Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Int J Food Microbiol ; 339: 109013, 2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33340943

ABSTRACT

Hepatitis E virus (HEV) is the causative agent of acute and chronic hepatitis in humans. The zoonotic HEV genotype 3 is the main genotype in Europe. The foodborne transmission via consumption of meat and meat products prepared from infected pigs or wild boars is considered the major transmission route of this genotype. High hydrostatic pressure processing (HPP) is a technique, which can be used for inactivation of pathogens in food. Here, preparations of a cell culture-adapted HEV genotype 3 strain in phosphate-buffered saline (PBS) were subjected to HPP and the remaining infectivity was titrated in cell culture by counting fluorescent foci of replicating virus. A gradual decrease in infectivity was found by application of 100 to 600 MPa for 2 min. At 20 °C, infectivity reduction of 0.5 log10 at 200 MPa and 1 log10 at 400 MPa were observed. Slightly higher infectivity reduction of 1 log10 at 200 MPa and 2 log10 at 400 MPa were found by application of the pressure at 4 °C. At both temperatures, the virus was nearly completely inactivated (>3.5 log10 infectivity decrease) at 600 MPa; however, low amounts of remaining infectious virus were observed in one of three replicates in both cases. Transmission electron microscopy showed disassembled and distorted particles in the preparations treated with 600 MPa. Time-course experiments at 400 MPa showed a continuous decline of infectivity from 30 s to 10 min, leading to a 2 log10 infectivity decrease at 20 °C and to a 2.5 log10 infectivity decrease at 4 °C for a 10 min pressure application each. Predictive models for inactivation of HEV by HPP were generated on the basis of the generated data. The results show that HPP treatment can reduce HEV infectivity, which is mainly dependent on pressure height and duration of the HPP treatment. Compared to other viruses, HEV appears to be relatively stable against HPP and high pressure/long time combinations have to be applied for significant reduction of infectivity.


Subject(s)
Food Microbiology , Hepatitis E virus/physiology , Hydrostatic Pressure , Meat Products/virology , Virus Inactivation , Animals , Europe , Hepatitis E/transmission , Hepatitis E/virology , Hepatitis E virus/ultrastructure , Humans , Meat/virology , Microscopy, Electron, Transmission , Models, Biological , Sus scrofa , Swine , Temperature
2.
Viruses ; 11(10)2019 09 20.
Article in English | MEDLINE | ID: mdl-31547135

ABSTRACT

Hepatitis E virus (HEV) infection is a major cause of acute hepatitis worldwide. It is transmitted enterically but replicates in the liver. Recent studies indicate that HEV exists in two forms: naked, nonenveloped virions that are shed into feces to mediate inter-host transmission, and membrane-cloaked, quasienveloped virions that circulate in the bloodstream to mediate virus spread within a host. Both virion types are infectious, but differ in the way they infect cells. Elucidating the entry mechanism for both virion types is essential to understand HEV biology and pathogenesis, and is relevant to the development of treatments and preventions for HEV. This review summarizes the current understanding of the cell entry mechanism for these two HEV virion types.


Subject(s)
Hepatitis E virus/physiology , Virus Internalization , Hepatitis E/metabolism , Hepatitis E/virology , Hepatitis E virus/ultrastructure , Humans , Lysosomes/metabolism , Lysosomes/virology , Receptors, Virus/metabolism , Virion/physiology , Virion/ultrastructure , Virus Uncoating
3.
Rev Med Virol ; 29(6): e2078, 2019 11.
Article in English | MEDLINE | ID: mdl-31456241

ABSTRACT

Hepatitis A (HAV) and E (HEV) viruses are able to cause liver disease in humans. Among the five classical hepatotropic viruses, they are mainly transmitted via the fecal-oral route. Historically, many similarities have thus been described between them according to their incidence and their pathogenicity, especially in countries with poor sanitary conditions. However, recent advances have provided new insights, and the gap is widening between them. Indeed, while HAV infection incidence tends to decrease in developed countries along with public health improvement, HEV is currently considered as an underdiagnosed emerging pathogen. HEV autochthonous infections are increasingly observed and are mainly associated with zoonotic transmissions. Extra hepatic signs resulting in neurological or renal impairments have also been reported for HEV, as well as a chronic carrier state in immunocompromised patients, arguing in favor of differential pathogenesis between those two viruses. Recent molecular tools have allowed studies of viral genome variability and investigation of links between viral plasticity and clinical evolution. The identification of key functional mutations in viral genomes may improve the knowledge of their clinical impact and is analyzed in depth in the present review.


