Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
J Gen Virol ; 86(Pt 9): 2585-2593, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16099918

ABSTRACT

Hepatitis E virus (HEV), the causative agent of hepatitis E, is an important human pathogen. Increasing evidence indicates that hepatitis E is a zoonosis. Avian HEV was recently discovered in chickens with hepatitis-splenomegaly syndrome in the USA. Like swine HEV from pigs, avian HEV is also genetically and antigenically related to human HEV. The objective of this study was to construct and characterize an infectious cDNA clone of avian HEV for future studies of HEV replication and pathogenesis. Three full-length cDNA clones of avian HEV, pT7-aHEV-5, pT7G-aHEV-10 and pT7G-aHEV-6, were constructed and their infectivity was tested by in vitro transfection of leghorn male hepatoma (LMH) chicken liver cells and by direct intrahepatic inoculation of specific-pathogen-free (SPF) chickens with capped RNA transcripts from the three clones. The results showed that the capped RNA transcripts from each of the three clones were replication competent when transfected into LMH cells as demonstrated by detection of viral antigens with avian HEV-specific antibodies. SPF chickens intrahepatically inoculated with the capped RNA transcripts from each of the three clones developed active avian HEV infections as evidenced by seroconversion to avian HEV antibodies, viraemia and faecal virus shedding. The infectivity was further confirmed by successful infection of naïve chickens with the viruses recovered from chickens inoculated with the RNA transcripts. The results indicated that all three cDNA clones of avian HEV are infectious both in vitro and in vivo. The availability of these infectious clones for a chicken strain of HEV now affords an opportunity to study the mechanisms of HEV cross-species infection and tissue tropism by constructing chimeric viruses among human, swine and avian HEVs.


Subject(s)
Cloning, Molecular , DNA, Complementary/genetics , Hepatitis E virus/pathogenicity , Hepatitis E/veterinary , Hepatitis, Viral, Animal/physiopathology , Poultry Diseases/physiopathology , Animals , Cell Line, Tumor , Chickens , DNA, Complementary/metabolism , Feces/virology , Hepatitis E/physiopathology , Hepatitis E/virology , Hepatitis E virus/classification , Hepatitis E virus/genetics , Hepatitis, Viral, Animal/virology , Poultry Diseases/virology , RNA Caps/genetics , RNA Caps/metabolism , Transcription, Genetic , Virus Replication
2.
J Clin Microbiol ; 43(4): 1684-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15814985

ABSTRACT

Hepatitis E virus (HEV), the causative agent of hepatitis E, is an important public health concern in many developing countries. Increasing evidence indicates that hepatitis E is a zoonotic disease. There exist four major genotypes of HEV, and HEV isolates identified in samples from pigs belong to either genotype 3 or 4. Genotype 1 and 2 HEVs are found exclusively in humans. To determine whether genotype 1 and 2 HEVs also exist in pigs, a universal reverse transcription-PCR assay that is capable of detecting all four HEV genotypes was used to test for the presence of HEV RNA in serum and/or fecal samples from pigs in Thailand, where genotype 1 human HEV is prevalent, and from pigs in Mexico, where genotype 2 human HEV was epidemic. In Thailand, swine HEV RNA was detected in sera from 10/26 pigs of 2 to 4 months of age but not in sera from 50 pigs of other ages. In Mexico, swine HEV RNA was detected in 8/125 sera and 28/92 fecal samples from 2- to 4-month-old pigs. Antibodies to swine HEV were also detected in about 81% of the Mexican pigs. A total of 44 swine HEV isolates were sequenced for the open reading frame 2 gene region. Sequence analyses revealed that all swine HEV isolates identified in samples from pigs in Thailand and Mexico belong to genotype 3. Phylogenetic analyses revealed that minor branches associated with geographic origin exist among the swine HEV isolates. The results indicated that genotype 1 or 2 swine HEV does not exist in pigs from countries where the respective human HEV genotype 1 or 2 is prevalent. It is likely that only genotype 3 and 4 HEV strains have zoonotic potential.


Subject(s)
Blood/microbiology , Feces/virology , Hepatitis E virus/classification , Hepatitis E/veterinary , Swine Diseases/epidemiology , Animals , Genotype , Hepatitis E/epidemiology , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/isolation & purification , Humans , Mexico/epidemiology , Molecular Sequence Data , Prevalence , Sequence Analysis, DNA , Swine , Swine Diseases/virology , Thailand/epidemiology
3.
J Virol ; 79(6): 3429-37, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731237

ABSTRACT

Hepatitis E virus (HEV) is an important human pathogen. Due to the lack of a cell culture system and a practical animal model for HEV, little is known about its pathogenesis and replication. The discovery of a strain of HEV in chickens, designated avian HEV, prompted us to evaluate chickens as a model for the study of HEV. Eighty-five 60-week-old specific-pathogen-free chickens were randomly divided into three groups. Group 1 chickens (n=28) were each inoculated with 5 x 10(4.5) 50% chicken infectious doses of avian HEV by the oronasal route, group 2 chickens (n=29) were each inoculated with the same dose by the intravenous (i.v.) route, and group 3 chickens (n=28) were not inoculated and were used as controls. Two chickens from each group were necropsied at 1, 3, 5, 7, 10, 13, 16, 20, 24, 28, 35, and 42 days postinoculation (dpi), and the remaining chickens were necropsied at 56 dpi. Serum, fecal, and various tissue samples, including liver and spleen samples, were collected at each necropsy for pathological and virological testing. By 21 dpi, all oronasally and i.v. inoculated chickens had seroconverted. Fecal virus shedding was detected variably from 1 to 20 dpi for the i.v. group and from 10 to 56 dpi for the oronasal group. Avian HEV RNA was detected in serum, bile, and liver samples from both i.v. and oronasally inoculated chickens. Gross liver lesions, characterized by subcapsular hemorrhages or enlargement of the right intermediate lobe, were observed in 7 of 28 oronasally and 7 of 29 i.v. inoculated chickens. Microscopic liver lesions were mainly lymphocytic periphlebitis and phlebitis. The lesion scores were higher for oronasal (P=0.0008) and i.v. (P=0.0029) group birds than for control birds. Slight elevations of the plasma liver enzyme lactate dehydrogenase were observed in infected chickens. The results indicated that chickens are a useful model for studying HEV replication and pathogenesis. This is the first report of HEV transmission via its natural route in a homologous animal model.


