Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Virol ; 98(2): e0167723, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38240590

ABSTRACT

Rotavirus infection is a leading cause of gastroenteritis in children worldwide; the genome of this virus is composed of 11 segments of dsRNA packed in a triple-layered protein capsid. Here, we investigated the role of nucleolin, a protein with diverse RNA-binding domains, in rotavirus infection. Knocking down the expression of nucleolin in MA104 cells by RNA interference resulted in a remarkable 6.3-fold increase in the production of infectious rhesus rotavirus (RRV) progeny, accompanied by an elevated synthesis of viral mRNA and genome copies. Further analysis unveiled an interaction between rotavirus segment 10 (S10) and nucleolin, potentially mediated by G-quadruplex domains on the viral genome. To determine whether the nucleolin-RNA interaction regulates RRV replication, MA104 cells were transfected with AGRO100, a compound that forms G4 structures and selectively inhibits nucleolin-RNA interactions by blocking the RNA-binding domains. Under these conditions, viral production increased by 1.5-fold, indicating the inhibitory role of nucleolin on the yield of infectious viral particles. Furthermore, G4 sequences were identified in all 11 RRV dsRNA segments, and transfection of oligonucleotides representing G4 sequences in RRV S10 induced a significant increase in viral production. These findings show that rotavirus replication is negatively regulated by nucleolin through the direct interaction with the viral RNAs by sequences forming G4 structures.IMPORTANCEViruses rely on cellular proteins to carry out their replicative cycle. In the case of rotavirus, the involvement of cellular RNA-binding proteins during the replicative cycle is a poorly studied field. In this work, we demonstrate for the first time the interaction between nucleolin and viral RNA of rotavirus RRV. Nucleolin is a cellular protein that has a role in the metabolism of ribosomal rRNA and ribosome biogenesis, which seems to have regulatory effects on the quantity of viral particles and viral RNA copies of rotavirus RRV. Our study adds a new component to the current model of rotavirus replication, where cellular proteins can have a negative regulation on rotavirus replication.


Subject(s)
Nucleolin , RNA, Viral , Rotavirus Infections , Rotavirus , Humans , Nucleolin/metabolism , RNA, Viral/genetics , Rotavirus/physiology , Rotavirus Infections/virology , Virus Replication
2.
J Virol ; 96(14): e0066522, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35762760

ABSTRACT

Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients, and its recent propagation in cell culture has opened the possibility to study its biology. Unlike classical human astroviruses, VA1 growth was found to be independent of trypsin during virus replication in vitro. In this work, we show that despite its independence on trypsin activation for cell infection, the VA1 capsid precursor protein, of 86 kDa (VP86), is processed intracellularly, and this proteolytic processing is important for astrovirus VA1 infectivity. Antibodies raised against different regions of the capsid precursor showed that the polyprotein can be processed starting at either its amino- or carboxy-terminal end, and they allowed us to identify those proteins of about 33 (VP33) and 38 (VP38) kDa constitute the core and the spike proteins of the mature infectious virus particles, respectively. The amino-terminal end of the spike protein was found to be Thr-348. Whether the protease involved in intracellular cleavage of the capsid precursor is of viral or cellular origin remains to be determined, but the cleavage is independent of caspases. Also, trypsin is able to degrade the capsid precursor but has no effect on VP33 and VP38 proteins when assembled into virus particles. These studies provide the basis for advancement of the knowledge of astrovirus VA1 cell entry and replication. IMPORTANCE Human astrovirus VA1 has been associated with neurological disease in immunocompromised patients. Its recent propagation in cell culture has facilitated the study of its biology. In this work, we show that despite the ability of this virus to grow in the absence of trypsin, a marked feature of human classical astroviruses, the capsid precursor protein of astrovirus VA1 is cleaved intracellularly to yield the mature infectious particles, formed by two polypeptides, VP33 that constitutes the core domain of the virus particle, and VP38 that forms the spike of the virus. These studies provide a platform to advance our knowledge on astrovirus VA1 cell entry and replication.