Subject(s)
Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Hepatitis A virus , Hepatitis A/epidemiology , Hepatitis A/virology , Hepatitis E virus , Hepatitis E/epidemiology , Hepatitis E/virology , Communicable Diseases, Emerging/diagnosis , Communicable Diseases, Emerging/transmission , Genetic Variation , Genotype , Geography, Medical , Global Health , Hepatitis A/diagnosis , Hepatitis A/transmission , Hepatitis A virus/physiology , Hepatitis A virus/ultrastructure , Hepatitis E/diagnosis , Hepatitis E/transmission , Hepatitis E virus/physiology , Hepatitis E virus/ultrastructure , Humans , Phenotype , Phylogeography
4.
Vaccine ; 36(16): 2104-2111, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29544686

ABSTRACT

Hepatitis E virus (HEV) is associated with acute hepatitis disease. Numerous truncated HEV capsid proteins have been successfully expressed using different expression systems. Among these, p495, a protein truncated at its N- and C-termini by 111 and 54 amino acids (aa), respectively (HEV ORF2 aa 112-606) can self-assemble into T = 1 virus-like particles (VLPs) when expressed by insect cells. A shorter p239 (aa 368-606) protein is a particulate antigen that we have previously used in our commercialized HEV vaccine, Hecolin. Here, we sought to express p495 in its soluble form (named Ep495) in E. coli and in baculovirus-infected Tn5 insect cells (named BTp495) as a back-to-back control. Characterization of p495 particles derived from these two expression systems showed similarities in particle size, morphology, and sedimentation coefficient. Antigenicity assays using a panel of anti-HEV monoclonal antibodies also showed similar strong reactivities for Ep495 and BTp495, as well as similar binding profiles that were congruent with p239. Mouse immunization results showed that Ep495 particles had comparable immunogenicity with that of BTp495 VLPs, as well as p239. Overall, our findings suggest that p495 particles produced in E. coli are ideal for the development of next-generation prophylactic vaccines against hepatitis E.


Subject(s)
Capsid Proteins/genetics , Capsid Proteins/immunology , Gene Expression , Hepatitis E virus/immunology , Recombinant Proteins , Vaccines, Virus-Like Particle , Antigens, Viral/immunology , Capsid Proteins/isolation & purification , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Hepatitis E virus/genetics , Hepatitis E virus/ultrastructure , Immunogenicity, Vaccine , Protein Binding , Protein Multimerization , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/ultrastructure , Viral Hepatitis Vaccines/immunology
5.
Viruses ; 8(8)2016 08 18.
Article in English | MEDLINE | ID: mdl-27548201

ABSTRACT

Hepatitis E virus (HEV), an enterically transmitted hepatotropic virus, was thought to be non-enveloped for decades. However, recent studies have revealed that the virus circulating in the patient's blood is completely cloaked in host membranes and resistant to neutralizing antibodies. The discovery of this novel enveloped form of HEV has raised a series of questions about the fundamental biology of HEV and the way this virus, which has been understudied in the past, interacts with its host. Here, we review recent advances towards understanding this phenomenon and discuss its potential impact on various aspects of the HEV life cycle and immunity.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E virus/ultrastructure , Host-Pathogen Interactions , Viral Structures , Virus Assembly , Animals , Humans , Models, Biological
6.
Emerg Infect Dis ; 19(2): 264-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23347828

ABSTRACT

We investigated viability of hepatitis E virus (HEV) identified in contaminated pork liver sausages obtained from France. HEV replication was demonstrated in 1 of 4 samples by using a 3-dimensional cell culture system. The risk for human infection with HEV by consumption of these sausages should be considered to be high.