Subject(s)
Chickens/virology , Disease Models, Animal , Hepatitis E virus/physiology , Hepatitis E/virology , Virus Replication , Animals , Bile/virology , Feces/virology , Hepatitis Antibodies/blood , Hepatitis E/pathology , Hepatitis E virus/genetics , Hepatitis E virus/immunology , L-Lactate Dehydrogenase/blood , Liver/pathology , Liver/virology , Phlebitis/pathology , RNA, Viral/analysis , Serum/enzymology , Serum/virology , Spleen/pathology , Spleen/virology
4.
J Clin Microbiol ; 42(6): 2658-62, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15184448

ABSTRACT

Avian hepatitis E virus (HEV), a novel virus identified from chickens with hepatitis-splenomegaly syndrome in the United States, is genetically and antigenically related to human HEV. In order to further characterize avian HEV, an infectious viral stock with a known infectious titer must be generated, as HEV cannot be propagated in vitro. Bile and feces collected from specific-pathogen-free (SPF) chickens experimentally infected with avian HEV were used to prepare an avian HEV infectious stock as a 10% suspension of positive fecal and bile samples in phosphate-buffered saline. The infectivity titer of this infectious stock was determined by inoculating 1-week-old SPF chickens intravenously with 200 microl of each of serial 10-fold dilutions (10(-2) to 10(-6)) of the avian HEV stock (two chickens were inoculated with each dilution). All chickens inoculated with the 10(-2) to 10(-4) dilutions of the infectious stock and one of the two chickens inoculated with the 10(-5) dilution, but neither of the chickens inoculated with the 10(-6) dilution, became seropositive for anti-avian HEV antibody at 4 weeks postinoculation (wpi). Two serologically negative contact control chickens housed together with chickens inoculated with the 10(-2) dilution also seroconverted at 8 wpi. Viremia and shedding of virus in feces were variable in chickens inoculated with the 10(-2) to 10(-5) dilutions but were not detectable in those inoculated with the 10(-6) dilution. The infectivity titer of the infectious avian HEV stock was determined to be 5 x 10(5) 50% chicken infectious doses (CID(50)) per ml. Eight 1-week-old turkeys were intravenously inoculated with 10(5) CID(50) of avian HEV, and another group of nine turkeys were not inoculated and were used as controls. The inoculated turkeys seroconverted at 4 to 8 wpi. In the inoculated turkeys, viremia was detected at 2 to 6 wpi and shedding of virus in feces was detected at 4 to 7 wpi. A serologically negative contact control turkey housed together with the inoculated ones also became infected through direct contact. This is the first demonstration of cross-species infection by avian HEV.


Subject(s)
Chickens/virology , Hepatitis E virus/pathogenicity , Hepatitis E/veterinary , Hepatitis, Viral, Animal/virology , Poultry Diseases/virology , Turkeys/virology , Animals , Disease Susceptibility , Hepatitis E/virology , Specific Pathogen-Free Organisms
5.
J Gen Virol ; 85(Pt 6): 1609-1618, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15166445

ABSTRACT

Avian hepatitis E virus (avian HEV), recently identified from a chicken with hepatitis-splenomegaly syndrome in the United States, is genetically and antigenically related to human and swine HEVs. In this study, sequencing of the genome was completed and an attempt was made to infect rhesus monkeys with avian HEV. The full-length genome of avian HEV, excluding the poly(A) tail, is 6654 bp in length, which is about 600 bp shorter than that of human and swine HEVs. Similar to human and swine HEV genomes, the avian HEV genome consists of a short 5' non-coding region (NCR) followed by three partially overlapping open reading frames (ORFs) and a 3'NCR. Avian HEV shares about 50 % nucleotide sequence identity over the complete genome, 48-51 % identity in ORF1, 46-48 % identity in ORF2 and only 29-34 % identity in ORF3 with human and swine HEV strains. Significant genetic variations such as deletions and insertions, particularly in ORF1 of avian HEV, were observed. However, motifs in the putative functional domains of ORF1, such as the helicase and methyltransferase, were relatively conserved between avian HEV and mammalian HEVs, supporting the conclusion that avian HEV is a member of the genus Hepevirus. Phylogenetic analysis revealed that avian HEV represents a branch distinct from human and swine HEVs. Swine HEV infects non-human primates and possibly humans and thus may be zoonotic. An attempt was made to determine whether avian HEV also infects across species by experimentally inoculating two rhesus monkeys with avian HEV. Evidence of virus infection was not observed in the inoculated monkeys as there was no seroconversion, viraemia, faecal virus shedding or serum liver enzyme elevation. The results from this study confirmed that avian HEV is related to, but distinct from, human and swine HEVs; however, unlike swine HEV, avian HEV is probably not transmissible to non-human primates.


