Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 87(19): 10423-34, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903828

ABSTRACT

To successfully complete their replication cycles, picornaviruses modify several host proteins to alter the cellular environment to favor virus production. One such target of viral proteinase cleavage is AU-rich binding factor 1 (AUF1), a cellular protein that binds to AU-rich elements, or AREs, in the 3' noncoding regions (NCRs) of mRNAs to affect the stability of the RNA. Previous studies found that, during poliovirus or human rhinovirus infection, AUF1 is cleaved by the viral proteinase 3CD and that AUF1 can interact with the long 5' NCR of these viruses in vitro. Here, we expand on these initial findings to demonstrate that all four isoforms of AUF1 bind directly to stem-loop IV of the poliovirus 5' NCR, an interaction that is inhibited through proteolytic cleavage of AUF1 by the viral proteinase 3CD. Endogenous AUF1 was observed to relocalize to the cytoplasm of infected cells in a viral protein 2A-driven manner and to partially colocalize with the viral protein 3CD. We identify a negative role for AUF1 in poliovirus infection, as AUF1 inhibited viral translation and, ultimately, overall viral titers. Our findings also demonstrate that AUF1 functions as an antiviral factor during infection by coxsackievirus or human rhinovirus, suggesting a common mechanism that targets these related picornaviruses.


Subject(s)
Coxsackievirus Infections/virology , Enterovirus/pathogenicity , Heterogeneous-Nuclear Ribonucleoprotein D/physiology , Picornaviridae Infections/virology , RNA Stability , Rhinovirus/pathogenicity , 3C Viral Proteases , Animals , Cells, Cultured , Coxsackievirus Infections/genetics , Coxsackievirus Infections/metabolism , Cysteine Endopeptidases/metabolism , Cytoplasm/metabolism , Cytoplasm/virology , Electrophoretic Mobility Shift Assay , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/virology , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Fluorescent Antibody Technique , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Humans , Mice , Mice, Knockout , Picornaviridae Infections/genetics , Picornaviridae Infections/metabolism , Poliovirus/genetics , Protein Biosynthesis , Protein Isoforms , RNA, Untranslated/genetics , RNA, Viral/genetics , Rabbits , Viral Proteins/immunology , Viral Proteins/metabolism
2.
mBio ; 3(6): e00431-12, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23131833

ABSTRACT

UNLABELLED: Due to the limited coding capacity of picornavirus genomic RNAs, host RNA binding proteins play essential roles during viral translation and RNA replication. Here we describe experiments suggesting that AUF1, a host RNA binding protein involved in mRNA decay, plays a role in the infectious cycle of picornaviruses such as poliovirus and human rhinovirus. We observed cleavage of AUF1 during poliovirus or human rhinovirus infection, as well as interaction of this protein with the 5' noncoding regions of these viral genomes. Additionally, the picornavirus proteinase 3CD, encoded by poliovirus or human rhinovirus genomic RNAs, was shown to cleave all four isoforms of recombinant AUF1 at a specific N-terminal site in vitro. Finally, endogenous AUF1 was found to relocalize from the nucleus to the cytoplasm in poliovirus-infected HeLa cells to sites adjacent to (but distinct from) putative viral RNA replication complexes. IMPORTANCE: This study derives its significance from reporting how picornaviruses like poliovirus and human rhinovirus proteolytically cleave a key player (AUF1) in host mRNA decay pathways during viral infection. Beyond cleavage of AUF1 by the major viral proteinase encoded in picornavirus genomes, infection by poliovirus results in the relocalization of this host cell RNA binding protein from the nucleus to the cytoplasm. The alteration of both the physical state of AUF1 and its cellular location illuminates how small RNA viruses manipulate the activities of host cell RNA binding proteins to ensure a faithful intracellular replication cycle.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Host-Pathogen Interactions , Poliovirus/physiology , RNA, Viral/metabolism , Rhinovirus/physiology , Virus Replication , 3C Viral Proteases , Cysteine Endopeptidases/metabolism , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Humans , Proteolysis , Viral Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 109(36): 14634-9, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22908287