Subject(s)
Hepatitis E virus/physiology , Meat Products/virology , Virion/physiology , Animals , Cell Line, Tumor , Food Microbiology , France , Hepatitis E virus/genetics , Hepatitis E virus/ultrastructure , Humans , Liver , Molecular Typing , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sus scrofa , Virion/genetics , Virion/ultrastructure , Virus Replication
7.
J Virol Methods ; 187(2): 327-32, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23159673

ABSTRACT

Hepatitis E is an acute, viral hepatitis epidemic in developing regions, but which is detected with increasing frequency in sporadic form in developed regions. Pigs and possibly some other mammals are considered reservoirs of zoonotic infection with hepatitis E virus (HEV). However, whilst the relative significance of potential transmission routes from pigs to people is still unclear, the consumption of raw or undercooked pig meat has been implicated as a source of HEV infection. The lack of information about HEV zoonotic transmission is due in part to the difficulties of in vitro propagation of HEV. The Rotating Wall Vessel (RVW) has been described as a useful tool for the culture of cell lines in a 3-dimensional (3D) configuration. The aim of this work was to develop a 3D cell culture system for HEV to facilitate studies into the viability of virions contaminating pig tissues. This study, demonstrated that HEV can replicate efficiently in the RWV in human hepatoblastoma PLC/PRF/5 cells for up to 5 months not only by real time RT-PCR but also by detection of complete virions via electron microscopy. Furthermore, the replication of HEV progeny was observed by detecting HEV RNA by RT-PCR. The progeny were able to infect fresh 3D cultures, showing that this method is able to produce infectious hepatitis E virions.


Subject(s)
Hepatitis E virus/physiology , Virus Replication , Cell Culture Techniques , Cell Line, Tumor , Hepatitis E virus/genetics , Hepatitis E virus/ultrastructure , Hepatocytes/virology , Humans , Microscopy, Electron , RNA, Viral/analysis , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Virus Cultivation/methods
8.
J Mol Biol ; 422(5): 650-658, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22743104

ABSTRACT

Human astroviruses (HAstVs) are a major cause of gastroenteritis. HAstV assembles from the structural protein VP90 and undergoes a cascade of proteolytic cleavages. Cleavage to VP70 is required for release of immature particles from cells, and subsequent cleavage by trypsin confers infectivity. We used electron cryomicroscopy and icosahedral image analysis to determine the first experimentally derived, three-dimensional structures of an immature VP70 virion and a fully proteolyzed, infectious virion. Both particles display T=3 icosahedral symmetry and nearly identical solid capsid shells with diameters of ~350Å. Globular spikes emanate from the capsid surface, yielding an overall diameter of ~440Å. While the immature particles display 90 dimeric spikes, the mature capsid only displays 30 spikes, located on the icosahedral 2-fold axes. Loss of the 60 peripentonal spikes likely plays an important role in viral infectivity. In addition, immature HAstV bears a striking resemblance to the structure of hepatitis E virus (HEV)-like particles, as previously predicted from structural similarity of the crystal structure of the astrovirus spike domain with the HEV P-domain [Dong, J., Dong, L., Méndez, E. & Tao, Y. (2011). Crystal structure of the human astrovirus capsid spike. Proc. Natl. Acad. Sci. USA108, 12681-12686]. Similarities between their capsid shells and dimeric spikes and between the sequences of their capsid proteins suggest that these viral families are phylogenetically related and may share common assembly and activation mechanisms.


Subject(s)
Mamastrovirus/ultrastructure , Virion/ultrastructure , Amino Acid Sequence , Cryoelectron Microscopy , Hepatitis E virus/ultrastructure , Humans , Image Processing, Computer-Assisted , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA
10.
J Biol Chem ; 285(43): 33175-33183, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20720013

ABSTRACT

Hepatitis E virus (HEV) induces acute hepatitis in humans with a high fatality rate in pregnant women. There is a need for anti-HEV research to understand the assembly process of HEV native capsid. Here, we produced a large virion-sized and a small T=1 capsid by expressing the HEV capsid protein in insect cells with and without the N-terminal 111 residues, respectively, for comparative structural analysis. The virion-sized capsid demonstrates a T=3 icosahedral lattice and contains RNA fragment in contrast to the RNA-free T=1 capsid. However, both capsids shared common decameric organization. The in vitro assembly further demonstrated that HEV capsid protein had the intrinsic ability to form decameric intermediate. Our data suggest that RNA binding is the extrinsic factor essential for the assembly of HEV native capsids.