Subject(s)
Astroviridae Infections , Capsid Proteins , Mamastrovirus , Protein Precursors , Astroviridae Infections/virology , Caco-2 Cells , Capsid/metabolism , Capsid Proteins/metabolism , Humans , Intracellular Space/virology , Mamastrovirus/physiology , Protein Precursors/metabolism , Trypsin/metabolism
3.
PLoS One ; 17(1): e0263114, 2022.
Article in English | MEDLINE | ID: mdl-35077513

ABSTRACT

In many countries a second wave of infections caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has occurred, triggering a shortage of reagents needed for diagnosis and compromising the capacity of laboratory testing. There is an urgent need to develop methods to accelerate the diagnostic procedures. Pooling samples represents a strategy to overcome the shortage of reagents, since several samples can be tested using one reaction, significantly increasing the number and speed with which tests can be carried out. We have reported the feasibility to use a direct lysis procedure of saliva as source for RNA to SARS-CoV-2 genome detection by reverse transcription quantitative-PCR (RT-qPCR). Here, we show that the direct lysis of saliva pools, of either five or ten samples, does not compromise the detection of viral RNA. In addition, it is a sensitive, fast, and inexpensive method that can be used for massive screening, especially considering the proximity of the reincorporation of activities in universities, offices, and schools.


Subject(s)
COVID-19 Nucleic Acid Testing/methods , COVID-19/diagnosis , Saliva/virology , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19 Nucleic Acid Testing/standards , Humans , Mass Screening/methods , Mass Screening/standards , Quarantine/standards , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , SARS-CoV-2/pathogenicity , Sensitivity and Specificity
4.
Viruses ; 13(5)2021 04 23.
Article in English | MEDLINE | ID: mdl-33922604

ABSTRACT

Swine enteric viral infections are responsible for substantial economic losses in the pork industry worldwide. Porcine epidemic diarrhea (PEDV) is one of the main causative agents of diarrhea in lactating pigs, and reports of PEDV coinfection with other enteric viruses highlight the importance of viral interactions for disease presentation and outcomes. Using next-generation sequencing (NGS) and sequence analyses from samples taken from piglets with acute diarrhea, we explored the possible interactions between PEDV and other less reported pathogens. PEDV coinfection with porcine kobuvirus (PKV) was detected in 36.4% (27/74) of samples. Full genomes from porcine coronavirus and kobuvirus were obtained, as was a partial porcine sapovirus genome (PSaV). The phylogenetic results show the clustering of these strains corresponding to the geographical relationship. To our knowledge, this is the first full genome and isolation report for porcine kobuvirus in México, as well as the first phylogenetic analysis for porcine sapovirus in the country. The NGS approach provides a better perspective of circulating viruses and other pathogens in affected production units.


Subject(s)
Coinfection/virology , Coronavirus Infections/virology , Kobuvirus/genetics , Kobuvirus/isolation & purification , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/isolation & purification , Animals , Coinfection/epidemiology , Coronavirus Infections/epidemiology , Diarrhea/virology , Feces/virology , Genome, Viral , Kobuvirus/classification , Mexico/epidemiology , Molecular Diagnostic Techniques , Phylogeny , Porcine epidemic diarrhea virus/classification , Sapovirus/genetics , Sequence Analysis , Swine , Swine Diseases/virology
5.
Curr Opin Virol ; 44: 57-65, 2020 10.
Article in English | MEDLINE | ID: mdl-32683123

ABSTRACT

Astroviruses are one of the main causes of gastroenteritis of medical and veterinary relevance worldwide. Recently, these viruses were associated with neurological disease in mammals, including humans. Reverse genetics systems are the most powerful tool to improve our understanding of the virus replication, and eventually to develop safe vaccine candidates. In the present review, it is summarized the current knowledge on the different strategies used to develop reverse genetics systems for mamastroviruses and avastroviruses, and some of the biological answers that have provided are discussed.