ABSTRACT

A previously described mammalian cell activity, called VPg unlinkase, specifically cleaves a unique protein-RNA covalent linkage generated during the viral genomic RNA replication steps of a picornavirus infection. For over three decades, the identity of this cellular activity and its normal role in the uninfected cell had remained elusive. Here we report the purification and identification of VPg unlinkase as the DNA repair enzyme, 5'-tyrosyl-DNA phosphodiesterase-2 (TDP2). Our data show that VPg unlinkase activity in different mammalian cell lines correlates with their differential expression of TDP2. Furthermore, we show that recombinant TDP2 can cleave the protein-RNA linkage generated by different picornaviruses without impairing the integrity of viral RNA. Our results reveal a unique RNA repair-like function for TDP2 and suggest an unusual role in host-pathogen interactions for this cellular enzyme. On the basis of the identification of TDP2 as a potential antiviral target, our findings may lead to the development of universal therapeutics to treat the millions of individuals afflicted annually with diseases caused by picornaviruses, including myocarditis, aseptic meningitis, encephalitis, hepatitis, and the common cold.


Subject(s)
Nuclear Proteins/metabolism , Picornaviridae/metabolism , Ribonucleoproteins/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , DNA-Binding Proteins , Electrophoresis, Polyacrylamide Gel , Evolution, Molecular , Fluorescent Antibody Technique , HeLa Cells , Host-Pathogen Interactions , Humans , Microscopy, Confocal , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases , Picornaviridae/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics
4.
PLoS Pathog ; 7(12): e1002433, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22174690

ABSTRACT

SARS coronavirus (SCoV) nonstructural protein (nsp) 1, a potent inhibitor of host gene expression, possesses a unique mode of action: it binds to 40S ribosomes to inactivate their translation functions and induces host mRNA degradation. Our previous study demonstrated that nsp1 induces RNA modification near the 5'-end of a reporter mRNA having a short 5' untranslated region and RNA cleavage in the encephalomyocarditis virus internal ribosome entry site (IRES) region of a dicistronic RNA template, but not in those IRES elements from hepatitis C or cricket paralysis viruses. By using primarily cell-free, in vitro translation systems, the present study revealed that the nsp1 induced endonucleolytic RNA cleavage mainly near the 5' untranslated region of capped mRNA templates. Experiments using dicistronic mRNAs carrying different IRESes showed that nsp1 induced endonucleolytic RNA cleavage within the ribosome loading region of type I and type II picornavirus IRES elements, but not that of classical swine fever virus IRES, which is characterized as a hepatitis C virus-like IRES. The nsp1-induced RNA cleavage of template mRNAs exhibited no apparent preference for a specific nucleotide sequence at the RNA cleavage sites. Remarkably, SCoV mRNAs, which have a 5' cap structure and 3' poly A tail like those of typical host mRNAs, were not susceptible to nsp1-mediated RNA cleavage and importantly, the presence of the 5'-end leader sequence protected the SCoV mRNAs from nsp1-induced endonucleolytic RNA cleavage. The escape of viral mRNAs from nsp1-induced RNA cleavage may be an important strategy by which the virus circumvents the action of nsp1 leading to the efficient accumulation of viral mRNAs and viral proteins during infection.


Subject(s)
Gene Expression Regulation, Viral/genetics , RNA Stability , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Nonstructural Proteins/metabolism , RNA, Messenger/genetics , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Reverse Transcriptase Polymerase Chain Reaction , Severe acute respiratory syndrome-related coronavirus/genetics , Templates, Genetic , Viral Nonstructural Proteins/genetics
5.
PLoS One ; 6(3): e16559, 2011 Mar 07.
Article in English | MEDLINE | ID: mdl-21408223