Subject(s)
Chickens/virology , Genome, Viral , Hepatitis E virus/genetics , Hepatitis E/veterinary , Macaca mulatta/virology , Poultry Diseases/virology , Amino Acid Sequence , Animals , Base Sequence , Hepatitis E/transmission , Hepatitis E virus/classification , Molecular Sequence Data , Phylogeny , Swine
6.
J Gen Virol ; 85(Pt 3): 693-700, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14993655

ABSTRACT

Avian hepatitis E virus (HEV), a novel virus identified from chickens with hepatitis-splenomegaly (HS) syndrome, is genetically and antigenically related to human HEV. Recently, it was found that avian HEV antibody is also prevalent in healthy chickens. A prospective study was done on a known seropositive but healthy chicken farm to identify avian HEV isolates from healthy chickens. Fourteen chickens were randomly selected, tagged and monitored under natural conditions for 19 weeks. All 14 chickens were seronegative at the beginning of the study at 12 weeks of age. By 21 weeks of age, all 14 chickens had seroconverted to avian HEV antibody. None of the chickens had any sign of HS syndrome. Partial helicase gene and capsid gene sequences of avian HEV isolates recovered from a healthy chicken were determined and found to share 75-97 % nucleotide sequence identity with the corresponding regions of avian HEV isolates from chickens with HS syndrome. Thus far, only one strain of avian HEV from a chicken with HS syndrome has been genetically characterized for its capsid gene, therefore the capsid gene region of an additional 14 isolates from chickens with HS syndrome were also characterized. The capsid genes of avian HEV isolates from chickens with HS syndrome were found to be heterogeneic, sharing 76-100 % nucleotide sequence identity with each other. This study indicates that avian HEV is enzootic in chicken flocks and spreads subclinically among chickens in the United States and that the virus is heterogeneic.


Subject(s)
Chickens/virology , Hepatitis E virus/genetics , Hepatitis, Viral, Animal/virology , Splenomegaly/veterinary , Aging , Animals , Antibodies, Viral/blood , Base Sequence , Bile/virology , Capsid , DNA Primers , Hepatitis E virus/classification , Hepatitis E virus/immunology , Hepatitis E virus/isolation & purification , Hepatitis, Viral, Animal/immunology , Immunoglobulin G/blood , Open Reading Frames , Phylogeny , Poultry Diseases/virology , Splenomegaly/virology
7.
Vet Microbiol ; 96(2): 165-76, 2003 Oct 17.
Article in English | MEDLINE | ID: mdl-14519334

ABSTRACT

Hepatitis E virus (HEV), the causative agent of human hepatitis E, is an important public health problem in many developing countries and is also endemic in many industrialized countries including the US. The discoveries of avian and swine HEVs by our group from chickens and pigs, respectively, suggest that hepatitis E may be a zoonosis. Current methods for molecular epidemiological studies of HEV require PCR amplification of field strains of HEV followed by DNA sequencing and sequence analyses, which are laborious and expensive. As novel or variant strains of HEV continue to evolve rapidly both in humans and other animals, it is important to develop a rapid pre-sequencing screening method to select field isolates for further molecular characterization. In this study, we developed two heteroduplex mobility assays (HMA) (one for swine HEV based on the ORF2 region, and the other for avian HEV based on the ORF1 region) to genetically differentiate field strains of avian and swine HEVs from known reference strains. The ORF2 regions of 22 swine HEV isolates and the ORF1 regions of 13 avian HEV isolates were amplified by PCR, sequenced and analyzed by HMA against reference prototype swine HEV strain and reference prototype avian HEV strain, respectively. We showed that, in general, the HMA profiles correlate well with nucleotide sequence identities and with phylogenetic clustering between field strains and the reference swine HEV or avian HEV strains. Field isolates with similar HMA patterns generally showed similar sequence identities with the reference strains and clustered together in the phylogenetic trees. Therefore, by using different HEV isolates as references, the HMA developed in this study can be used as a pre-sequencing screening tool to identify variant HEV isolates for further molecular epidemiological studies.


Subject(s)
Chickens , Hepatitis E virus/isolation & purification , Hepatitis E/veterinary , Heteroduplex Analysis/veterinary , Poultry Diseases/virology , Swine Diseases/virology , Animals , Base Sequence , Gene Amplification , Hepatitis E/diagnosis , Hepatitis E/virology , Hepatitis E virus/classification , Hepatitis E virus/genetics , Heteroduplex Analysis/methods , Phylogeny , Poultry Diseases/diagnosis , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sensitivity and Specificity , Sequence Homology, Nucleic Acid , Swine , Swine Diseases/diagnosis
8.
J Clin Microbiol ; 41(6): 2433-9, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12791860

ABSTRACT

Porcine reproductive and respiratory syndrome has been devastating the swine industry since the late 1980s. The disease has been controlled, to some extent, through the use of modified live-attenuated (MLV) vaccines once available. However, such a practice periodically resulted in isolation or detection of vaccine-like viruses from pigs as determined by a partial genomic sequencing. In this study, we developed a heteroduplex mobility assay (HMA) for quickly identifying porcine reproductive and respiratory syndrome virus (PRRSV) isolates with significant nucleotide sequence identities (>/=98%) with the modified live-attenuated vaccines. The major envelope gene (ORF5) of 51 PRRSV field isolates recovered before and after the introduction of the vaccines was amplified, denatured, and reannealed with the HMA reference vaccine strains Ingelvac PRRS MLV and Ingelvac PRRS ATP, respectively. Nine of the 51 field isolates and the VR2332 parent virus of Ingelvac PRRS MLV, which were all highly related to Ingelvac PRRS MLV with

Subject(s)
Heteroduplex Analysis/methods , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/isolation & purification , Vaccines, Attenuated , Viral Vaccines , Animals , Molecular Sequence Data , Phylogeny , Porcine respiratory and reproductive syndrome virus/classification , Porcine respiratory and reproductive syndrome virus/genetics , Sequence Analysis, DNA , Swine , Swine Diseases/virology
9.
J Clin Microbiol ; 40(11): 4197-202, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12409397