Subject(s)
Astroviridae Infections/prevention & control , Astroviridae Infections/veterinary , Astroviridae/genetics , Astroviridae/immunology , Genome, Viral , Reverse Genetics/methods , Animals , Astroviridae Infections/immunology , Cell Line , Gastroenteritis/prevention & control , Gastroenteritis/veterinary , Gastroenteritis/virology , Humans , Plasmids/genetics , Plasmids/immunology
6.
J Clin Microbiol ; 58(10)2020 09 22.
Article in English | MEDLINE | ID: mdl-32703816

ABSTRACT

As part of any plan to lift or ease the confinement restrictions that are in place in many different countries, there is an urgent need to increase the capacity of laboratory testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Detection of the viral genome through reverse transcription-quantitative PCR (RT-qPCR) is the gold standard for this virus; however, the high demand of the materials and reagents needed to sample individuals, purify the viral RNA, and perform the RT-qPCR has resulted in a worldwide shortage of several of these supplies. Here, we show that directly lysed saliva samples can serve as a suitable source for viral RNA detection that is less expensive and can be as efficient as the classical protocol, which involves column purification of the viral RNA. In addition, it bypasses the need for swab sampling, decreases the risk of the health care personnel involved in the testing process, and accelerates the diagnostic procedure.


Subject(s)
Betacoronavirus/isolation & purification , Saliva/virology , Specimen Handling/methods , Betacoronavirus/genetics , COVID-19 Testing , Clinical Laboratory Techniques , Coronavirus Infections/diagnosis , Coronavirus Infections/epidemiology , Diagnostic Tests, Routine , Genome, Viral/genetics , Humans , Nasopharynx/virology , Oropharynx/virology , RNA, Viral/genetics , RNA, Viral/isolation & purification , Real-Time Polymerase Chain Reaction , SARS-CoV-2 , Viral Load
7.
Virology ; 535: 130-135, 2019 09.
Article in English | MEDLINE | ID: mdl-31299489

ABSTRACT

Human astroviruses (HAstVs) are a frequent cause of gastroenteritis in young children and immunocompromised patients. The current report describes a new approach to recover genetically defined HAstVs through the use of a reverse genetics system based on a single DNA plasmid. This plasmid, carrying the full-length virus genome under a T7 promoter, is directly transfected into cells expressing T7 RNA polymerase, resulting in the rapid and robust recovery of infectious HAstV. The efficiency of the system was tested with the generation of a chimeric astrovirus having the HAstV serotype 1 replication machinery and the capsid derived from a HAstV serotype 8 virus. This new system provides an efficient and reproducible method to deepen our knowledge of astrovirus biology.


Subject(s)
Mamastrovirus/growth & development , Mamastrovirus/genetics , Reverse Genetics/methods , DNA, Complementary/genetics , Genetic Vectors , Genome, Viral , Humans , Plasmids
8.
Viruses ; 10(8)2018 07 26.
Article in English | MEDLINE | ID: mdl-30049969

ABSTRACT

Hepatitis E virus (HEV) is an emerging public health problem with an estimated 20 million infections each year. In Mexico, Orthohepevirus A, genotype 2, has been reported in humans, but genotype 3 has only been reported in swine (zoonotic). No diagnostic tests are publicly available in Mexico, and only partial sequences have been reported from swine samples. Hence, research is necessary to determine circulating strains, understand the features and dynamics of infection on pig farms, determine how to implement surveillance programs, and to assess public health risks. In this study, a next-generation sequencing (NGS) approach was applied to obtain a complete genome of swine HEV. Liver, feces, and bile samples were taken at slaughterhouses and a farm in Mexico. RT-PCR was used to determine positive samples and confirmed by Sanger sequencing. Of the 64 slaughterhouse samples, one bile sample was positive (B1r) (1.56%). Of 21 sample pools from farm animals, 14 were positive (66.66%), representing all stages of production. A complete sequence strain MXCDg3_B1c|_2016 was obtained from the bile of a domestic swine in the fattening stage. In addition, two partial sequences-MXCDg3_H2cons|_2016 (1473 nt) and MXCDg3_C3Acons|_2016 (4777 nt)-were obtained from sampled farm animals. Comparison with all reported genome HEV sequences showed similarity to genotype 3 subgenotype a (G3a), which has been previously reported in acute cases of human hepatitis in the US, Colombia, China, and Japan.


