Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Arch Virol ; 164(10): 2451-2458, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31273469

ABSTRACT

Avian reovirus (ARV) is a member of the genus Orthoreovirus in the family Reoviridae and causes a severe syndrome including viral arthritis that leads to considerable losses in the poultry industry. Innate immunity plays a significant role in host defense against ARV. Here, we explored the interaction between ARV and the host innate immune system by measuring mRNA expression levels of several genes associated with the MDA5 signaling pathway. The results showed that expression peaks for MDA5, MAVS, TRAF3, TRAF6, IRF7, IKKɛ, TBK1 and NF-κB occurred at 3 days postinfection (dpi). Moreover, type I IFN (IFN-α, IFN-ß) and IL-12 expression levels peaked at 3 dpi, while type II IFN (IFN-γ), IL-6, IL-17 and IL-18 expression reached a maximum level at 1 dpi. IL-8 changed at 5 dpi, and IL-1ß and TNF-α changed at 7 dpi. Interestingly, several key IFN-stimulated genes (ISGs), including IFITM1, IFITM2, IFITM5, Mx1 and OASL, were simultaneously upregulated and reached maximum values at 3 dpi. These data indicate that the MDA5 signaling pathway and innate immune cytokines were induced after ARV infection, which would contribute to the ARV-host interaction, especially at the early infection stage.


Subject(s)
Interferon-Induced Helicase, IFIH1/biosynthesis , Lymphocytes/pathology , Orthoreovirus, Avian/growth & development , Poultry Diseases/pathology , Reoviridae Infections/veterinary , Signal Transduction , Transcriptome , Animals , Chickens , Cytokines/biosynthesis , Host-Pathogen Interactions , Immunity, Innate
2.
Virology ; 487: 104-11, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26517397

ABSTRACT

We have previously shown that the replication of avian reovirus (ARV) in chicken embryo fibroblasts (CEF) is more resistant to the antiviral action of interferon (IFN) than the replication of vesicular stomatitis virus (VSV) or vaccinia virus (VV). In this study we examined the capacity of these three viruses to induce the expression of IFN when infecting avian cells. Efficient expression of both type-α and type-ß IFNs, as well as of the double-stranded RNA (dsRNA)-activated protein kinase (PKR), takes place in ARV-infected CEF, but not in cells infected with VSV or VV. PKR expression is not directly induced by ARV infection, but by the IFN secreted by ARV-infected cells. IFN induction in ARV-infected cells requires viral uncoating, but not viral gene expression, a situation similar to that reported for apoptosis induction by ARV-infected cells. However, our results demonstrate that IFN induction by ARV-infected CEF occurs by a caspase-independent mechanism.


Subject(s)
Interferon-alpha/immunology , Interferon-beta/immunology , Orthoreovirus, Avian/immunology , Vaccinia virus/immunology , Vesicular stomatitis Indiana virus/immunology , eIF-2 Kinase/immunology , Animals , Apoptosis/immunology , Caspases/metabolism , Cell Proliferation , Cells, Cultured , Chick Embryo , Chickens , Cricetinae , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Orthoreovirus, Avian/growth & development , Primary Cell Culture , Vaccinia virus/growth & development , Vesicular stomatitis Indiana virus/growth & development , Virus Replication , eIF-2 Kinase/biosynthesis
3.
Arch Virol ; 160(6): 1449-61, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25854689

ABSTRACT

Avian reovirus (ARV) causes viral arthritis, chronic respiratory diseases, retarded growth and malabsorption syndrome. It is well established that the ARV sigma-C protein induces apoptosis in host cells. However, the underlying molecular mechanism of this induction is still unclear. We report here the identification of eukaryotic elongation factor 1 alpha 1 (EEF1A1) as the interacting partner of σC. We found that σC-induced apoptosis in DF-1 cells could be completely abolished by knockdown of EEF1A1 by siRNA. Furthermore, knockdown of EEF1A1 markedly reduced ARV-induced apoptosis associated with decreased caspase-9 and -3 activation and cytochrome C release, leading to increased ARV growth in host cells. Thus, EEF1A1 plays a critical role in σC-induced apoptosis and inhibition of viral growth.