ABSTRACT

We recently identified and characterized a novel virus, designated avian hepatitis E virus (avian HEV), from chickens with hepatitis-splenomegaly syndrome (HS syndrome) in the United States. Avian HEV is genetically related to but distinct from human and swine HEVs. To determine the extent of genetic variation and the seroprevalence of avian HEV infection in chicken flocks, we genetically identified and characterized 11 additional avian HEV isolates from chickens with HS syndrome and assessed the prevalence of avian HEV antibodies from a total of 1,276 chickens of different ages and breeds from 76 different flocks in five states (California, Colorado, Connecticut, Virginia, and Wisconsin). An enzyme-linked immunosorbent assay using a truncated recombinant avian HEV ORF2 antigen was developed and used to determine avian HEV seroprevalence. About 71% of chicken flocks and 30% of chickens tested in the study were positive for antibodies to avian HEV. About 17% of chickens younger than 18 weeks were seropositive, whereas about 36% of adult chickens were seropositive. By using a reverse transcription-PCR (RT-PCR) assay, we tested 21 bile samples from chickens with HS syndrome in California, Connecticut, New York, and Wisconsin for the presence of avian HEV RNA. Of the 21 bile samples, 12 were positive for 30- to 35-nm HEV-like virus particles by electron microscopy (EM). A total of 11 of the 12 EM-positive bile samples and 6 of the 9 EM-negative bile samples were positive for avian HEV RNA by RT-PCR. The sequences of a 372-bp region within the helicase gene of 11 avian HEV isolates were determined. Sequence analyses revealed that the 11 field isolates of avian HEV had 78 to 100% nucleotide sequence identities to each other, 79 to 88% identities to the prototype avian HEV, 76 to 80% identities to chicken big liver and spleen disease virus, and 56 to 61% identities to other known strains of human and swine HEV. The data from this study indicated that, like swine and human HEVs, avian HEV isolates are genetically heterogenic and that avian HEV infection is enzoonotic in chicken flocks in the United States.


Subject(s)
Chickens/virology , Genetic Variation , Hepatitis Antibodies/blood , Hepatitis E virus/immunology , Hepatitis E/veterinary , Poultry Diseases/epidemiology , Animals , Hepatitis E/epidemiology , Hepatitis E/virology , Hepatitis E virus/genetics , Molecular Sequence Data , Poultry Diseases/virology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Seroepidemiologic Studies , Splenomegaly/epidemiology , Splenomegaly/veterinary , Splenomegaly/virology , United States
10.
J Gen Virol ; 83(Pt 9): 2201-2209, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12185274

ABSTRACT

We recently identified a novel virus, designated avian hepatitis E virus (avian HEV), from chickens with hepatitis-splenomegaly (HS) syndrome in the USA. We showed that avian HEV is genetically related to swine and human HEVs. Here we report the antigenic cross-reactivity of the putative open reading frame 2 (ORF2) capsid protein of avian HEV with those of swine and human HEVs and the Australian chicken big liver and spleen disease virus (BLSV). The region encoding the C-terminal 268 amino acid residues of avian HEV ORF2 was cloned into expression vector pRSET-C. The truncated ORF2 protein was expressed in E. coli as a fusion protein and purified by affinity chromatography. Western blot analysis revealed that the avian HEV ORF2 protein reacted with antisera against the Sar-55 strain of human HEV and with convalescent antisera against swine HEV and the US2 strain of human HEV, as well as with antiserum against BLSV. Convalescent sera from specific-pathogen-free chickens experimentally infected with avian HEV also reacted with the recombinant capsid proteins of swine HEV and Sar-55 human HEV. Antisera against the US2 human HEV also reacted with recombinant ORF2 proteins of both swine HEV and Sar-55 human HEV. The antigenic cross-reactivity of the avian HEV putative capsid protein with those of swine and human HEVs was further confirmed, for the most part, by ELISA assays. The data indicate that avian HEV shares certain antigenic epitopes in its putative capsid protein with swine and human HEVs, as well as with BLSV. The results have implications for HEV diagnosis and taxonomy.


Subject(s)
Antigens, Viral/immunology , Capsid/immunology , Chickens/virology , Hepatitis E virus/immunology , Viral Fusion Proteins/immunology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , Bird Diseases/immunology , Capsid/genetics , Cross Reactions , Epitopes/immunology , Hepatitis E/immunology , Hepatitis E/veterinary , Hepatitis E virus/classification , Hepatitis E virus/genetics , Immune Sera , Molecular Sequence Data , Open Reading Frames , Sequence Alignment
11.
Vet Microbiol ; 88(2): 189-202, 2002 Aug 25.
Article in English | MEDLINE | ID: mdl-12135637

ABSTRACT

Brucella abortus resists the microbicidal mechanisms of macrophages, and the expression of its heat shock proteins (HSPs) such as GroEL, GroES and HtrA may play a role in this resistance. Bacterial HSPs can be very immunogenic, inducing protective immunity in various types of bacterial infections. However, the significance of immune responses directed against B. abortus HSPs in the protection against brucellosis is currently unresolved. To elucidate the role of these proteins in protection against Brucella challenge, individual, divalent or trivalent baculovirus (BV) recombinants of B. abortus GroEL, GroES and/or HtrA were injected into BALB/c mice either as protein-expressing whole cells or as purified proteins. The preparations were given to mice in combination with Freund's or Ribi adjuvant, respectively. In addition, some mice were primed with a vaccinia virus-GroEL recombinant, followed by inoculation with purified GroEL-Ribi adjuvant combination. Antibodies were observed against B. abortus GroEL and HtrA, but not against GroES. Cellular immune response was demonstrated by observing significant IFN-gamma release by lymphocytes of mice immunized with the purified HtrA-Ribi adjuvant combination. However, none of the mice inoculated with individual, divalent or trivalent HSP-expressing cells combined with complete Freund's adjuvant or inoculated with purified B. abortus HSPs combined with Ribi adjuvant, were protected against challenge with B. abortus virulent strain 2308. Priming with vaccinia virus-GroEL recombinant and boosting with GroEL-Ribi combination did not induce protective immunity. Based on the results obtained, we suggest that although humoral and cell-mediated immune responses are induced, but protective immune response is not induced by B. abortus HSPs.


