engle RE, Faulk K, Dalton HR, Bendall RP, Keane FE, Purcell RH, Emerson SU. Cross-species infections of cultured cells by hepatitis E virus and discovery of an infectious virus-host recombinant. Proc Natl Acad Sci U S A. 2011;108(6):2438–43.CrossRef Shukla P, Nguyen HT, Torian U, Engle RE, Faulk K, Dalton HR, Bendall RP, Keane FE, Purcell RH, Emerson SU. Cross-species infections of cultured cells by hepatitis E virus and discovery of an infectious virus-host recombinant. Proc Natl Acad Sci U S A. 2011;108(6):2438–43.CrossRef
17.
Zurück zum Zitat Shukla P, Nguyen HT, Faulk K, Mather K, Torian U, Engle RE, Emerson SU. Adaptation of a genotype 3 hepatitis E virus to efficient growth in cell culture depends on an inserted human gene segment acquired by recombination. J Virol. 2012;86(10):5697–707.CrossRef Shukla P, Nguyen HT, Faulk K, Mather K, Torian U, Engle RE, Emerson SU. Adaptation of a genotype 3 hepatitis E virus to efficient growth in cell culture depends on an inserted human gene segment acquired by recombination. J Virol. 2012;86(10):5697–707.CrossRef
18.
Zurück zum Zitat Tanaka T, Takahashi M, Takahashi H, Ichiyama K, Hoshino Y, Nagashima S, Mizuo H, Okamoto H. Development and characterization of a genotype 4 hepatitis E virus cell culture system using a HE-JF5/15F strain recovered from a fulminant hepatitis patient. J Clin Microbiol. 2009;47(6):1906–10.CrossRef Tanaka T, Takahashi M, Takahashi H, Ichiyama K, Hoshino Y, Nagashima S, Mizuo H, Okamoto H. Development and characterization of a genotype 4 hepatitis E virus cell culture system using a HE-JF5/15F strain recovered from a fulminant hepatitis patient. J Clin Microbiol. 2009;47(6):1906–10.CrossRef
19.
Zurück zum Zitat Huang YW, Haqshenas G, Kasorndorkbua C, Halbur PG, Emerson SU, Meng XJ. Capped RNA transcripts of full-length cDNA clones of swine hepatitis E virus are replication competent when transfected into Huh7 cells and infectious when intrahepatically inoculated into pigs. J Virol. 2005;79(3):1552–8.CrossRef Huang YW, Haqshenas G, Kasorndorkbua C, Halbur PG, Emerson SU, Meng XJ. Capped RNA transcripts of full-length cDNA clones of swine hepatitis E virus are replication competent when transfected into Huh7 cells and infectious when intrahepatically inoculated into pigs. J Virol. 2005;79(3):1552–8.CrossRef
20.
Zurück zum Zitat Cordoba L, Feagins AR, Opriessnig T, Cossaboom CM, Dryman BA, Huang YW, Meng XJ. Rescue of a genotype 4 human hepatitis E virus from cloned cDNA and characterization of intergenotypic chimeric viruses in cultured human liver cells and in pigs. J Gen Virol. 2012;93(Pt 10):2183–94.CrossRef Cordoba L, Feagins AR, Opriessnig T, Cossaboom CM, Dryman BA, Huang YW, Meng XJ. Rescue of a genotype 4 human hepatitis E virus from cloned cDNA and characterization of intergenotypic chimeric viruses in cultured human liver cells and in pigs. J Gen Virol. 2012;93(Pt 10):2183–94.CrossRef
21.
Zurück zum Zitat Huang F, Zhang W, Gong G, Yuan C, Yan Y, Yang S, Cui L, Zhu J, Yang Z, Hua X. Experimental infection of Balb/c nude mice with hepatitis E virus. BMC Infect Dis. 2009;9:93.CrossRef Huang F, Zhang W, Gong G, Yuan C, Yan Y, Yang S, Cui L, Zhu J, Yang Z, Hua X. Experimental infection of Balb/c nude mice with hepatitis E virus. BMC Infect Dis. 2009;9:93.CrossRef
22.
Zurück zum Zitat Fu H, Wang L, Zhu Y, Geng J, Li L, Wang X, Bu Q, Zhuang H. Analysing complete genome sequence of swine hepatitis E virus (HEV), strain CHN-XJSW13 isolated from Xinjiang, China: putative host range, and disease severity determinants in HEV. Infect Genet Evol. 2011;11(3):618–23.CrossRef Fu H, Wang L, Zhu Y, Geng J, Li L, Wang X, Bu Q, Zhuang H. Analysing complete genome sequence of swine hepatitis E virus (HEV), strain CHN-XJSW13 isolated from Xinjiang, China: putative host range, and disease severity determinants in HEV. Infect Genet Evol. 2011;11(3):618–23.CrossRef
23.
Zurück zum Zitat Huang FF, Haqshenas G, Guenette DK, Halbur PG, Schommer SK, Pierson FW, Toth TE, Meng XJ. Detection by reverse transcription-PCR and genetic characterization of field isolates of swine hepatitis E virus from pigs in different geographic regions of the United States. J Clin Microbiol. 2002;40(4):1326–32.CrossRef Huang FF, Haqshenas G, Guenette DK, Halbur PG, Schommer SK, Pierson FW, Toth TE, Meng XJ. Detection by reverse transcription-PCR and genetic characterization of field isolates of swine hepatitis E virus from pigs in different geographic regions of the United States. J Clin Microbiol. 2002;40(4):1326–32.CrossRef