Subject(s)
Apoptosis , Capsid Proteins/physiology , Eukaryotic Initiation Factor-1/physiology , Orthoreovirus, Avian/physiology , Reoviridae Infections/physiopathology , Animals , Apoptosis/physiology , Blotting, Western , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , Chick Embryo/virology , Fluorescent Antibody Technique , HEK293 Cells/virology , Humans , Immunoprecipitation , Microscopy, Confocal , Orthoreovirus, Avian/growth & development , Peptide Elongation Factor 1/physiology , Reoviridae Infections/veterinary , Reoviridae Infections/virology , Two-Hybrid System Techniques
4.
J Biol Chem ; 288(5): 3571-84, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23233667

ABSTRACT

Autophagy has been shown to facilitate replication or production of avian reovirus (ARV); nevertheless, how ARV induces autophagy remains largely unknown. Here, we demonstrate that the nonstructural protein p17 of ARV functions as an activator of autophagy. ARV-infected or p17-transfected cells present a fast and strong induction of autophagy, resulting in an increased level of autophagic proteins Beclin 1 and LC3-II. Although autophagy was suppressed by 3-methyladenine or shRNAs targeting autophagic proteins (Beclin 1, ATG7, and LC3) as well as by overexpression of Bcl-2, viral transcription, σC protein synthesis, and virus yield were all significantly reduced, suggesting a key role of autophagosomes in supporting ARV replication. Furthermore, we revealed for the first time that p17 positively regulates phosphatase and tensin deleted on chromosome 10 (PTEN), AMP-activated protein kinase (AMPK), and dsRNA dependent protein kinase RNA (PKR)/eIF2α signaling pathways, accompanied by down-regulation of Akt and mammalian target of rapamycin complex 1, thereby triggering autophagy. By using p53, PTEN, PKR, AMPK, and p17 short hairpin RNA (shRNA), activation of signaling pathways and LC3-II levels was significantly suppressed, suggesting that p17 triggers autophagy through activation of p53/PTEN, AMPK, and PKR signaling pathways. Furthermore, colocalization of LC3 with viral proteins (p17 and σC), p62 with LAMP2 and LC3 with Rab7 was observed under a fluorescence microscope. The expression level of p62 was increased at 18 h postinfection and then slightly decreased 24 h postinfection compared with mock infection and thapsigargin treatment. Furthermore, disruption of autophagosome-lysosome fusion by shRNAs targeting LAMP2 or Rab7a resulted in inhibition of viral protein synthesis and virus yield, suggesting that formation of autolysosome benefits virus replication. Taken together, our results suggest that ARV induces formation of autolysosome but does not induce complete autophagic flux.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy , Orthoreovirus, Avian/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , AMP-Activated Protein Kinases/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Chickens , Enzyme Activation , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice , Microtubule-Associated Proteins/metabolism , Models, Biological , Orthoreovirus, Avian/growth & development , Orthoreovirus, Avian/physiology , PTEN Phosphohydrolase/genetics , Phagosomes/metabolism , RNA, Double-Stranded/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism , eIF-2 Kinase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Vet Pathol ; 44(2): 185-95, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17317795

ABSTRACT

The pathogenesis of 4 isolates of turkey-origin reovirus (NC/SEP-R44/03, NC/98, TX/98, and NC/85) and 1 chicken-origin reovirus (1733) was examined by infecting specific pathogen free (SPF) poults. These turkey-origin reovirus (TRV) isolates were collected from turkey flocks experiencing poult enteritis and are genetically distinct from previously reported avian reoviruses. Microscopic examination of the tissues collected from the TRV-infected poults revealed different degrees of bursal atrophy characterized by lymphoid depletion and increased fibroplasia between the bursal follicles. To understand the relationship between virus spread and replication, and the induction of lesions, immunohistochemical staining (IHC) for viral antigen, in situ hybridization (ISH) for the detection of viral RNA, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay for the detection of apoptosis in affected tissues was performed. Both IHC and ISH revealed viral antigen and RNA in the surface epithelial cells of the bursa, in macrophages in the interstitium of the bursa and, to lesser degree, in splenic red pulp macrophages and intestinal epithelial cells. Increased apoptosis of bursal lymphocytes and macrophages was observed at 2 and 5 days postinoculation. No lesions were found in tissues from poults inoculated with the virulent chicken-origin strain, however viral antigen was detected in the bursa and the intestine. Although all TRVs studied displayed similar tissue tropism, there were substantial differences in the severity of the lesions produced. Poults inoculated with NC/SEP-R44/03 or NC/98 had moderate to severe bursal atrophy, whereas poults inoculated with TX/98 or NC/85 presented a mild to moderate bursal lymphoid depletion. The lymphoid depletion observed in the bursa appears to be the effect of an indirectly induced apoptosis and would most likely result in immune dysfunction in poults infected with TRV.