Subject(s)
Antibodies, Bacterial/biosynthesis , Bacterial Vaccines/immunology , Brucella abortus/immunology , Heat-Shock Proteins/immunology , Animals , Baculoviridae , Brucellosis/prevention & control , Chaperonin 10/immunology , Chaperonin 60/immunology , Female , Immunity, Cellular , Mice , Mice, Inbred BALB C , Periplasmic Proteins/immunology , Serine Endopeptidases/immunology , Vaccination , Vaccines, Synthetic/immunology , Vaccinia virus
12.
J Clin Microbiol ; 40(4): 1326-32, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11923352

ABSTRACT

Hepatitis E virus (HEV) is an important public health concern in many developing countries. HEV is also endemic in some industrialized counties, including the United States. With our recent discovery of swine HEV in pigs that is genetically closely related to human HEV, hepatitis E is now considered a zoonotic disease. Human strains of HEV are genetically heterogenic. So far in the United States, only one strain of swine HEV has been identified and characterized from a pig. To determine the extent of genetic variations and the nature of swine HEV infections in U.S. pigs, we developed a universal reverse transcription-PCR (RT-PCR) assay that is capable of detecting genetically divergent strains of HEV. By using this universal RT-PCR assay, we tested fecal and serum samples of pigs of 2 to 4 months of age from 37 different U.S. swine farms for the presence of swine HEV RNA. Thirty-four of the 96 pigs (35%) and 20 of the 37 swine herds (54%) tested were positive for swine HEV RNA. The sequences of a 348-bp region within the ORF2 gene of 27 swine HEV isolates from different geographic regions were determined. Sequence analyses revealed that the 27 U.S. swine HEV isolates shared 88 to 100% nucleotide sequence identities with each other and 89 to 98% identities with the prototype U.S. strain of swine HEV. These U.S. swine HEV isolates are only distantly related to the Taiwanese strains of swine HEV, with about 74 to 78% nucleotide sequence identities; to most known human strains of HEV worldwide, with <79% sequence identities; and to avian HEV, with 54 to 56% sequence identities. Phylogenetic analysis showed that all the U.S. swine HEV isolates identified in this study clustered in the same genotype with the prototype U.S. swine HEV and the two U.S. strains of human HEV. The data from this study indicated that swine HEV is widespread and enzoonotic in U.S. swine herds and that, as is with human HEV, swine HEV isolates from different geographic regions of the world are also genetically heterogenic. These data further raise potential concerns for zoonosis, xenozoonosis, and food safety.


Subject(s)
Hepatitis E virus/classification , Hepatitis E virus/isolation & purification , Hepatitis E/veterinary , Reverse Transcriptase Polymerase Chain Reaction , Swine Diseases/virology , Animals , Genetic Variation , Hepatitis E/epidemiology , Hepatitis E/virology , Hepatitis E virus/genetics , Molecular Sequence Data , Phylogeny , Sensitivity and Specificity , Sequence Analysis, DNA , Swine , Swine Diseases/epidemiology , United States/epidemiology
13.
J Virol Methods ; 101(1-2): 71-8, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11849685

ABSTRACT

The objective of this study was to assess the risk of transmission of swine hepatitis E virus (swine HEV) to naïve pigs by inoculation with tissues or feces collected from pigs infected experimentally with swine HEV. Seventy-five, 3-week-old pigs were assigned randomly to 24 groups of 3-4 pigs and inoculated with homogenates of tissues (liver, heart, pancreas, or skeletal muscle) or a suspension of feces from swine HEV-infected pigs collected at 3, 7, 14, 20, 27, or 55 days post inoculation (DPI). Each inoculum was prepared as a 10% suspension (w/v) in PBS buffer and tested by a semi-quantitative RT-PCR for swine HEV RNA and by the swine bioassay. The inoculation route was intravenous for liver, heart and pancreas, and via stomach tube for skeletal muscle and fecal suspension. The liver homogenate inocula and feces collected at 3-7 and 14-20 DPI were positive for swine HEV RNA by RT-PCR. The pigs inoculated with liver homogenates collected at 3-7 and 14-20 DPI developed anti-HEV antibodies and swine HEV RNA was detected in their sera. Pigs inoculated with heart, pancreas, skeletal muscle homogenates or fecal suspensions failed to develop anti-HEV antibodies. These findings suggest that there is a potential risk of transmission of swine HEV via liver tissue from infected pigs in the early stages (3-20 DPI) of infection and the in vitro RT-PCR assay correlates well with the swine bioassay.