Subject(s)
Genome, Viral , Genotype , Hepatitis E virus/genetics , Hepatitis E/veterinary , Phylogeny , Swine Diseases/virology , Abattoirs , Animals , Animals, Domestic , Bile/virology , Feces/virology , High-Throughput Nucleotide Sequencing , Liver/virology , Mexico , Swine/virology , Zoonoses/virology
9.
mBio ; 8(1)2017 02 14.
Article in English | MEDLINE | ID: mdl-28196955

ABSTRACT

The Hom-1 vesivirus was reported in 1998 following the inadvertent transmission of the animal calicivirus San Miguel sea lion virus to a human host in a laboratory. We characterized the Hom-1 strain and investigated the mechanism by which human cells could be infected. An expression library of 3,559 human plasma membrane proteins was screened for reactivity with Hom-1 virus-like particles, and a single interacting protein, human junctional adhesion molecule 1 (hJAM1), was identified. Transient expression of hJAM1 conferred susceptibility to Hom-1 infection on nonpermissive Chinese hamster ovary (CHO) cells. Virus infection was markedly inhibited when CHO cells stably expressing hJAM were pretreated with anti-hJAM1 monoclonal antibodies. Cell lines of human origin were tested for growth of Hom-1, and efficient replication was observed in HepG2, HuH7, and SK-CO15 cells. The three cell lines (of hepatic or intestinal origin) were confirmed to express hJAM1 on their surface, and clustered regularly interspaced short palindromic repeats/Cas9-mediated knockout of the hJAM1 gene in each line abolished Hom-1 propagation. Taken together, our data indicate that entry of the Hom-1 vesivirus into these permissive human cell lines is mediated by the plasma membrane protein hJAM1 as a functional receptor.IMPORTANCE Vesiviruses, such as San Miguel sea lion virus and feline calicivirus, are typically associated with infection in animal hosts. Following the accidental infection of a laboratory worker with San Miguel sea lion virus, a related virus was isolated in cell culture and named Hom-1. In this study, we found that Hom-1 could be propagated in a number of human cell lines, making it the first calicivirus to replicate efficiently in cultured human cells. Screening of a library of human cell surface membrane proteins showed that the virus could utilize human junctional adhesion molecule 1 as a receptor to enter cells and initiate replication. The Hom-1 virus presents a new system for the study of calicivirus biology and species specificity.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Vesivirus/physiology , Virus Replication , Animals , CHO Cells , Cats , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/isolation & purification , Cell Membrane/chemistry , Cell Membrane/genetics , Cricetinae , Cricetulus , Humans , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/isolation & purification , Receptors, Virus/isolation & purification , Vesivirus/growth & development
10.
Viruses ; 8(6)2016 06 07.
Article in English | MEDLINE | ID: mdl-27338442

ABSTRACT

The general stress and innate immune responses are closely linked and overlap at many levels. The outcomes of these responses serve to reprogram host expression patterns to prevent viral invasions. In turn, viruses counter attack these cell responses to ensure their replication. The mechanisms by which viruses attempt to control host cell responses are as varied as the number of different virus families. One of the most recurrent strategies used by viruses to control the antiviral response of the cell is to hijack the translation machinery of the host, such that viral proteins are preferentially synthesized, while the expression of the stress and antiviral responses of the cell are blocked at the translation level. Here, we will review how rotaviruses, an important agent of acute severe gastroenteritis in children, overcome the stress responses of the cell to establish a productive infectious cycle.