Subject(s)
Orthoreovirus, Avian/growth & development , Poultry Diseases/pathology , Poultry Diseases/virology , Reoviridae Infections/veterinary , Turkeys , Animals , Apoptosis/physiology , Bursa of Fabricius/pathology , Bursa of Fabricius/virology , Immunohistochemistry/veterinary , In Situ Hybridization/veterinary , In Situ Nick-End Labeling/veterinary , Jejunum/pathology , Jejunum/virology , Liver/pathology , Liver/virology , Orthoreovirus, Avian/genetics , Reoviridae Infections/pathology , Reoviridae Infections/virology , Specific Pathogen-Free Organisms , Spleen/pathology , Spleen/virology
6.
Curr Top Microbiol Immunol ; 309: 67-85, 2006.
Article in English | MEDLINE | ID: mdl-16909897

ABSTRACT

Avian reoviruses are important pathogens that may cause considerable economic losses in poultry farming. Their genome expresses at least eight structural and four nonstructural proteins, three of them encoded by the S1 gene. These viruses enter cells by receptor-mediated endocytosis, and acidification of virus-containing endosomes is necessary for the virus to uncoat and release transcriptionally active cores into the cytosol. Avian reoviruses replicate within cytoplasmic inclusions of globular morphology, termed viral factories, which are not microtubule-associated, and which are formed by the nonstructural protein muNS. This protein also mediates the association of some viral proteins (but not of others) with inclusions, suggesting that the recruitment of viral proteins into avian reovirus factories has specificity. Avian reovirus morphogenesis is a complex and temporally controlled process that takes place exclusively within viral factories of infected cells. Core assembly takes place within the first 30 min after the synthesis of their protein components, and fully formed cores are then coated by outer-capsid polypeptides over the next 30 min to generate mature infectious reovirions. Based on data from avian reovirus studies and on results reported for other members of the Reoviridae family, we present a model for avian reovirus gene expression and morphogenesis.


Subject(s)
Morphogenesis , Orthoreovirus, Avian/growth & development , Animals , Gene Expression , Orthoreovirus, Avian/genetics , Virus Assembly
7.
Antiviral Res ; 64(1): 55-61, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15451179

ABSTRACT

Avian reoviruses (ARV) are economically important pathogens, especially in the poultry industry, where they cause viral arthritis and tenosynovitis. Mycophenolic acid (MPA) is an inhibitor of inosine monophosphate dehydrogenase (mainly used clinically for immunosuppression) that inhibits the replication of several viruses. We demonstrate in this study that MPA also is capable of inhibiting ARV replication in QM5 quail fibrosarcoma cells. The selectivity index of MPA in QM5 cells was determined as approximately 41. Concentrations of > or =3 microg/ml MPA inhibited infectious ARV progeny production in QM5 cells by more than 100-fold. Inhibition of ARV replication also was seen in other cell lines, including HD-11 and Vero. Addition of exogenous guanosine to MPA-treated ARV-infected QM5 cells restored viral replicative capacity to nearly normal levels.


Subject(s)
Antiviral Agents/pharmacology , Mycophenolic Acid/pharmacology , Orthoreovirus, Avian/drug effects , Orthoreovirus, Avian/physiology , Virus Replication/drug effects , Animals , Cell Line , Guanosine/pharmacology , Orthoreovirus, Avian/growth & development , Orthoreovirus, Avian/pathogenicity , Quail , Ribavirin/pharmacology
8.
J Mol Biol ; 341(2): 361-74, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15276829

ABSTRACT

We have recently shown that the avian reovirus non-structural protein microNS forms cytoplasmic inclusions in transfected cells and recruits sigmaNS to these structures. In the present study we further demonstrate that microNS mediates the association of the major core protein lambdaA, but not of sigmaA or sigmaC, with inclusions, indicating that the recruitment of viral proteins into avian reovirus factories has specificity. Thus, some proteins appear to be initially recruited to factories by association with microNS, whereas others are recruited subsequently through interaction with as-yet-unknown factors. We next used metabolic pulse-chase radiolabeling combined with cell fractionation and antibody immunoprecipitation to study the recruitment of newly synthesized viral polypeptides into viral factories and virus particles. The results of this combined approach revealed that avian reovirus morphogenesis is a complex and temporally controlled process that takes place exclusively within globular viral factories that are not microtubule-associated. Our findings further suggest that cores are assembled within the first 30 minutes after the synthesis of their polypeptide components, and that reovirion morphogenesis is completed over the next 30 minutes by the subsequent addition of outer capsid proteins.