Subject(s)
Biological Assay/methods , Hepatitis E virus/isolation & purification , Hepatitis E/veterinary , Hepatitis, Viral, Animal/transmission , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Swine Diseases/virology , Animals , Disease Models, Animal , Feces/virology , Heart/virology , Hepatitis Antibodies/blood , Hepatitis E/immunology , Hepatitis E/transmission , Hepatitis E/virology , Hepatitis E virus/genetics , Hepatitis E virus/immunology , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/virology , Injections, Intravenous , Liver/virology , Muscle, Skeletal/virology , Pancreas/virology , RNA, Viral/blood , RNA, Viral/genetics , RNA, Viral/isolation & purification , Swine
14.
J Clin Microbiol ; 40(1): 117-22, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11773103

ABSTRACT

Hepatitis E virus (HEV) is endemic in many developing and some industrialized countries. It has been hypothesized that animals may be the source of infection. The recent identification of swine HEV in U.S. pigs and the demonstration of its ability to infect across species have lent credence to this hypothesis. To assess the potential risk of zoonotic HEV infection, we tested a total of 468 veterinarians working with swine (including 389 U.S. swine veterinarians) and 400 normal U.S. blood donors for immunoglobulin G anti-HEV. Recombinant capsid antigens from a U.S. strain of swine HEV and from a human HEV strain (Sar-55) were each used in an enzyme-linked immunosorbent assay. The anti-HEV prevalence assayed with the swine HEV antigen showed 97% concordance with that obtained with the human HEV antigen (kappa = 92%). Among the 295 swine veterinarians tested from the eight U.S. states (Minnesota, Indiana, Nebraska, Iowa, Illinois, Missouri, North Carolina, and Alabama) from which normal blood donor samples were available, 26% were positive with Sar-55 antigen and 23% were positive with swine HEV antigen. In contrast, 18% of the blood donors from the same eight U.S. states were positive with Sar-55 antigen and 17% were positive with swine HEV antigen. Swine veterinarians in the eight states were 1.51 times more likely when tested with swine HEV antigen (95% confidence interval, 1.03 to 2.20) and 1.46 times more likely when tested with Sar-55 antigen (95% confidence interval, 0.99 to 2.17) to be anti-HEV positive than normal blood donors. We did not find a difference in anti-HEV prevalence between veterinarians who reported having had a needle stick or cut and those who had not or between those who spent more time (> or = 80% of the time) and those who spent less time (< or = 20% of the time) working with pigs. Similarly, we did not find a difference in anti-HEV prevalence according to four job categories (academic, practicing, student, and industry veterinarians). There was a difference in anti-HEV prevalence in both swine veterinarians and blood donors among the eight selected states, with subjects from Minnesota six times more likely to be anti-HEV positive than those from Alabama. Age was not a factor in the observed differences from state to state. Anti-HEV prevalence in swine veterinarians and normal blood donors was age specific and paralleled increasing age. The results suggest that swine veterinarians may be at somewhat higher risk of HEV infection than are normal blood donors.


Subject(s)
Hepatitis Antibodies/blood , Hepatitis E virus/immunology , Hepatitis E/epidemiology , Swine Diseases/epidemiology , Veterinarians , Zoonoses/epidemiology , Adult , Aged , Animals , Blood Donors , Hepatitis E/immunology , Humans , Middle Aged , Prevalence , Risk Factors , Swine/virology , Swine Diseases/virology , United States/epidemiology
15.
J Virol ; 76(2): 541-51, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11752145

ABSTRACT

Infection of animals with a molecular viral clone is critical to study the genetic determinants of viral replication and virulence in the host. Type 2 porcine circovirus (PCV2) has been incriminated as the cause of postweaning multisystemic wasting syndrome (PMWS), an emerging disease in pigs. We report here for the first time the construction and use of an infectious molecular DNA clone of PCV2 to characterize the disease and pathologic lesions associated with PCV2 infection by direct in vivo transfection of pigs with the molecular clone. The PCV2 molecular clone was generated by ligating two copies of the complete PCV2 genome in tandem into the pBluescript SK (pSK) vector and was shown to be infectious in vitro when transfected into PK-15 cells. Forty specific-pathogen-free pigs at 4 weeks of age were randomly assigned to four groups of 10 each. Group 1 pigs served as uninoculated controls. Pigs in group 2 were each inoculated intranasally with about 1.9 x 10(5) 50% tissue culture infective doses of a homogeneous PCV2 live virus stock derived from the molecular clone. Pigs in group 3 were each injected intrahepatically with 200 microg of the cloned PCV2 plasmid DNA, and pigs in group 4 were each injected into the superficial iliac lymph nodes with 200 microg of the cloned PCV2 plasmid DNA. Animals injected with the cloned PCV2 plasmid DNA developed infection resembling that induced by intranasal inoculation with PCV2 live virus stock. Seroconversion to PCV2-specific antibody was detected in the majority of pigs from the three inoculated groups at 35 days postinoculation (DPI). Viremia, beginning at 14 DPI and lasting 2 to 4 weeks, was detected in the majority of the pigs from all three inoculated groups. There were no remarkable clinical signs of PMWS in control or any of the inoculated pigs. Gross lesions in pigs of the three inoculated groups were similar and were characterized by systemically enlarged, tan lymph nodes and lungs that failed to collapse. Histopathological lesions and PCV2-specific antigen were detected in numerous tissues and organs, including brain, lung, heart, kidney, tonsil, lymph nodes, spleen, ileum, and liver of infected pigs. This study more definitively characterizes the clinical course and pathologic lesions exclusively attributable to PCV2 infection. The data from this study indicate that the cloned PCV2 genomic DNA may replace infectious virus for future PCV2 pathogenesis and immunization studies. The data also suggest that PCV2, although essential for development of PMWS, may require other factors or agents to induce the full spectrum of clinical signs and lesions associated with advanced cases of PMWS.


