Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Comp Med ; 70(4): 359-369, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32674749

ABSTRACT

The unexpected seroconversion of sentinel mice in our facility to murine T lymphotrophic virus (MTLV) positivity led to our identification of a novel murine astrovirus that we designated murine astrovirus 2 (MuAstV-2). During our investigation, MuAstV-2 was found to be a contaminant of the T helper cell line (D10. G4.1) that was used to generate the MTLV antigen that we included in the multiplex fluorometric immunoassay (MFIA) that we used for sentinel screening. We eventually determined that cross-reactivity with the astrovirus generated a positive result in the MTLV assay. A confirmatory immunofluorometric assay (IFA) using the same MTLV-infected cell line yielded a similar result. However, the use of antigen prepared from MTLV-infected neonatal mouse thymus did not reproduce a positive result, leading us to suspect that the seroreactivity we had observed was not due to infection with MTLV. A mouse antibody production test showed that mice inoculated with naïve D10. G4.1 cells and their contact sentinels tested positive for MTLV using cell-line generated antigen, but tested negative in assays using MTLV antigen produced in mice. Metagenomic analysis was subsequently used to identify MuAstV-2 in feces from 2 sentinel mice that had recently seroconverted to MTLV. Two closely related astrovirus sequences (99.6% capsid identity) were obtained and shared 95% capsid amino acid identity with the MuAstV-2 virus sequenced from the D10. G4.1 cell line. These viruses are highly divergent from previously identified murine astroviruses, displaying <30% capsid identity, yet were closely related to murine astrovirus 2 (85% capsid identity), which had recently been isolated from feral mice in New York City. A MuAstV-2 specific PCR assay was developed and used to eradicate MuAstV-2 from the infected colony using a test and cull strategy. The newly identified MuAstV2 readily transmits to immunocompetent mouse strains by fecal-oral exposure, but fails to infect NOD-Prkdcem26Cd52Il2rgem26Cd22/NjuCrl (NCG) mice, which have significantly impaired adaptive and innate immune systems. Neither immunocompetent nor immunodeficient mice showed any astrovirus-associated pathology. MuAstV-2 may provide a valuable model for the study of specific aspects of astrovirus pathogenesis and virus-host interactions.


Subject(s)
Astroviridae Infections/metabolism , Animals , Astroviridae , Astroviridae Infections/virology , Cell Line , Feces/virology , Genome, Viral , Immunocompetence/genetics , Mice/virology , Rodent Diseases/virology , T-Lymphocytes, Helper-Inducer/immunology
2.
Poult Sci ; 99(4): 1967-1974, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32241477

ABSTRACT

In 2018, a new goose astrovirus (GAstrV) was reported in China, which causes 2 to 20% deaths in 4- to 16-day-old goslings causing great damages to the livestock industry. Gout is the typical feature of GAstrV infection in goslings. However, the mechanism of gout formation remains unclear. In the present study, 2-day-old goslings were infected intramuscularly with GAstrV for 14 D. One quarter of the infected goslings died, and typical gout pathological changes were found in the dead infected goslings. Pathological changes were observed in the morphology of the kidney and liver, such as degeneration, necrosis, and inflammatory cell infiltration. Accordingly, a high virus load was found in both organs. The serum level of uric acid in the inoculated goslings was higher, whereas no differences were found in levels of creatinine, calcium, and phosphorus. Moreover, the xanthine dehydrogenase (XOD) and adenosine deaminase (ADA) activities and the mRNA levels of xanthine dehydrogenase, adenosine deaminase, phosphoribosyl pyrophosphate amidotransferase, and phosphoribosyl pyrophosphate synthetase 1 in livers increased, wheres the multidrug resistance-associated protein 4 mRNA level and Na-K-ATPase activity in the kidneys decreased. These results showed that GAstrV infection could cause lesions on the liver and kidney and then increase the expression or activity of enzymes related to uric acid production in the liver and decrease renal excretion function, which contribute to hyperuricemia and gout formation.


Subject(s)
Astroviridae Infections/veterinary , Avian Proteins/genetics , Carrier Proteins/genetics , Geese , Gout/veterinary , Poultry Diseases/metabolism , Uric Acid/metabolism , Animals , Astroviridae Infections/complications , Astroviridae Infections/metabolism , Astroviridae Infections/virology , Avastrovirus/physiology , Avian Proteins/metabolism , Carrier Proteins/metabolism , Disease Models, Animal , Feces/chemistry , Gout/metabolism , Gout/virology , Kidney/metabolism , Liver/chemistry , Poultry Diseases/virology , Purines/metabolism
3.
Viruses ; 13(1)2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33396308

