Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 10(1): e0226521, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35138149

ABSTRACT

Avian hepatitis E virus (HEV) causes liver diseases and multiple extrahepatic disorders in chickens. However, the mechanisms involved in avian HEV entry remain elusive. Herein, we identified the RAS-related protein 1b (Rap1b) as a potential HEV-ORF2 protein interacting candidate. Experimental infection of chickens and cells with an avian HEV isolate from China (CaHEV) led to upregulated expression and activation of Rap1b both in vivo and in vitro. By using CaHEV capsid as mimic of virion to treat cell in vitro, it appears that the interaction between the viral capsid and Rap1b promoted cell membrane recruitment of the downstream effector Rap1-interacting molecule (RIAM). In turn, RIAM further enhanced Talin-1 membrane recruitment and retention, which led to the activation of integrin α5/ß1, as well as integrin-associated membrane protein kinases, including focal adhesion kinase (FAK). Meanwhile, FAK activation triggered activation of downstream signaling molecules, such as Ras-related C3 botulinum toxin substrate 1 RAC1 cell division cycle 42 (CDC42), p21-activated kinase 1 (PAK1), and LIM domain kinase 1 (LIMK1). Finally, F-actin rearrangement induced by Cofilin led to the formation of lamellipodia, filopodia, and stress fibers, contributes to plasma membrane remodeling, and might enhance CaHEV virion internalization. In conclusion, our data suggested that Rap1b activation was triggered during CaHEV infection and appeared to require interaction between CaHEV-ORF2 and Rap1b, thereby further inducing membrane recruitment of Talin-1. Membrane-bound Talin-1 then activates key Integrin-FAK-Cofilin cascades involved in modulation of actin kinetics, and finally leads to F-actin rearrangement and membrane remodeling to potentially facilitate internalization of CaHEV virions into permissive cells. IMPORTANCE Rap1b is a multifunctional protein that is responsible for cell adhesion, growth, and differentiation. The inactive form of Rap1b is phosphorylated and distributed in the cytoplasm, while active Rap1b is prenylated and loaded with GTP to the cell membrane. In this study, the activation of Rap1b was induced during the early stage of avian HEV infection under the regulation of PKA and SmgGDS. Continuously activated Rap1b recruited its effector RIAM to the membrane, thereby inducing the membrane recruitment of Talin-1 that led to the activation of membrane α5/ß1 integrins. The triggering of the signaling pathway-associated Integrin α5/ß1-FAK-CDC42&RAC1-PAK1-LIMK1-Cofilin culminated in F-actin polymerization and membrane remodeling that might promote avian HEV virion internalization. These findings suggested a novel mechanism that is potentially utilized by avian HEV to invade susceptible cells.


Subject(s)
Cytoskeleton/metabolism , Hepatitis, Viral, Animal/metabolism , Hepevirus/pathogenicity , Poultry Diseases/metabolism , Viral Proteins/metabolism , Virus Internalization , rap GTP-Binding Proteins/metabolism , Actins/genetics , Actins/metabolism , Animals , Chickens , Cytoskeleton/genetics , Cytoskeleton/virology , Hepatitis, Viral, Animal/genetics , Hepatitis, Viral, Animal/virology , Hepevirus/genetics , Host-Pathogen Interactions , Poultry Diseases/genetics , Poultry Diseases/virology , Protein Binding , Viral Proteins/genetics , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , rap GTP-Binding Proteins/genetics
2.
Viruses ; 13(9)2021 08 31.
Article in English | MEDLINE | ID: mdl-34578311