Subject(s)
Circoviridae Infections/pathology , Circoviridae Infections/virology , Circovirus/physiology , Genome, Viral , Liver/virology , Lymph Nodes/virology , Swine/virology , Administration, Intranasal , Animals , Antigens, Viral/analysis , Cell Line , Circoviridae Infections/veterinary , Circovirus/genetics , Circovirus/immunology , Circovirus/pathogenicity , Cloning, Molecular , DNA, Recombinant/genetics , DNA, Viral/genetics , DNA, Viral/physiology , Immunohistochemistry , Lung/pathology , Lung/virology , Lymph Nodes/pathology , Necrosis , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Swine Diseases/pathology , Swine Diseases/virology , Transfection , Viremia/pathology , Viremia/virology
16.
Vet Microbiol ; 83(3): 249-63, 2001 Nov 26.
Article in English | MEDLINE | ID: mdl-11574173

ABSTRACT

Swine herds in the US have experienced recent outbreaks of a severe form of porcine reproductive and respiratory syndrome (designated acute or atypical PRRS) characterized by abortion and high mortality in pregnant sows. Most of the affected herds had been vaccinated with modified live-vaccines (MLVs) against PRRS. To explore the possible mechanism of the emergence of acute PRRS, the open reading frame 5 (ORF5) gene encoding the major envelope protein (GP5) of acute PRRSV isolates was characterized. The complete ORF5 gene of eight acute PRRSV isolates from herds experiencing acute PRRS outbreaks in Iowa and North Carolina was amplified and sequenced. Sequence analyses revealed that these acute PRRSV isolates shared 88-95% nucleotide and 88-96% amino acid sequence identities to each other, 87-97% nucleotide and 84-96% amino acid sequence identities with other North American PRRSV isolates and the MLVs. Most of the amino acid substitutions locate in the putative signal sequence and two short hypervariable regions at the amino terminus. The ORF5 gene sequence of the acute PRRSV isolate 98-37120-2 from a non-vaccinated swine herd in Iowa is very closely related to that of the RespPRRS MLV, with 97% nucleotide and 96% amino acid sequence identities. Phylogenetic analysis revealed that all eight acute PRRSV isolates are clustered within the North American genotype. Several minor branches that are not associated with geographic origins were also identified within the North American genotype. One acute PRRSV isolate (98-37120-2) is clustered with the RespPRRS MLV and several Danish isolates that were confirmed to be derived from the RespPRRS MLV. The ORF5 gene sequences of other seven acute isolates are more related to those of several earlier PRRSV isolates and the PrimePac MLV than to that of the RespPRRS MLV. Our results showed that the acute PRRSV isolates analyzed in this study differed from each other in ORF5 genes, although they all clustered within the North American genotype. The data from this study do not fully support the hypothesis that the emergence of acute PRRS is due to reversion of MLVs to a pathogenic phenotype, as only one of the eight acute isolates was shown to be very closely related to the RespPRRS MLV.


Subject(s)
Genetic Variation , Porcine Reproductive and Respiratory Syndrome/epidemiology , Porcine respiratory and reproductive syndrome virus/classification , Porcine respiratory and reproductive syndrome virus/genetics , Viral Proteins/genetics , Acute Disease , Amino Acid Sequence , Animals , Base Sequence , Gene Amplification , Genotype , Iowa/epidemiology , Molecular Sequence Data , Multigene Family , North Carolina/epidemiology , Phylogeny , Porcine Reproductive and Respiratory Syndrome/virology , Swine , Viral Envelope Proteins , Viral Proteins/chemistry
17.
J Clin Microbiol ; 39(9): 3040-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11526125

ABSTRACT

Hepatitis E virus (HEV) is the major cause of enterically transmitted non-A, non-B hepatitis in many developing countries and is also endemic in many industrialized countries. Due to the lack of an effective cell culture system and a practical animal model, the mechanisms of HEV pathogenesis and replication are poorly understood. Our recent identification of swine HEV from pigs affords us an opportunity to systematically study HEV replication and pathogenesis in a swine model. In an early study, we experimentally infected specific-pathogen-free pigs with two strains of HEV: swine HEV and the US-2 strain of human HEV. Eighteen pigs (group 1) were inoculated intravenously with swine HEV, 19 pigs (group 2) were inoculated with the US-2 strain of human HEV, and 17 pigs (group 3) were used as uninoculated controls. The clinical and pathological findings have been previously reported. In this expanded study, we aim to identify the potential extrahepatic sites of HEV replication using the swine model. Two pigs from each group were necropsied at 3, 7, 14, 20, 27, and 55 days postinoculation (DPI). Thirteen different types of tissues and organs were collected from each necropsied animal. Reverse transcriptase PCR (RT-PCR) was used to detect the presence of positive-strand HEV RNA in each tissue collected during necropsy at different DPI. A negative-strand-specific RT-PCR was standardized and used to detect the replicative, negative strand of HEV RNA from tissues that tested positive for the positive-strand RNA. As expected, positive-strand HEV RNA was detected in almost every type of tissue at some time point during the viremic period between 3 and 27 DPI. Positive-strand HEV RNA was still detectable in some tissues in the absence of serum HEV RNA from both swine HEV- and human HEV-inoculated pigs. However, replicative, negative-strand HEV RNA was detected primarily in the small intestines, lymph nodes, colons, and livers. Our results indicate that HEV replicates in tissues other than the liver. The data from this study may have important implications for HEV pathogenesis, xenotransplantation, and the development of an in vitro cell culture system for HEV.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/virology , Liver/virology , Virus Replication , Animals , Disease Models, Animal , Hepatitis E/physiopathology , Hepatitis E virus/pathogenicity , Humans , RNA, Viral/analysis , Swine
18.
Vet Ophthalmol ; 4(2): 119-26, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11422993