ABSTRACT

Although human astroviruses (HAstVs) are important agents of gastroenteritis in young children, the studies aimed at characterizing their biology have been limited, in particular regarding their cell entry process. It has been shown that HAstV serotype 8 enters human cells by a classical clathrin-mediated endocytosis pathway; however, the cell receptor or other cell entry factors that may be relevant for an efficient viral infection are unknown. In this work we used a far-Western blotting approach to identify cellular proteins that interact with the recombinant capsid spike proteins of HAstV serotypes 1, 2, and 8, synthesized in Escherichia coli. We identified the 72 kDa protein disulfide isomerase A4 (PDIA4) as a binding partner for HAstV-1 and -8 spikes, but not for the HAstV-2 spike. In agreement with this observation, the PDI inhibitor 16F16 strongly blocked infection by HAstV serotypes 1 and 8, but not serotype 2. RNA interference of PDIA4 expression selectively blocked HAstV-8 infectivity. We also showed that the PDI activity does not affect virus binding or internalization but is required for uncoating of the viral genome.


Subject(s)
Astroviridae Infections/metabolism , Astroviridae Infections/virology , Host-Pathogen Interactions , Mamastrovirus/physiology , Protein Disulfide-Isomerases/metabolism , Virus Uncoating , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Cells, Cultured , Humans , Mamastrovirus/drug effects , Protein Binding , Virus Internalization
4.
J Virol ; 88(5): 2452-60, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24335315

ABSTRACT

Human astroviruses (HAstV) are a frequent cause of gastroenteritis in young children and immunocompromised patients. To understand the early steps of HAstV infection in the highly permissive Caco-2 cell line, the binding and entry processes of the virus were characterized. The half-time of virus binding to the cell surface was about 10 min, while virus decapsidation took around 130 min. Drugs affecting clathrin-mediated endocytosis, endosome acidification, and actin filament polymerization, as well as those that reduce the presence of cholesterol in the cell membrane, decreased the infectivity of the virus. The infection was also reduced by silencing the expression of the clathrin heavy chain (CHC) by RNA interference or by overexpression of dominant-negative mutants of dynamin 2 and Eps15. Furthermore, the entry of HAstV apparently depends on the maturation of endosomes, since the infection was reduced by silencing the expression of Rab7, a small GTPase involved in the early- to late-endosome maturation. Altogether, our results suggest that HAstV enters Caco-2 cells using a clathrin-dependent pathway and reaches late endosomes to enter cells. Here, we have characterized the mechanism used by human astroviruses, important agents of gastroenteritis in children, to gain entry into their host cells. Using a combination of biochemical and genetic tools, we found that these viruses enter Caco-2 cells using a clathrin-dependent endocytic pathway, where they most likely need to travel to late endosomes to reach the cytoplasm and begin their replication cycle.


Subject(s)
Mamastrovirus/physiology , Virus Internalization , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Antiviral Agents/pharmacology , Astroviridae Infections/genetics , Astroviridae Infections/metabolism , Astroviridae Infections/virology , Cell Line , Clathrin/genetics , Clathrin/metabolism , Dynamins/genetics , Dynamins/metabolism , Endoribonucleases/metabolism , Fungal Proteins/metabolism , Gene Silencing , Humans , Mamastrovirus/drug effects , Mutation , Virus Attachment , Virus Release , Virus Replication/drug effects , Virus Uncoating , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
5.
J Virol ; 86(22): 12262-70, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22951832

ABSTRACT

The mechanisms of astrovirus pathogenesis are largely unknown, in part due to a lack of a small-animal model of disease. Using shotgun sequencing and a custom analysis pipeline, we identified two novel astroviruses capable of infecting research mice, murine astrovirus (MuAstV) STL1 and STL2. Subsequent analysis revealed the presence of at least two additional viruses (MuAstV STL3 and STL4), suggestive of a diverse population of murine astroviruses in research mice. Complete genomic characterization and subsequent phylogenetic analysis showed that MuAstV STL1 to STL4 are members of the mamastrovirus genus and are likely members of a new mamastrovirus genogroup. Using Rag1(-/-) mice deficient in B and T cells, we demonstrate that adaptive immunity is required to control MuAstV infection. Furthermore, using Stat1(-/-) mice deficient in innate signaling, we demonstrate a role for the innate immune response in the control of MuAstV replication. Our results demonstrate that MuAstV STL permits the study of the mechanisms of astrovirus infection and host-pathogen interactions in a genetically manipulable small-animal model. Finally, we detected MuAstV in commercially available mice, suggesting that these viruses may be present in academic and commercial research mouse facilities, with possible implications for interpretation of data generated in current mouse models of disease.


Subject(s)
Adaptive Immunity , Astroviridae Infections/metabolism , Astroviridae Infections/virology , Mamastrovirus/metabolism , Animals , Disease Models, Animal , Female , Genome, Viral , Homeodomain Proteins/genetics , Mamastrovirus/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Open Reading Frames , Phylogeny , STAT1 Transcription Factor/genetics , Sequence Analysis, DNA , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...