Subject(s)
Host-Pathogen Interactions , Protein Biosynthesis , Rotavirus Infections/immunology , Rotavirus Infections/pathology , Rotavirus/immunology , Rotavirus/pathogenicity , Stress, Physiological , Gene Expression Regulation , Humans , Immunity, Innate , Virus Replication
11.
Virology ; 492: 232-41, 2016 May.
Article in English | MEDLINE | ID: mdl-26971245

ABSTRACT

Noroviruses are diverse positive-strand RNA viruses associated with acute gastroenteritis. Cross-reactive epitopes have been mapped primarily to conserved sequences in the capsid VP1 Shell (S) domain, and strain-specific epitopes to the highly variable Protruding (P) domain. In this work, we investigated a strain-specific linear epitope defined by MAb NV10 that was raised against prototype (Genogroup I.1) strain Norwalk virus (NV). Using peptide scanning and mutagenesis, the epitope was mapped to amino acids 21-32 (LVPEVNASDPLA) of the NV S domain, and its specificity was verified by epitope transfer and reactivity with a recombinant MAb NV10 single-chain variable fragment (scFv). Comparative structural modeling of the NV10 strain-specific and the broadly cross-reactive TV20 epitopes identified two internal non-overlapping sites in the NV shell, corresponding to variable and conserved amino acid sequences among strains, respectively. The S domain, like the P domain, contains strain-specific epitopes that contribute to the antigenic diversity among the noroviruses.


Subject(s)
Antibodies, Viral/chemistry , Capsid Proteins/chemistry , Epitopes/chemistry , Norwalk virus/immunology , Single-Chain Antibodies/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Antibodies, Viral/biosynthesis , Calicivirus, Feline/genetics , Calicivirus, Feline/metabolism , Capsid/chemistry , Capsid/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , Gene Expression , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Norwalk virus/genetics , Protein Structure, Tertiary , Sequence Alignment , Single-Chain Antibodies/biosynthesis
12.
Virology ; 489: 51-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26707270

ABSTRACT

Feline calicivirus depends on host-cell proteins for its replication. We previously showed that knockdown of nucleolin (NCL), a phosphoprotein involved in ribosome biogenesis, resulted in the reduction of FCV protein synthesis and virus yield. Here, we found that NCL may not be involved in FCV binding and entry into cells, but it binds to both ends of the FCV genomic RNA, and stimulates its translation in vitro. AGRO100, an aptamer that specifically binds and inactivates NCL, caused a strong reduction in FCV protein synthesis. This effect could be reversed by the addition of full-length NCL but not by a ΔrNCL, lacking the N-terminal domain. Consistent with this, FCV infection of CrFK cells stably expressing ΔrNCL led to a reduction in virus protein translation. These results suggest that NCL is part of the FCV RNA translational complex, and that the N-terminal part of the protein is required for efficient FCV replication.


Subject(s)
Caliciviridae Infections/veterinary , Calicivirus, Feline/genetics , Cat Diseases/metabolism , Cat Diseases/virology , Phosphoproteins/metabolism , Protein Biosynthesis , RNA, Viral/genetics , RNA-Binding Proteins/metabolism , Animals , Caliciviridae Infections/genetics , Caliciviridae Infections/metabolism , Caliciviridae Infections/virology , Calicivirus, Feline/physiology , Cat Diseases/genetics , Cats , Cell Line , Host-Pathogen Interactions , Phosphoproteins/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Nucleolin
13.
J Virol Methods ; 217: 1-7, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25698463

ABSTRACT

Reverse genetics systems constitute one of the most important and powerful tools to study the molecular biology of viruses. We developed a new strategy for the recovery of murine norovirus from a single plasmid in which a bacteriophage T7 RNA polymerase (T7pol) promoter for transcription and an EMCV IRES for efficient translation were engineered immediately upstream of the viral genome. Infectious noroviruses were recovered following transfection of the newly designed plasmid into nonpermissive BHK-21 and HEK293T cell lines that were engineered to express T7pol constitutively. Recovery of the virus did not require the presence of a ribozyme at the 3'-end of the virus genome. The strategy worked also for the efficient recovery of feline calicivirus in these normally nonpermissive cell types. This simplified reverse genetics approach may be broadly applicable to other caliciviruses.