ABSTRACT

OBJECTIVES: To determine the presence of feline leukemia virus (FeLV) in the corneas of FeLV-infected cats. ANIMALS STUDIED: Thirty-four random source cats. PROCEDURES: Seventeen cats were found positive for FeLV serum p27 antigen by enzyme-linked immunosorbent assay (ELISA). Twelve ELISA positive cats were also positive on peripheral blood samples by immunofluorescent antibody (IFA) testing. Seventeen ELISA negative cats served as negative controls. Full thickness corneal specimens were collected from the left cornea of all cats and analyzed for FeLV proviral DNA and gp70 antigen by polymerase chain reaction (PCR) and immunohistochemical (IHC) testing, respectively. RESULTS: Eleven (64.7%) positive corneal PCR results were obtained from 17 ELISA positive cats. Of 12 cats that were both ELISA and IFA positive on peripheral blood, 10 (83.3%) had positive corneal PCR results. All corneal tissues from ELISA negative cats were PCR negative. IHC staining of corneal sections revealed the presence of FeLV gp70 in corneal tissues of nine (52.9%) ELISA positive cats. Of the 12 cats that were both ELISA and IFA positive on peripheral blood, eight (66.7%) had positive corneal IHC results. Positive IHC staining was localized to the corneal epithelium. Corneal tissues of all ELISA negative cats and all IFA negative cats were negative on IHC testing. CONCLUSIONS: FeLV antigens and proviral DNA are present in corneal tissues of some FeLV-infected cats. Screening corneal donors for FeLV infection is warranted.


Subject(s)
Cat Diseases/virology , Cornea/virology , Leukemia Virus, Feline/isolation & purification , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Antigens, Viral/isolation & purification , Case-Control Studies , Cats , DNA Primers , DNA, Viral/isolation & purification , Electrophoresis, Agar Gel/veterinary , Female , Immunohistochemistry/veterinary , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/immunology , Male , Polymerase Chain Reaction/veterinary , Retroviridae Infections/virology , Tumor Virus Infections/virology
19.
J Clin Microbiol ; 39(3): 918-23, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11230404

ABSTRACT

Specific-pathogen-free pigs were inoculated with one of two hepatitis E viruses (HEV) (one recovered from a pig and the other from a human) to study the relative pathogenesis of the two viruses in swine. Fifty-four pigs were randomly assigned to three groups. Seventeen pigs in group 1 served as uninoculated controls, 18 pigs in group 2 were intravenously inoculated with the swine HEV recovered from a pig in the United States, and 19 pigs in group 3 were intravenously inoculated with the US-2 strain of human HEV recovered from a hepatitis patient in the United States. Two to four pigs from each group were necropsied at 3, 7, 14, 20, 27, or 55 days postinoculation (DPI). Evidence of clinical disease or elevation of liver enzymes or bilirubin was not found in pigs from any of the three groups. Enlarged hepatic and mesenteric lymph nodes were observed in both HEV-inoculated groups. Multifocal lymphoplasmacytic hepatitis was observed in 9 of 17, 15 of 18, and 16 of 19 pigs in groups 1 to 3, respectively. Focal hepatocellular necrosis was observed in 5 of 17, 10 of 18, and 13 of 19 pigs in groups 1 to 3, respectively. Hepatitis lesions were very mild in group 1 pigs, mild to moderate in group 2 pigs, and moderate to severe in group 3 pigs. Hepatic inflammation and hepatocellular necrosis peaked in severity at 20 DPI and were still moderately severe at 55 DPI in the group inoculated with human HEV. Hepatitis lesions were absent or nearly resolved by 55 DPI in the swine-HEV-inoculated pigs. All HEV-inoculated pigs seroconverted to anti-HEV immunoglobulin G. HEV RNA was detected by reverse transcriptase PCR in feces, liver tissue, and bile of pigs in both HEV-inoculated groups from 3 to 27 DPI. Based on evaluation of microscopic lesions, the US-2 strain of human HEV induced more severe and persistent hepatic lesions in pigs than did swine HEV. Pig livers or cells from the livers of HEV-infected pigs may represent a risk for transmission of HEV from pigs to human xenograft recipients. Since HEV was shed in the feces of infected pigs, exposure to feces from infected pigs represents a risk for transmission of HEV, and pigs should be considered a reservoir for HEV.


Subject(s)
Disease Models, Animal , Hepatitis E virus/pathogenicity , Swine Diseases/virology , Swine , Animals , Hepatitis Antibodies/blood , Hepatitis E/physiopathology , Hepatitis E/virology , Hepatitis E virus/isolation & purification , Humans , Liver/pathology , RNA, Viral/analysis , RNA, Viral/blood , Swine Diseases/physiopathology
20.
J Feline Med Surg ; 3(3): 133-41, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11876630

ABSTRACT

The purpose of this study was to determine if polymerase chain reaction (PCR) could be used to detect FeLV proviral DNA in bone marrow samples of cats with varying suspicion of latent infection. Blood and bone marrow samples from 50 cats and bone marrow from one fetus were collected, including 16 cats with diseases suspected to be FeLV-associated. Serum enzyme-linked immunosorbent assay (ELISA), blood and bone marrow immunofluorescent antibody test (IFA), and blood and bone marrow PCR were performed on each cat, and IFA and PCR on bone marrow of the fetus. Forty-one cats were FeLV negative. Five cats and one fetus were persistently infected with FeLV. Four cats had discordant test results. No cats were positive on bone marrow PCR only. It appears persistent or latent FeLV infection is not always present in conditions classically associated with FeLV.


Subject(s)
DNA, Viral/isolation & purification , Leukemia Virus, Feline/genetics , Leukemia, Feline/diagnosis , Animals , Bone Marrow/virology , Cats , DNA Primers , DNA, Viral/blood , Enzyme-Linked Immunosorbent Assay/veterinary , Fluorescent Antibody Technique/veterinary , Leukemia Virus, Feline/isolation & purification , Leukemia, Feline/pathology , Polymerase Chain Reaction/veterinary , Predictive Value of Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...