Subject(s)
Chick Embryo/virology , Inclusion Bodies, Viral/chemistry , Orthoreovirus, Avian/metabolism , Viral Core Proteins/analysis , Viral Proteins/analysis , Animals , Cells, Cultured , Inclusion Bodies, Viral/metabolism , Molecular Sequence Data , Morphogenesis , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/growth & development , Precipitin Tests , Reoviridae Infections/genetics , Reoviridae Infections/metabolism , Siphoviridae , Viral Core Proteins/genetics , Viral Core Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins
9.
Vet Microbiol ; 91(4): 309-23, 2003 Feb 25.
Article in English | MEDLINE | ID: mdl-12477645

ABSTRACT

Monoclonal antibodies (MAbs) were prepared against avian reovirus S1133 protein sigmaA (esigmaA) synthesized in Escherichia coli. MAbs were characterized and used to develop a diagnostic test. Ten MAbs were selected for competitive binding assay following coupling with horseradish peroxidase. The results indicated that these MAbs delineated two epitopes I and II of esigmaA. An immuno-dot binding assay was used to detect the effect of denaturation on antibody recognition of the epitopes. All MAbs bound to esigmaA in its native form. After denaturation by boiling in SDS and 2-mercaptoethanol, the binding of MAbs recognizing epitope I was fully abolished. However, the reactivity of MAbs recognizing epitope II was not affected. MAbs 31 and 32, recognizing epitopes I and II, respectively, were selected for the cross-reactivity to heterologous reovirus strains. The results suggest that the two epitopes are highly conserved among these virus strains. A MAb capture enzyme-linked immunosorbent assay (ELISA) procedure was developed using MAbs 32 and 31 to detect reovirus protein sigmaA in samples from tendon tissues of infected bird and chicken embryo fibroblast (CEF) cell cultures. Avian reovirus sigmaA antigens in tendon specimens were detected from the inoculated birds as early as 2 days post-inoculation (PI), approximated a peak at 7 days PI, and maintained this until 16 days PI, then decreased gradually. A clear difference in absorbance values between the tendon samples of the avian reovirus- and mock-infected birds is obtained. Positive results were also obtained from avian reovirus-infected CEF and from the tendon tissues of naturally infected broilers. These results indicated that the MAb capture ELISA is a useful methods for the detection of avian reovirus from chickens suspected to have avian reovirus infections.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Chickens , Orthoreovirus, Avian/immunology , Poultry Diseases/virology , Reoviridae Infections/veterinary , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Viral , Antigens, Viral/chemistry , Antigens, Viral/genetics , Binding, Competitive , Chick Embryo , Enzyme-Linked Immunosorbent Assay/methods , Enzyme-Linked Immunosorbent Assay/veterinary , Epitope Mapping , Escherichia coli/genetics , Escherichia coli/metabolism , Immunoglobulin Isotypes , Mice , Mice, Inbred BALB C , Orthoreovirus, Avian/growth & development , Poultry Diseases/diagnosis , Precipitin Tests/veterinary , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Reoviridae Infections/diagnosis , Reoviridae Infections/virology , Specific Pathogen-Free Organisms , Tendons/virology
10.
Avian Pathol ; 32(4): 419-25, 2003 Aug.
Article in English | MEDLINE | ID: mdl-17585466

ABSTRACT

Two strains of avian reovirus were tested for their ability to survive on materials common to most poultry houses. The viruses survived longest and for at least 10 days on feathers, wood shavings and chicken feed, and for the shortest periods on wood (2 days), paper and cotton (4 days). There were some differences in survivability between the two strains. In most instances, the presence of faecal material increased the survival time, although in others it had the opposite effect. Reovirus survived for at least 10 days on the surface of eggshells when organic material was present. In drinking water, it survived for at least 10 weeks with little loss of infectivity. This could have implications for contamination of water supplies in poultry houses. It was shown that if cotton swabs are used for sampling, reovirus survives longer if they are pre-moistened with culture medium rather than used dry.


Subject(s)
Chickens/virology , Housing, Animal , Orthoreovirus, Avian/growth & development , Orthoreovirus, Avian/isolation & purification , Animal Feed/virology , Animals , Cotton Fiber , Egg Shell/virology , Feathers/virology , Feces , Glass , Metals , Paper , Polyethylene , Rubber , Time Factors , Vinyl Compounds , Wood/virology
11.
Poult Sci ; 81(11): 1661-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12455593