24.
Zurück zum Zitat Nanda SK, Panda SK, Durgapal H, Jameel S. Detection of the negative strand of hepatitis E virus RNA in the livers of experimentally infected rhesus monkeys: evidence for viral replication. J Med Virol. 1994;42(3):237–40.CrossRef Nanda SK, Panda SK, Durgapal H, Jameel S. Detection of the negative strand of hepatitis E virus RNA in the livers of experimentally infected rhesus monkeys: evidence for viral replication. J Med Virol. 1994;42(3):237–40.CrossRef
25.
Zurück zum Zitat Baylis SA, Blumel J, Mizusawa S, Matsubayashi K, Sakata H, Okada Y, Nubling CM, Hanschmann KM. World Health Organization international standard to harmonize assays for detection of hepatitis E virus RNA. Emerg Infect Dis. 2013;19(5):729–35.CrossRef Baylis SA, Blumel J, Mizusawa S, Matsubayashi K, Sakata H, Okada Y, Nubling CM, Hanschmann KM. World Health Organization international standard to harmonize assays for detection of hepatitis E virus RNA. Emerg Infect Dis. 2013;19(5):729–35.CrossRef
26.
Zurück zum Zitat Huang F, Hua X, Yang S, Yuan C, Zhang W. Effective inhibition of hepatitis E virus replication in A549 cells and piglets by RNA interference (RNAi) targeting RNA-dependent RNA polymerase. Antivir Res. 2009;83(3):274–81.CrossRef Huang F, Hua X, Yang S, Yuan C, Zhang W. Effective inhibition of hepatitis E virus replication in A549 cells and piglets by RNA interference (RNAi) targeting RNA-dependent RNA polymerase. Antivir Res. 2009;83(3):274–81.CrossRef
27.
Zurück zum Zitat Okamoto H. Hepatitis E virus cell culture models. Virus Res. 2011;161(1):65–77.CrossRef Okamoto H. Hepatitis E virus cell culture models. Virus Res. 2011;161(1):65–77.CrossRef
28.
Zurück zum Zitat Aggarwal R, Kamili S, Spelbring J, Krawczynski K. Experimental studies on subclinical hepatitis E virus infection in cynomolgus macaques. J Infect Dis. 2001;184(11):1380–5.CrossRef Aggarwal R, Kamili S, Spelbring J, Krawczynski K. Experimental studies on subclinical hepatitis E virus infection in cynomolgus macaques. J Infect Dis. 2001;184(11):1380–5.CrossRef
29.
Zurück zum Zitat Panda SK, Ansari IH, Durgapal H, Agrawal S, Jameel S. The in vitro-synthesized RNA from a cDNA clone of hepatitis E virus is infectious. J Virol. 2000;74(5):2430–7.CrossRef Panda SK, Ansari IH, Durgapal H, Agrawal S, Jameel S. The in vitro-synthesized RNA from a cDNA clone of hepatitis E virus is infectious. J Virol. 2000;74(5):2430–7.CrossRef
30.
Zurück zum Zitat Zhu Y, Yu X, Zhang Y, Ni Y, Si F, Yu R, Dong S, Huang Y, Li Z. Infectivity of a genotype 4 hepatitis E virus cDNA clone by intrahepatic inoculation of laboratory rats. Vet Microbiol. 2013;166(3–4):405–11.CrossRef Zhu Y, Yu X, Zhang Y, Ni Y, Si F, Yu R, Dong S, Huang Y, Li Z. Infectivity of a genotype 4 hepatitis E virus cDNA clone by intrahepatic inoculation of laboratory rats. Vet Microbiol. 2013;166(3–4):405–11.CrossRef
31.
Zurück zum Zitat Li TC, Suzaki Y, Ami Y, Tsunemitsu H, Miyamura T, Takeda N. Mice are not susceptible to hepatitis E virus infection. J Vet Med Sci. 2008;70(12):1359–62.CrossRef Li TC, Suzaki Y, Ami Y, Tsunemitsu H, Miyamura T, Takeda N. Mice are not susceptible to hepatitis E virus infection. J Vet Med Sci. 2008;70(12):1359–62.CrossRef
Metadaten
Titel
Successful infection of BALB/c mice by a swine hepatitis E virus clone constructed with reverse genetics
verfasst von
Wenhai Yu
Chenchen Yang
Xianhui Hao
Tianwu Ma
Fen Huang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Infectious Diseases / Ausgabe 1/2018
Elektronische ISSN: 1471-2334
DOI
https://doi.org/10.1186/s12879-018-3544-4

Weitere Artikel der Ausgabe 1/2018

BMC Infectious Diseases 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Reizdarmsyndrom: Diäten wirksamer als Medikamente

29.04.2024 Reizdarmsyndrom Nachrichten

Bei Reizdarmsyndrom scheinen Diäten, wie etwa die FODMAP-arme oder die kohlenhydratreduzierte Ernährung, effektiver als eine medikamentöse Therapie zu sein. Das hat eine Studie aus Schweden ergeben, die die drei Therapieoptionen im direkten Vergleich analysierte.

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.