ABSTRACT

Using poliovirus, the prototypic member of Picornaviridae, we have further characterized a host cell enzymatic activity found in uninfected cells, termed "unlinkase," that recognizes and cleaves the unique 5' tyrosyl-RNA phosphodiester bond found at the 5' end of picornavirus virion RNAs. This bond connects VPg, a viral-encoded protein primer essential for RNA replication, to the viral RNA; it is cleaved from virion RNA prior to its engaging in protein synthesis as mRNA. Due to VPg retention on nascent RNA strands and replication templates, but not on viral mRNA, we hypothesize that picornaviruses utilize unlinkase activity as a means of controlling the ratio of viral RNAs that are translated versus those that either serve as RNA replication templates or are encapsidated. To test our hypothesis and further characterize this enzyme, we have developed a novel assay to detect unlinkase activity. We demonstrate that unlinkase activity can be detected using this assay, that this unique activity remains unchanged over the course of a poliovirus infection in HeLa cells, and that unlinkase activity is unaffected by the presence of exogenous VPg or anti-VPg antibodies. Furthermore, we have determined that unlinkase recognizes and cleaves a human rhinovirus-poliovirus chimeric substrate with the same efficiency as the poliovirus substrate.


Subject(s)
Genetic Engineering , Phosphoric Diester Hydrolases/metabolism , Poliovirus/genetics , Poliovirus/metabolism , RNA, Viral/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Cell Extracts , Enzyme Assays , Genome, Viral/genetics , HeLa Cells , Humans , Isotope Labeling , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Rhinovirus/metabolism , Ribonuclease T1/metabolism , Substrate Specificity , Viral Proteins/chemistry , Viral Proteins/metabolism , Virion/metabolism
6.
J Virol ; 85(1): 638-43, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21047955

ABSTRACT

The nsp1 protein of transmissible gastroenteritis virus (TGEV), an alphacoronavirus, efficiently suppressed protein synthesis in mammalian cells. Unlike the nsp1 protein of severe acute respiratory syndrome coronavirus, a betacoronavirus, the TGEV nsp1 protein was unable to bind 40S ribosomal subunits or promote host mRNA degradation. TGEV nsp1 also suppressed protein translation in cell-free HeLa cell extract; however, it did not affect translation in rabbit reticulocyte lysate (RRL). Our data suggested that HeLa cell extracts and cultured host cells, but not RRL, contain a host factor(s) that is essential for TGEV nsp1-induced translational suppression.


Subject(s)
Gene Expression Regulation , Protein Biosynthesis/drug effects , Transmissible gastroenteritis virus/pathogenicity , Viral Nonstructural Proteins/pharmacology , Animals , Cell Extracts , Cell Line , HeLa Cells/virology , Humans , Kidney/cytology , Kidney/virology , Male , Rabbits , Reticulocytes/virology , Swine , Testis/cytology , Testis/virology , Viral Nonstructural Proteins/metabolism
7.
Virology ; 400(2): 240-7, 2010 May 10.
Article in English | MEDLINE | ID: mdl-20189623

ABSTRACT

The hnRNP C heterotetramer [(C1(3))C2] binds RNA polymerase II transcripts in the nucleus, along with other proteins of the core hnRNP complex, and plays an important role in mRNA biogenesis and transport. Infection of HeLa cells with poliovirus causes hnRNP C to re-localize from the nucleus, where it is normally retained during interphase, to the cytoplasm. We have proposed that in the cytoplasm, the protein isoforms of hnRNP C participate in the recognition of viral specific RNAs by the poliovirus replication proteins and/or in the assembly of membrane-bound RNA replication complexes. In SK-OV-3 cells, which express reduced levels of hnRNP C compared to HeLa cells or 293 cells, the kinetics of poliovirus replication are delayed. hnRNP C is also re-localized from the nucleus to the cytoplasm in SK-OV-3 cells infected with poliovirus. Increased expression of hnRNP C in SK-OV-3 cells by transient transfection increases the rate of virus production and overall yield over that seen in mock-transfected cells. We propose that hnRNP C interacts with poliovirus RNA and replication proteins to increase the efficiency of viral genomic RNA synthesis.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Host-Pathogen Interactions , Poliovirus/physiology , RNA, Viral/biosynthesis , Virus Replication , Cell Line, Tumor , Cell Nucleus/chemistry , Cytoplasm/chemistry , Gene Expression , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Humans , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...