Subject(s)
Calicivirus, Feline/physiology , DNA-Directed RNA Polymerases/metabolism , Encephalomyocarditis virus/genetics , Internal Ribosome Entry Sites , Norovirus/physiology , Plasmids , Reverse Genetics/methods , Viral Proteins/metabolism , Animals , Calicivirus, Feline/genetics , Cell Line , Cricetinae , DNA-Directed RNA Polymerases/genetics , Humans , Norovirus/genetics , Protein Biosynthesis , Transcription, Genetic , Viral Proteins/genetics
14.
Virus Genes ; 48(1): 96-110, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24217871

ABSTRACT

Feline calicivirus (FCV) is a common cause of mild to severe upper respiratory tract disease (URTD) in cats. FCV strain 21223 was isolated from a kitten with severe pneumonia in a disease outbreak with unusually high mortality (35 %) that occurred in a Missouri feline colony in 1995-1996. Phylogenetic analysis of the genome sequence of strain 21223 indicated the emergence of a new FCV strain. Analysis of the full-length genome sequence of a closely related (99.5 % nucleotide identity) strain, 3786, obtained from an asymptomatic animal in the same colony four months later, showed the presence of seven amino acid substitutions, with six of them located in the VP1 capsid sequence encoded by ORF2. Comparative analysis of the E-region sequences (426-521 aa ORF2) presumably involved in virus-host cell receptor interactions did not identify amino acid substitutions unique to the virulent strain. We determined the complete genome sequences of four virus isolates that were collected in regional catteries in the months following the outbreak that were associated with different manifestations of the disease (URTD, chronic stomatitis, and gingivitis). We show that genetically distinct FCV strains were cocirculating in the area, and no apparent correlation could be made between overall sequence and observed disease.


Subject(s)
Caliciviridae Infections/veterinary , Calicivirus, Feline/classification , Calicivirus, Feline/genetics , Cat Diseases/pathology , Cat Diseases/virology , Animals , Asymptomatic Diseases , Caliciviridae Infections/pathology , Caliciviridae Infections/virology , Calicivirus, Feline/isolation & purification , Capsid Proteins/genetics , Cats , Cluster Analysis , Disease Outbreaks , Genome, Viral , Missouri/epidemiology , Molecular Sequence Data , Mutation, Missense , Phylogeny , RNA, Viral/genetics , Sequence Analysis, DNA , Sequence Homology
15.
PLoS One ; 8(6): e67592, 2013.
Article in English | MEDLINE | ID: mdl-23805319

ABSTRACT

Noroviruses are major pathogens associated with acute gastroenteritis. They are diverse viruses, with at least six genogroups (GI-GVI) and multiple genotypes defined by differences in the major capsid protein, VP1. This diversity has challenged the development of broadly cross-reactive vaccines as well as efficient detection methods. Here, we report the characterization of a broadly cross-reactive monoclonal antibody (MAb) raised against the capsid protein of a GII.3 norovirus strain. The MAb reacted with VLPs and denatured VP1 protein from GI, GII, GIV and GV noroviruses, and mapped to a linear epitope located in the inner shell domain. An alignment of all available VP1 sequences showed that the putative epitope (residues 52-56) is highly conserved across the genus Norovirus. This broadly cross-reactive MAb thus constitutes a valuable reagent for the diagnosis and study of these diverse viruses.


Subject(s)
Capsid/metabolism , Norovirus/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/immunology , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Capsid/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/metabolism , Chlorocebus aethiops , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Epitopes/genetics , Epitopes/immunology , Epitopes/metabolism , Genotype , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Mutagenesis , Norovirus/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Sequence Alignment , Vero Cells , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
16.
J Virol ; 87(6): 3003-17, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269802

ABSTRACT

Open reading frame 2 (ORF2) of the feline calicivirus (FCV) genome encodes a capsid precursor that is posttranslationally processed to release the mature capsid protein (VP1) and a small protein of 124 amino acids, designated the leader of the capsid (LC). To investigate the role of the LC protein in the virus life cycle, mutations and deletions were introduced into the LC coding region of an infectious FCV cDNA clone. Three cysteine residues that are conserved among all vesivirus LC sequences were found to be critical for the recovery of FCV with a characteristic cytopathic effect in feline kidney cells. A cell-rounding phenotype associated with the transient expression of wild-type and mutagenized forms of the LC correlated with the cytopathic and growth properties of the corresponding engineered viruses. The host cellular protein annexin A2 was identified as a binding partner of the LC protein, consistent with a role for the LC in mediating host cell interactions that alter the integrity of the cell and enable virus spread.