Subject(s)
Capsid Proteins/metabolism , Capsid/metabolism , Hepatitis E virus/physiology , RNA, Viral/metabolism , Virus Assembly/physiology , Animals , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cell Line , Female , Hepatitis E/mortality , Hepatitis E/virology , Hepatitis E virus/chemistry , Hepatitis E virus/ultrastructure , Humans , Moths , Pregnancy , Pregnancy Complications, Infectious/mortality , Pregnancy Complications, Infectious/virology , RNA, Viral/chemistry , RNA, Viral/genetics
11.
J Biomed Sci ; 14(5): 555-63, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17487571

ABSTRACT

Twenty-seven monoclonal antibodies (Mabs) recognizing the open reading frame 2 structural protein of the Pakistan strain of hepatitis E virus (HEV) were generated by conventional hybridoma technique. These Mabs were characterized by ELISA, affinity-capture reverse transcriptase-polymerase chain reaction (AC/RT-PCR), immune electron microscopy (IEM), and a RT-PCR based seroneutralization assay. Twenty-seven Mabs were positive by ELISA. By AC/RT-PCR, 24 Mabs bound to Pakistan and Namibia HEV strains. Thirteen Mabs were examined by IEM. Nine Mabs, positive by ELISA and AC/RT-PCR, bound and aggregated to Mexican HEV strain. We tested five Mabs that were positive by ELISA, AC/RT/PCR, and IEM by a RT-PCR based seroneutralization assay. Only one Mab (Mab 7) showed activity that inhibited the ability of HEV to attach to Alexander hepatoma cells (PLC-PRF-5). When Mab 7 was diluted to 1: 160, its inhibition activity persisted suggesting that Mab 7 might be a potential candidate for further evaluation in primates (passive protection experiments).


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Capsid Proteins/immunology , Hepatitis E virus/immunology , Animals , Antibodies, Monoclonal/ultrastructure , Capsid Proteins/genetics , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Hepatitis E virus/ultrastructure , Humans , Mice , Microscopy, Immunoelectron , Neutralization Tests , Open Reading Frames , Reverse Transcriptase Polymerase Chain Reaction
14.
World J Gastroenterol ; 10(11): 1583-8, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15162530

ABSTRACT

AIM: To select the peptide mimicking the neutralization epitope of hepatitis E virus which bound to non-type-specific and conformational monoclonal antibodies (mAbs) 8C11 and 8H3 fromed 7-peptide phage display library, and expressed the peptide recombinant with HBcAg in E.coli, and to observe whether the recombinant HBcAg could still form virus like particle (VLP) and to test the activation of the recombinant polyprotein and chemo-synthesized peptide that was selected by mAb 8H3. METHODS: 8C11 and 8H3 were used to screen for binding peptides through a 7-peptide phage display library. After 4 rounds of panning, monoclonal phages were selected and sequenced. The obtained dominant peptide coding sequences was then synthesized and inserted into amino acid 78 to 83 of hepatitis B core antigen (HBcAg), and then expressed in E.coli. Activity of the recombinant proteins was detected by Western blotting, VLPs of the recombinant polyproteins were tested by transmission electron microscopy and binding activity of the chemo-synthesized peptide was confirmed by BIAcore biosensor. RESULTS: Twenty-one positive monoclonal phages (10 for 8C11, and 11 for 8H3) were selected and the inserted fragments were sequenced. The DNA sequence coding for the obtained dominant peptides 8C11 (N'-His-Pro-Thr-Leu-Leu-Arg-Ile-C', named 8C11A) and 8H3 (N'-Ser-Ile-Leu-Pro- Tyr-Pro-Tyr-C', named 8H3A) were then synthesized and cloned to the HBcAg vector, then expressed in E.coli. The recombinant proteins aggregated into homodimer or polymer on SDS-PAGE, and could bind to mAb 8C11 and 8H3 in Western blotting. At the same time, the recombinant polyprotein could form virus like particles (VLPs), which could be visualized on electron micrograph. The dominant peptide 8H3A selected by mAb 8H3 was further chemo-synthesized, and its binding to mAb 8H3 could be detected by BIAcore biosensor. CONCLUSION: These results implicate that conformational neutralizing epitope can be partially modeled by a short peptide, which provides a feasible route for subunit vaccine development.