ABSTRACT

The sole member of the Piscihepevirus genus (family Hepeviridae) is cutthroat trout virus (CTV) but recent metatranscriptomic studies have identified numerous fish hepevirus sequences including CTV-2. In the current study, viruses with sequences resembling both CTV and CTV-2 were isolated from salmonids in eastern and western Canada. Phylogenetic analysis of eight full genomes delineated the Canadian CTV isolates into two genotypes (CTV-1 and CTV-2) within the Piscihepevirus genus. Hepevirus genomes typically have three open reading frames but an ORF3 counterpart was not predicted in the Canadian CTV isolates. In vitro replication of a CTV-2 isolate produced cytopathic effects in the CHSE-214 cell line with similar amplification efficiency as CTV. Likewise, the morphology of the CTV-2 isolate resembled CTV, yet viral replication caused dilation of the endoplasmic reticulum lumen which was not previously observed. Controlled laboratory studies exposing sockeye (Oncorhynchus nerka), pink (O. gorbuscha), and chinook salmon (O. tshawytscha) to CTV-2 resulted in persistent infections without disease and mortality. Infected Atlantic salmon (Salmo salar) and chinook salmon served as hosts and potential reservoirs of CTV-2. The data presented herein provides the first in vitro and in vivo characterization of CTV-2 and reveals greater diversity of piscihepeviruses extending the known host range and geographic distribution of CTV viruses.


Subject(s)
Fish Diseases/virology , Hepevirus/classification , Hepevirus/genetics , Hepevirus/isolation & purification , Animals , Canada , Genotype , Hepevirus/pathogenicity , Persistent Infection/virology , Phylogeny , Salmo salar/virology , Salmon/virology , Trout , Virulence , Viruses, Unclassified/classification , Viruses, Unclassified/genetics , Viruses, Unclassified/isolation & purification , Viruses, Unclassified/pathogenicity
3.
Vet Microbiol ; 242: 108577, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32122587

ABSTRACT

Since 2016, severe outbreaks of hepatic rupture hemorrhage syndrome (HRHS) associated with infections of tentative novel avian hepatitis E virus (HEV) have emerged in chickens in China, causing increased mortality and decreased laying rate in adult hens and disturbing the hatching and breeding of chicks. To further identify the genotype and gain a better understanding of the genetic properties of the avian HEV responsible for that, a strain from Hebei province was isolated, purified and sequenced in this study. Results identified a novel avian HEV genotype, sharing 79.5-86.9% identities with other published avian HEV strains, and having higher identities with Orthohepevirus A HEV strains. More importantly, the new isolate contains various amino-acid substitutions in its functional proteins, including methyltransferase, helicase, RNA-dependent RNA polymerase. The data presented in this report will enhance the current understanding of the genetic diversity of the avian HEV and provide additional insight into the critical factors that determine the pathogenicity.


Subject(s)
Genome, Viral , Hemorrhage/veterinary , Hepatitis, Viral, Animal/virology , Hepevirus/genetics , Animals , Chickens/virology , China , Farms , Genetic Variation , Genotype , Hemorrhage/virology , Hepatitis, Viral, Animal/complications , Hepevirus/pathogenicity , Liver/pathology , Liver/virology , Mutation , Phylogeny , Poultry Diseases/virology , Sequence Analysis, DNA , Whole Genome Sequencing
4.
Vet Microbiol ; 222: 91-97, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30080679

ABSTRACT

Since 2016, severe outbreaks of hepatic rupture hemorrhage syndrome (HRHS) have emerged in chickens in several Chinese provinces and caused huge economic losses to the poultry industry, but the etiological characteristics and pathogenic potential of it has remained unclear. This study sequenced the partial helicase and capsid gene of the potentially novel avian hepatitis E virus (HEV) isolated from chickens with HRHS and tested the pathogenicity of it on SPF chicks, while the appearance of clinical signs, histopathological changes, viral distribution, viremia and viral shedding were monitored for 14 days post-infection (dpi). Analysis revealed that the HRHS related avian HEV belongs to a novel genotype, and infected chicks developed the typical symptoms of HRHS. Thus, this study successfully developed an experimental infection model for studying the pathogenicity and role of the novel avian HEV in HRHS. Meanwhile, the novel avian HEV mainly existed in the liver and spleen, inducing a rapid viremia and chronic viral shedding in infected chicks, and could cause 40% mortality before 14 dpi. In conclusion, this study found the novel genotype avian HEV and confirmed its role in HRHS.