Subject(s)
Calicivirus, Feline/pathogenicity , Capsid Proteins/metabolism , Cytopathogenic Effect, Viral , Virulence Factors/metabolism , Animals , Annexin A2/metabolism , Capsid Proteins/genetics , Cats , Cell Line , Host-Pathogen Interactions , Point Mutation , Protein Binding , Protein Processing, Post-Translational , Sequence Deletion , Virulence Factors/genetics
17.
Virol J ; 9: 297, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23190937

ABSTRACT

BACKGROUND: Vesiviruses in the family Caliciviridae infect a broad range of animal hosts including mammals, birds, fish, amphibians and reptiles. The vesivirus Cro1 strains were isolated from diseased snakes in the San Diego zoo in 1978 and reported as the first caliciviruses found in reptiles. The goal of this study was to characterize the Cro1 strain 780032I that was isolated in cell culture from a rock rattlesnake (Crotalus lepidus) in the original outbreak. RESULTS: We re-amplified the original virus stock in Vero cells, and determined its full-length genome sequence. The Cro1 genome is 8296 nucleotides (nt) in length and has a typical vesivirus organization, with three open reading frames (ORF), ORF1 (5643 nt), ORF2 (2121 nt), and ORF3 (348 nt) encoding a nonstructural polyprotein, the major capsid protein precursor, and a minor structural protein, respectively. Phylogenetic analysis of the full-length genome sequence revealed that the Cro1 virus clustered most closely with the VESV species of the genus Vesivirus, but was genetically distinct (82-83% identities with closest strains). CONCLUSIONS: This is the first description of a full-length genome sequence from a reptile calicivirus (Cro1). The availability of the Cro1 genome sequence should facilitate investigation of the molecular mechanisms involved in Cro1 virus evolution and host range.


Subject(s)
Caliciviridae Infections/veterinary , Crotalus/virology , Disease Outbreaks , Genome, Viral , RNA, Viral/genetics , Sequence Analysis, DNA , Vesivirus/genetics , Animals , Animals, Zoo , Caliciviridae Infections/epidemiology , Caliciviridae Infections/virology , California , Chlorocebus aethiops , Cluster Analysis , Molecular Sequence Data , Phylogeny , Sequence Homology, Nucleic Acid , Vero Cells , Vesivirus/isolation & purification , Virus Cultivation
18.
J Virol ; 86(13): 7414-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532688

ABSTRACT

Noroviruses are major etiological agents of acute viral gastroenteritis. In 2002, a GII.4 variant (Farmington Hills cluster) spread so rapidly in the human population that it predominated worldwide and displaced previous GII.4 strains. We developed and characterized a panel of six monoclonal antibodies (MAbs) directed against the capsid protein of a Farmington Hills-like GII.4 norovirus strain that was associated with a large hospital outbreak in Maryland in 2004. The six MAbs reacted with high titers against homologous virus-like particles (VLPs) by enzyme-linked immunoassay but did not react with denatured capsid protein in immunoblots. The expression and self-assembly of newly developed genogroup I/II chimeric VLPs showed that five MAbs bound to the GII.4 protruding (P) domain of the capsid protein, while one recognized the GII.4 shell (S) domain. Cross-competition assays and mutational analyses showed evidence for at least three distinct antigenic sites in the P domain and one in the S domain. MAbs that mapped to the P domain but not the S domain were able to block the interaction of VLPs with ABH histo-blood group antigens (HBGA), suggesting that multiple antigenic sites of the P domain are involved in HBGA blocking. Further analysis showed that two MAbs mapped to regions of the capsid that had been associated with the emergence of new GII.4 variants. Taken together, our data map antibody and HBGA carbohydrate binding to proximal regions of the norovirus capsid, showing that evolutionary pressures on the norovirus capsid protein may affect both antigenic and carbohydrate recognition phenotypes.