Subject(s)
Epitopes/genetics , Hepatitis E virus/genetics , Hepatitis E virus/immunology , Peptide Library , Viral Hepatitis Vaccines/genetics , Amino Acid Sequence , Antibodies, Monoclonal , Blotting, Western , Epitopes/chemistry , Epitopes/immunology , Hepatitis B Core Antigens/genetics , Hepatitis E virus/ultrastructure , Humans , Microscopy, Electron , Protein Conformation , Viral Hepatitis Vaccines/chemistry , Viral Hepatitis Vaccines/immunology , Virion/ultrastructure
16.
J Gen Virol ; 82(Pt 10): 2449-2462, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11562538

ABSTRACT

Hepatitis-splenomegaly (HS) syndrome is an emerging disease in chickens in North America; the cause of this disease is unknown. In this study, the genetic identification and characterization of a novel virus related to human hepatitis E virus (HEV) isolated from bile samples of chickens with HS syndrome is reported. Based upon the similar genomic organization and significant sequence identity of this virus with HEV, the virus has been tentatively named avian HEV in order to distinguish it from human and swine HEV. Electron microscopy revealed that avian HEV is a non-enveloped virus particle of 30-35 nm in diameter. The sequence of the 3' half of the viral genome ( approximately 4 kb) was determined. Sequence analyses revealed that this genomic region contains the complete 3' non-coding region, the complete genes from open reading frames (ORFs) 2 and 3, the complete RNA-dependent RNA polymerase (RdRp) gene and a partial helicase gene from ORF 1. The helicase gene is the most conserved gene between avian HEV and other HEV strains, displaying 58-61% aa and 57-60% nt sequence identities. The RdRp gene of avian HEV shares 47-50% aa and 52-53% nt sequence identities and the putative capsid gene (ORF 2) of avian HEV shares 48-49% aa and 48-51% nt sequence identities with the corresponding regions of other known HEV strains. Phylogenetic analysis indicates that avian HEV is genetically related to, but distinct from, other known HEV strains. This discovery has important implications for HEV animal models, nomenclature and natural history.


Subject(s)
Chickens/virology , Hepatitis E virus/classification , Poultry Diseases/virology , Splenomegaly/veterinary , 3' Untranslated Regions/chemistry , Amino Acid Sequence , Animals , Base Sequence , Hepatitis E virus/genetics , Hepatitis E virus/ultrastructure , Humans , Microscopy, Electron , Molecular Sequence Data , Open Reading Frames , Phylogeny , RNA, Viral/chemistry , Swine
17.
Vaccine ; 19(25-26): 3476-84, 2001 May 14.
Article in English | MEDLINE | ID: mdl-11348714

ABSTRACT

We evaluated the potential of recombinant hepatitis E virus (rHEV) virus-like particles (VLPs) as an oral immunogen by analyzing the response of serum IgM, IgG, and IgA and fecal IgA in mice after oral administration. The capsid proteins of HEV with its N-terminal 111 amino acids truncated were expressed with a recombinant baculovirus in insect cells, where the capsid proteins self-assembled into VLPs. Mice were orally inoculated four times with purified rHEV VLPs in concentrations ranging from 10 to 100 microg without adjuvant. Serum IgM response was obtained with as little as 10 microg of the VLPs, and the level reached its maximum in all mice groups within 2 weeks after the first administration. Serum IgG was detected by 4 weeks post-immunization (p.i.) in the majority of mice given doses of 50 and 100 microg and continuously increased at least until the 10 week mark. Serum IgA was also detected by 4 weeks p.i. in the majority of mice given doses of 50 and 100 microg, and the level reached the maximum at 8 weeks p.i. Furthermore, the maximum level of intestinal IgA responses was detected in the groups of mice receiving 50 and 100 microg rHEV VLPs at 8 weeks p.i. All these antibody responses were obtained without a mucosal adjuvant. We therefore concluded that oral immunization of rHEV VLPs is capable of inducing systemic as well as intestinal antibody responses. Furthermore, serum IgG and fecal IgA thus induced were reactive to the native HEV antigen, as determined by Western blot assays and antigen-capture ELISA.


Subject(s)
Hepatitis E virus/immunology , Viral Hepatitis Vaccines/administration & dosage , Administration, Oral , Animals , Female , Hepatitis Antibodies/biosynthesis , Hepatitis Antibodies/blood , Hepatitis E virus/ultrastructure , Immunity, Mucosal , Immunoglobulin A/biosynthesis , Immunoglobulin A/blood , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Injections, Intraperitoneal , Intestinal Mucosa/immunology , Kinetics , Mice , Mice, Inbred BALB C , Microscopy, Electron
18.
Braz J Infect Dis ; 4(5): 214-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11063553

ABSTRACT

Three new hepatitis viruses are reviewed; Hepatitis E Virus (HEV), Hepatitis G Virus (HGV), and Transfusion Transmitted Virus (TTV). Only HEV has been clearly associated with liver damage. It is transmitted by the fecal-oral route and tends to occur in poor socio-economic conditions. Acute disease is diagnosed by the presence of anti-HEV IgM antibodies in the serum. Hepatitis G virus and TTV are transmitted parenterally. HGV is the same agent as GBV-C. Although it is hepatotropic, and high levels of viremia may occur, pathogenicity to the liver has not been proven. TTV may also be transmitted by the fecal-oral route. It is abundant in liver tissue but, like HGV, pathogenicity has not been proven.