Subject(s)
Genotype , Hepatitis E/veterinary , Hepatitis, Viral, Animal/virology , Hepevirus/genetics , Liver Diseases/veterinary , Liver/pathology , Animals , Antibodies, Viral/blood , Chickens/virology , China/epidemiology , Disease Models, Animal , Genes, Viral/genetics , Hemorrhage , Hepatitis E/blood , Hepatitis E/virology , Hepatitis, Viral, Animal/blood , Hepatitis, Viral, Animal/epidemiology , Hepevirus/pathogenicity , High-Throughput Nucleotide Sequencing , Liver/virology , Liver Diseases/virology , Poultry Diseases/virology , Rupture, Spontaneous/veterinary , Rupture, Spontaneous/virology , Viremia/pathology , Virus Shedding
5.
Trends Microbiol ; 26(7): 598-610, 2018 07.
Article in English | MEDLINE | ID: mdl-29268982

ABSTRACT

Viruses from the Coronaviridae, Togaviridae, and Hepeviridae families ​all contain genes that encode a conserved protein domain, called a macrodomain; however, the role of this domain during infection has remained enigmatic. The recent discovery that mammalian macrodomain proteins enzymatically remove ADP-ribose, a common post-translation modification, from proteins has led to an outburst of studies describing both the enzymatic activity and function of viral macrodomains. These new studies have defined these domains as de-ADP-ribosylating enzymes, which indicates that these viruses have evolved to counteract antiviral ADP-ribosylation, likely mediated by poly-ADP-ribose polymerases (PARPs). Here, we comprehensively review this rapidly expanding field, describing the structures and enzymatic activities of viral macrodomains, and discussing their roles in viral replication and pathogenesis.


Subject(s)
Protein Domains , Viral Nonstructural Proteins/chemistry , Virus Replication , Viruses/genetics , Viruses/pathogenicity , Adenosine Diphosphate Ribose/metabolism , Coronaviridae/genetics , Coronaviridae/pathogenicity , Hepevirus/genetics , Hepevirus/pathogenicity , Histones , Poly(ADP-ribose) Polymerases , Protein Processing, Post-Translational , Togaviridae/genetics , Togaviridae/pathogenicity , Viral Nonstructural Proteins/metabolism , Viruses/enzymology
6.
Vet Microbiol ; 203: 174-180, 2017 May.
Article in English | MEDLINE | ID: mdl-28619140

ABSTRACT

To determine the relationship between decreased egg production and avian HEV infection, thirty healthy 23-week-old Hy-Line Variety Brown layer hens were randomly divided into 3 groups with 10 hens per group. Next, a genotype 3 avian HEV strain from China was used to inoculate laying hens via oronasal or intravenous routes using a 50% chicken infectious dose of 500. All hens were necropsied at 14 weeks postinoculation (wpi). Fecal virus shedding, viremia, seroconversion, serum alanine aminotransferase (ALT) increases and liver lesions showed that after intravenous (i.v.) and oronasal inoculation, the laying hens were successfully infected. Compared with the uninoculated group, the i.v. and oronasally inoculated groups exhibited egg production decreases at 1wpi and 2wpi, reaching peak production at 3wpi and 8wpi, respectively. In both groups, decreased production was evident for 12 weeks and overall decreases ranged from 10% to 30%. In addition, in the 7 field layer farms exhibiting decreased egg production, vaccination regimens had been completed against Newcastle disease, infectious bronchitis, avian influenza H9N2 and H5N1 and egg drop syndrome virus. However, circulating avian HEV was confirmed on these farms using tests to detect avian HEV IgG antibodies and RNA. Therefore, the experimental and field data indicate that avian HEV infection acting alone could account for observed decreases in egg production in laying hens.