ABSTRACT

An avian reovirus, ARV-CU98, has recently been isolated from poults experiencing poult enteritis and mortality syndrome (PEMS). To further understand ARV-CU98 and its role in PEMS, the current study investigates interactions of ARV-CU98 with various cell types in vitro. When macrophages, B cells, T cells, and liver cells of chicken or turkey origin were co-incubated with ARV-CU98, only cells of liver origin demonstrated cytopathic effects, the presence of viral antigen, and reduced metabolic activity over time. Furthermore, distinctive pockets of viral particles were evident in electron microscopic examination of a chicken hepatocellular carcinoma (LMH) cell line, but not in a chicken macrophage cell line (MQ-NCSU) co-incubated with virus. Additional evidence of viral replication in LMH, cells but not MQ-NCSU cells was demonstrated by the presence of two viral bands (43 and 145 kD size) in cell lysates from LMH cells exposed to ARV-CU98. Although not capable of being infected by ARV-CU98, MQ-NCSU cells do appear to be activated by the virus since IL-1 mRNA expression is increased in MQ-NCSU cells 2 h after addition of the virus. LMH cells exposed to the virus demonstrate a decrease in IL-1 mRNA expression by 8 to 10 h after addition of the virus, perhaps corresponding to the initiation of infection by the virus. In conclusion, this study demonstrates that ARV-CU98 actively infects and replicates in LMH cells, but not in lymphocytes or macrophages, suggesting that the liver may be a target and site of replication of ARV-CU98 in poults experiencing PEMS.


Subject(s)
Chickens , Orthoreovirus, Avian/pathogenicity , Poult Enteritis Mortality Syndrome/virology , Reoviridae Infections/veterinary , Turkeys , Animals , Antigens, Viral/analysis , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line , Cells, Cultured , Cytopathogenic Effect, Viral , Interleukin-1/genetics , Interleukin-1/metabolism , Liver/cytology , Liver/virology , Macrophages/metabolism , Macrophages/virology , Microscopy, Electron/veterinary , Orthoreovirus, Avian/growth & development , Orthoreovirus, Avian/immunology , Poult Enteritis Mortality Syndrome/metabolism , Poultry Diseases/metabolism , Poultry Diseases/virology , RNA, Messenger/metabolism , Reoviridae Infections/virology , Specific Pathogen-Free Organisms , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Tumor Cells, Cultured , Virus Replication
12.
J Virol ; 76(16): 7932-41, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12133997

ABSTRACT

The cytopathic effect evidenced by cells infected with avian reovirus S1133 suggests that this virus may induce apoptosis in primary cultures of chicken embryo fibroblasts. In this report we present evidence that avian reovirus infection of cultured cells causes activation of the intracellular apoptotic program and that this activation takes place during an early stage of the viral life cycle. The ability of avian reoviruses to induce apoptosis is not restricted to a particular virus strain or to a specific cell type, since different avian reovirus isolates were able to induce apoptosis in several avian and mammalian cell lines. Apoptosis was also provoked in ribavirin-treated avian reovirus-infected cells and in cells infected with UV-irradiated reovirions, indicating that viral mRNA synthesis and subsequent steps in viral replication are not needed for apoptosis induction in avian reovirus-infected cells and that the number of inoculated virus particles, not their infectivity, is the critical factor for apoptosis induction by avian reovirus. Our finding that apoptosis is no longer induced when intracellular viral uncoating is blocked indicates that intraendosomal virion disassembly is required for apoptosis induction and that attachment and uptake of parental reovirions are not sufficient to cause apoptosis. Taken together, our results suggest that apoptosis is triggered from within the infected cell by viral products generated after intraendosomal uncoating of parental reovirions.


Subject(s)
Apoptosis/physiology , Orthoreovirus, Avian/pathogenicity , Animals , Antimetabolites/pharmacology , Antiviral Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Capsid/physiology , Cells, Cultured , Chick Embryo , Gene Expression , Genes, Viral , HeLa Cells , Humans , L Cells , Mice , Orthoreovirus, Avian/genetics , Orthoreovirus, Avian/growth & development , Orthoreovirus, Avian/physiology , Ribavirin/pharmacology , Species Specificity , Ultraviolet Rays , Virulence
13.
Virus Res ; 81(1-2): 103-11, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11682129

ABSTRACT

The replication of two pathotypes of avian reovirus, 1733 and 2177 in transformed chicken lymphoid and myeloid cell lines was examined, showing that only the macrophage cell line, HD11, supports replication. The virulent strain 1733 causes a lytic infection producing 100-1000 fold more virus than the avirulent strain 2177. Cells infected with strain 2177 display delayed viral RNA and protein synthesis as well as a suppressed expression of the major capsid protein muB. These features may contribute to the lower virulence of the strain 2177 in their natural host in vivo.


Subject(s)
Macrophages/virology , Orthoreovirus, Avian/growth & development , Orthoreovirus, Avian/pathogenicity , Animals , B-Lymphocytes , Cell Line , Cell Line, Transformed , Chick Embryo , Chickens , Orthoreovirus, Avian/metabolism , Reoviridae Infections/virology , T-Lymphocytes , Virulence , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...