Subject(s)
ABO Blood-Group System/metabolism , Caliciviridae Infections/virology , Capsid Proteins/metabolism , Norovirus/pathogenicity , Protein Interaction Mapping , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Caliciviridae Infections/epidemiology , Capsid Proteins/genetics , Disease Outbreaks , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Genotype , Humans , Maryland/epidemiology , Mice , Mice, Inbred BALB C , Norovirus/genetics , Norovirus/isolation & purification , Protein Binding
19.
PLoS One ; 6(6): e21435, 2011.
Article in English | MEDLINE | ID: mdl-21738664

ABSTRACT

Antibody prevalence studies in laboratory mice indicate that murine norovirus (MNV) infections are common, but the natural history of these viruses has not been fully established. This study examined the extent of genetic diversity of murine noroviruses isolated from healthy laboratory mice housed in multiple animal facilities within a single, large research institute- the National Institute of Allergy and Infectious Diseases of the National Institutes of Health (NIAID-NIH) in Bethesda, Maryland, U.S. Ten distinct murine norovirus strains were isolated from various tissues and feces of asymptomatic wild type sentinel mice as well as asymptomatic immunodeficient (RAG 2(-/-)) mice. The NIH MNV isolates showed little cytopathic effect in permissive RAW264.7 cells in early passages, but all isolates examined could be adapted to efficient growth in cell culture by serial passage. The viruses, although closely related in genome sequence, were distinguishable from each other according to facility location, likely due to the introduction of new viruses into each facility from separate sources or vendors at different times. Our study indicates that the murine noroviruses are widespread in these animal facilities, despite rigorous guidelines for animal care and maintenance.


Subject(s)
Academies and Institutes , Norovirus/classification , Norovirus/genetics , Animals , Cell Line , Fluorescent Antibody Technique , Mice , Norovirus/isolation & purification , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , United States
20.
J Virol ; 85(16): 8056-68, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21680514

ABSTRACT

Cellular proteins play many important roles during the life cycle of all viruses. Specifically, host cell nucleic acid-binding proteins interact with viral components of positive-stranded RNA viruses and regulate viral translation, as well as RNA replication. Here, we report that nucleolin, a ubiquitous multifunctional nucleolar shuttling phosphoprotein, interacts with the Norwalk virus and feline calicivirus (FCV) genomic 3' untranslated regions (UTRs). Nucleolin can also form a complex in vitro with recombinant Norwalk virus NS6 and -7 (NS6/7) and can be copurified with the analogous protein from feline calicivirus (p76 or NS6/7) from infected feline kidney cells. Nucleolin RNA levels or protein were not modified during FCV infection; however, as a consequence of the infection, nucleolin was seen to relocalize from the nucleoli to the nucleoplasm, as well as to the perinuclear area where it colocalizes with the feline calicivirus NS6/7 protein. In addition, antibodies to nucleolin were able to precipitate viral RNA from feline calicivirus-infected cells, indicating a direct or indirect association of nucleolin with the viral RNA during virus replication. Small interfering RNA (siRNA)-mediated knockdown of nucleolin resulted in a reduction of the cytopathic effect and virus yield in CrFK cells. Taken together, these results demonstrate that nucleolin is a nucleolar component that interacts with viral RNA and NS6/7 and is required for feline calicivirus replication.


Subject(s)
3' Untranslated Regions , Calicivirus, Feline/physiology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Calicivirus, Feline/genetics , Calicivirus, Feline/metabolism , Cats , Cell Line , Kidney/virology , Norwalk virus/genetics , Norwalk virus/metabolism , Peptide Hydrolases , Phosphoproteins/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , Viral Proteins/genetics , Nucleolin
SELECTION OF CITATIONS
SEARCH DETAIL
...