Subject(s)
Chickens/virology , Hepatitis, Viral, Animal/virology , Hepevirus/pathogenicity , RNA Virus Infections/veterinary , Animals , China , Eggs , Female , Genotype , Hepevirus/genetics , Hepevirus/isolation & purification , RNA Virus Infections/virology , Virus Shedding
8.
Wei Sheng Wu Xue Bao ; 52(3): 279-85, 2012 Mar 04.
Article in Chinese | MEDLINE | ID: mdl-22712397

ABSTRACT

Avian hepatitis E virus (HEV), a member of Hepeviridae family, is genetically and antigenically related with human and swine HEV in the family. Since its discovery, avian HEV infection has been investigated in many countries from serology and molecular epidemiology studies. At present, five complete or near complete genomes of avian HEV isolates were reported in GenBank and were divided into three genotypes. The complete genome of avian HEV contains 3 ORFs of which ORF2 gene encodes capsid protein containing the primary epitopes of viral particles and is target gene for serodiagnostic antigen and vaccine candidate. Because avian HEV infection has significant impact on the poultry industry and potential zoonotic transmission, the researches on avian HEV have been given much attention. We here give a broad review of the research update on the aetiology, pathogenesis and the antigenicity of capsid protein of avian HEV based on identification of Chinese avian HEV isolate.


Subject(s)
Hepevirus/isolation & purification , Poultry Diseases/virology , RNA Virus Infections/veterinary , RNA Virus Infections/virology , Amino Acid Sequence , Animals , China , Hepevirus/genetics , Hepevirus/immunology , Hepevirus/pathogenicity , Humans , Molecular Sequence Data , Open Reading Frames , Poultry , Viral Proteins/genetics , Viral Proteins/immunology
9.
Vet Microbiol ; 147(3-4): 310-9, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-20708350

ABSTRACT

A genetically distinct strain of avian hepatitis E virus (avian HEV-VA strain) was isolated from a healthy chicken in Virginia, and thus it is important to characterize and compare its pathogenicity with the prototype strain (avian HEV-prototype) isolated from a diseased chicken. Here we first constructed an infectious clone of the avian HEV-VA strain. Capped RNA transcripts from the avian HEV-VA clone were replication-competent after transfection of LMH chicken liver cells. Chickens inoculated intrahepatically with RNA transcripts of avian HEV-VA clone developed active infection as evidenced by fecal virus shedding, viremia, and seroconversion. To characterize the pathogenicity, RNA transcripts of both avian HEV-VA and avian HEV-prototype clones were intrahepatically inoculated into the livers of chickens. Avian HEV RNA was detected in feces, serum and bile samples from 10/10 avian HEV-VA-inoculated and 9/9 avian HEV-prototype-inoculated chickens although seroconversion occurred only in some chickens during the experimental period. The histopathological lesion scores were lower for avian HEV-VA group than avian HEV-prototype group in the liver at 3 and 5 weeks post-inoculation (wpi) and in the spleen at 3 wpi, although the differences were not statistically significant. The liver/body weight ratio, indicative of liver enlargement, of both avian HEV-VA and avian HEV-prototype groups were significantly higher than that of the control group at 5 wpi. Overall, the avian HEV-VA strain still induces histological liver lesions even though it was isolated from a healthy chicken. The results also showed that intrahepatic inoculation of chickens with RNA transcripts of avian HEV infectious clone may serve as an alternative for live virus in animal pathogenicity studies.


Subject(s)
DNA, Complementary/metabolism , Hepatitis E/veterinary , Hepatitis, Viral, Animal/virology , Hepevirus/pathogenicity , Poultry Diseases/virology , Animals , Cells, Cultured , Chickens , Cloning, Molecular , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Hepatitis E/pathology , Hepatitis E/virology , Hepatitis, Viral, Animal/physiopathology , Hepevirus/genetics , Liver/pathology , Liver/virology , Poultry Diseases/pathology , RNA Caps/genetics , Specific Pathogen-Free Organisms , Virginia , Virus Shedding
10.
Vet Microbiol ; 139(3-4): 253-61, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19570623