Subject(s)
DNA Virus Infections/virology , Flaviviridae , Hepatitis E virus , Hepatitis, Viral, Human/virology , Liver/pathology , Torque teno virus , DNA Virus Infections/pathology , Flaviviridae/genetics , Flaviviridae/pathogenicity , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/pathogenicity , Hepatitis E virus/physiology , Hepatitis E virus/ultrastructure , Hepatitis, Viral, Human/pathology , Humans , Liver/virology , Torque teno virus/genetics , Torque teno virus/isolation & purification , Torque teno virus/pathogenicity
19.
Virology ; 265(1): 35-45, 1999 Dec 05.
Article in English | MEDLINE | ID: mdl-10603315

ABSTRACT

The three-dimensional structure of a self-assembled, recombinant hepatitis E virus particle has been solved to 22-A resolution by cryo-electron microscopy and three-dimensional image reconstruction. The single subunit of 50 kDa is derived from a truncated version of the open reading frame-2 gene of the virus expressed in a baculovirus system. This is the first structure of a T = 1 particle with protruding dimers at the icosahedral two-fold axes solved by cryo-electron microscopy. The protein shell of these hollow particles extends from a radius of 50 A outward to a radius of 135 A. In the reconstruction, the capsid is dominated by dimers that define the 30 morphological units. The outer domain of the homodimer forms a protrusion, which corresponds to the spike-like density seen in the cryo-electron micrograph. This particle retains native virus epitopes, suggesting its potential value as a vaccine.


Subject(s)
Capsid/ultrastructure , Hepatitis E virus/ultrastructure , Cryoelectron Microscopy , Epitopes/ultrastructure , Image Processing, Computer-Assisted , Models, Molecular , Molecular Weight , Protein Conformation , Protein Folding , Recombinant Proteins/ultrastructure , Virion/ultrastructure
20.
Virus Res ; 62(1): 59-65, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10513287

ABSTRACT

Previously, we have described that injection of an expression vector containing hepatitis E virus (HEV) open reading frame 2 (HEV-ORF-2) generated a strong antibody response in mice. To characterize the reaction of this antiserum with native HEV and to evaluate its potential diagnostic application, we tested the antiserum's ability to bind HEV using immune electron microscope (IEM) and affinity-capture reverse transcription polymerase chain reaction (RT-PCR) amplification. Antiserum to ORF-2 aggregated HEV virions as seen by electron microscopy, providing direct evidence that ORF-2 encodes a structural protein. Antiserum also captured HEV for RT-PCR amplification. This antiserum bound HEV from diverse origins (Asia, Africa, Mexico) at virus concentrations found in patient fecal specimens and bile from inoculated non-human primates. The specificity of the affinity binding was demonstrated when pre-immune sera or sera collected from mice injected with control DNA vector (lacking the HEV ORF-2 gene) failed to bind HEV for RT-PCR amplification and IEM. Specific RT-PCR amplification was confirmed by restriction enzyme digestion of PCR products. The sensitivity of the binding was evaluated by RT-PCR amplification of serially diluted bile containing a genetically divergent HEV, Mexico'86. HEV was detected in a 10(-8) dilution of this bile. This is the first report that antibodies elicited by a DNA vaccine recognize native HEV. Our results indicate that ORF-2 encodes a structural protein and that antiserum to this protein enables simple, sensitive, and specific HEV detection by affinity-capture RT-PCR.


Subject(s)
Antibodies, Viral/immunology , Hepatitis E virus/immunology , Hepatitis E/virology , Reverse Transcriptase Polymerase Chain Reaction/methods , Vaccines, DNA/immunology , Animals , Haplorhini , Hepatitis E/immunology , Hepatitis E virus/isolation & purification , Hepatitis E virus/ultrastructure , Humans , Mice , Microscopy, Immunoelectron/methods , Reverse Transcriptase Polymerase Chain Reaction/standards
SELECTION OF CITATIONS
SEARCH DETAIL
...