ABSTRACT

Avian hepatitis E virus (avian HEV) is the primary causative agent of Hepatitis-Splenomegaly (HS) syndrome in chickens. Recently, a genetically unique strain of avian HEV, designated avian HEV-VA, was recovered from healthy chickens in Virginia. The objective of this study was to experimentally compare the pathogenicity of the prototype strain recovered from a chicken with HS syndrome and the avian HEV-VA strain in specific-pathogen-free chickens. An infectious stock of the avian HEV-VA strain was first generated and its infectivity titer determined in chickens. For the comparative pathogenesis study, 54 chickens of 6-week-old were assigned to 3 groups of 18 chickens each. The group 1 chickens were each intravenously inoculated with 5x10(2.5) 50% chicken infectious dose of the prototype strain. The group 2 received the same dose of the avian HEV-VA strain, and the group 3 served as negative controls. Six chickens from each group were necropsied at 2, 3 and 4 weeks post-inoculation (wpi). Most chickens in both inoculated groups seroconverted by 3wpi, and the mean anti-avian HEV antibody titers were higher for the prototype strain group than the avian HEV-VA strain group. There was no significant difference in the patterns of viremia and fecal virus shedding. Blood analyte profiles did not differ between treatment groups except for serum creatine phosphokinase levels which were higher for prototype avian HEV group than avian HEV-VA group. The hepatic lesion score was higher for the prototype strain group than the other two groups. The results indicated that the avian HEV-VA strain is only slightly attenuated compared to the prototype strain, suggesting that the full spectrum of HS syndrome is likely associated with other co-factors.


Subject(s)
Chickens , Hepatitis, Viral, Animal/virology , Hepevirus/pathogenicity , Poultry Diseases/virology , Animals , Antibodies, Viral/blood , Feces/virology , Hepatitis, Viral, Animal/pathology , Hepevirus/genetics , Hepevirus/immunology , Immunoglobulin G/blood , Molecular Sequence Data , Poultry Diseases/pathology , Specific Pathogen-Free Organisms , Swine , Viremia/genetics , Viremia/immunology , Viremia/virology , Virginia
11.
Vaccine ; 25(15): 2892-9, 2007 Apr 12.
Article in English | MEDLINE | ID: mdl-17074425

ABSTRACT

Avian hepatitis E virus (avian HEV) is an emerging virus associated with hepatitis-splenomegaly syndrome in chickens in North America. Avian HEV is genetically and antigenically related to human HEV, the causative agent of hepatitis E in humans. In the lack of a practical animal model, avian HEV infection in chickens has been used as a model to study human HEV replication and pathogenesis. A 32 kDa recombinant ORF2 capsid protein of avian HEV expressed in Escherichia coli was found having similar antigenic structure as that of human HEV containing major neutralizing epitopes. To determine if the capsid protein of avian HEV can be used as a vaccine, 20 chickens were immunized with purified avian HEV recombinant protein with aluminum as adjuvant and another 20 chickens were mock immunized with KLH precipitated in aluminum as controls. Both groups of chickens were subsequently challenged with avian HEV. All the tested mock-immunized control chickens developed typical avian HEV infection characterized by viremia, fecal virus shedding and seroconversion to avian HEV antibodies. Gross hepatic lesions were also found in portion of these chickens. In contrast, none of the tested chickens immunized with avian HEV capsid protein had detectable viremia, fecal virus shedding or observable gross hepatitis lesions. The results from this study suggested that immunization of chickens with avian HEV recombinant ORF2 capsid protein with aluminum as adjuvant can induce protective immunity against avian HEV infection. Chickens are a useful small animal model to study anti-HEV immunity and pathogenesis.


Subject(s)
Hepevirus/immunology , Poultry Diseases/prevention & control , RNA Virus Infections/prevention & control , RNA Virus Infections/veterinary , Viral Hepatitis Vaccines/immunology , Viral Proteins/immunology , Animals , Antibodies, Viral/biosynthesis , Chickens , Hepevirus/pathogenicity , Humans , L-Lactate Dehydrogenase/blood , Poultry Diseases/immunology , Poultry Diseases/virology , RNA Virus Infections/immunology , RNA Virus Infections/virology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Hepatitis Vaccines/pharmacology , Viral Proteins/genetics , Viremia/immunology , Viremia/prevention & control , Viremia/veterinary , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL
...