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










Publication year range
1.
PLoS One ; 7(3): e32857, 2012.
Article in English | MEDLINE | ID: mdl-22412934

ABSTRACT

Severe acute respiratory syndrome (SARS) is a zoonotic disease caused by SARS-related coronavirus (SARS-CoV) that emerged in 2002 to become a global health concern. Although the original outbreak was controlled by classical public health measures, there is a real risk that another SARS-CoV could re-emerge from its natural reservoir, either in its original form or as a more virulent or pathogenic strain; in which case, the virus would be difficult to control in the absence of any effective antiviral drugs or vaccines. Using the well-studied SARS-CoV isolate HKU-39849, we developed a vaccinia virus-based SARS-CoV reverse genetic system that is both robust and biosafe. The SARS-CoV genome was cloned in separate vaccinia virus vectors, (vSARS-CoV-5prime and vSARS-CoV-3prime) as two cDNAs that were subsequently ligated to create a genome-length SARS-CoV cDNA template for in vitro transcription of SARS-CoV infectious RNA transcripts. Transfection of the RNA transcripts into permissive cells led to the recovery of infectious virus (recSARS-CoV). Characterization of the plaques produced by recSARS-CoV showed that they were similar in size to the parental SARS-CoV isolate HKU-39849 but smaller than the SARS-CoV isolate Frankfurt-1. Comparative analysis of replication kinetics showed that the kinetics of recSARS-CoV replication are similar to those of SARS-CoV Frankfurt-1, although the titers of virus released into the culture supernatant are approximately 10-fold less. The reverse genetic system was finally used to generate a recSARS-CoV reporter virus expressing Renilla luciferase in order to facilitate the analysis of SARS-CoV gene expression in human dendritic cells (hDCs). In parallel, a Renilla luciferase gene was also inserted into the genome of human coronavirus 229E (HCoV-229E). Using this approach, we demonstrate that, in contrast to HCoV-229E, SARS-CoV is not able to mediate efficient heterologous gene expression in hDCs.


Subject(s)
Reassortant Viruses/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Vaccinia virus/genetics , Animals , Cell Line , Chlorocebus aethiops , Cloning, Molecular , DNA, Complementary , Dendritic Cells/virology , Gene Expression Regulation, Viral , Gene Order , Genome, Viral , Humans , Molecular Sequence Data , Recombination, Genetic , Severe acute respiratory syndrome-related coronavirus/growth & development , Sequence Analysis, DNA , Viral Plaque Assay , Virus Replication
2.
Nucleic Acids Res ; 40(4): 1737-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22039154

ABSTRACT

Uniquely among RNA viruses, replication of the ~30-kb SARS-coronavirus genome is believed to involve two RNA-dependent RNA polymerase (RdRp) activities. The first is primer-dependent and associated with the 106-kDa non-structural protein 12 (nsp12), whereas the second is catalysed by the 22-kDa nsp8. This latter enzyme is capable of de novo initiation and has been proposed to operate as a primase. Interestingly, this protein has only been crystallized together with the 10-kDa nsp7, forming a hexadecameric, dsRNA-encircling ring structure [i.e. nsp(7+8), consisting of 8 copies of both nsps]. To better understand the implications of these structural characteristics for nsp8-driven RNA synthesis, we studied the prerequisites for the formation of the nsp(7+8) complex and its polymerase activity. We found that in particular the exposure of nsp8's natural N-terminal residue was paramount for both the protein's ability to associate with nsp7 and for boosting its RdRp activity. Moreover, this 'improved' recombinant nsp8 was capable of extending primed RNA templates, a property that had gone unnoticed thus far. The latter activity is, however, ~20-fold weaker than that of the primer-dependent nsp12-RdRp at equal monomer concentrations. Finally, site-directed mutagenesis of conserved D/ExD/E motifs was employed to identify residues crucial for nsp(7+8) RdRp activity.


Subject(s)
RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cations, Divalent/chemistry , DNA-Directed RNA Polymerases/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Multimerization , Protons , RNA/metabolism , RNA, Double-Stranded/metabolism , RNA-Dependent RNA Polymerase/genetics , Sequence Homology, Amino Acid , Viral Proteins/genetics
3.
J Virol ; 85(11): 5669-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21430047

ABSTRACT

The RNA replication and transcription complex of coronaviruses is associated with an elaborate reticulovesicular network (RVN) of modified endoplasmic reticulum. Using cycloheximide and puromycin, we have studied the effect of translation inhibition on the RNA synthesis of severe acute respiratory syndrome coronavirus and mouse hepatitis virus. Both inhibitors prevented the usual exponential increase in viral RNA synthesis, with immunofluorescence and electron microscopy indicating that RVN development came to a standstill. Nevertheless, limited RNA synthesis was supported, implying that continued translation is not an absolute requirement and suggesting a direct link between RVN formation and accumulation of coronavirus proteins.


Subject(s)
Murine hepatitis virus/physiology , RNA, Viral/biosynthesis , Severe acute respiratory syndrome-related coronavirus/physiology , Transcription, Genetic , Virus Replication , Animals , Chlorocebus aethiops , Cycloheximide/metabolism , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/metabolism , Puromycin/metabolism , Vero Cells , Viral Proteins/metabolism
4.
PLoS Pathog ; 6(11): e1001176, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-21079686

ABSTRACT

Increasing the intracellular Zn(2+) concentration with zinc-ionophores like pyrithione (PT) can efficiently impair the replication of a variety of RNA viruses, including poliovirus and influenza virus. For some viruses this effect has been attributed to interference with viral polyprotein processing. In this study we demonstrate that the combination of Zn(2+) and PT at low concentrations (2 µM Zn(2+) and 2 µM PT) inhibits the replication of SARS-coronavirus (SARS-CoV) and equine arteritis virus (EAV) in cell culture. The RNA synthesis of these two distantly related nidoviruses is catalyzed by an RNA-dependent RNA polymerase (RdRp), which is the core enzyme of their multiprotein replication and transcription complex (RTC). Using an activity assay for RTCs isolated from cells infected with SARS-CoV or EAV--thus eliminating the need for PT to transport Zn(2+) across the plasma membrane--we show that Zn(2+) efficiently inhibits the RNA-synthesizing activity of the RTCs of both viruses. Enzymatic studies using recombinant RdRps (SARS-CoV nsp12 and EAV nsp9) purified from E. coli subsequently revealed that Zn(2+) directly inhibited the in vitro activity of both nidovirus polymerases. More specifically, Zn(2+) was found to block the initiation step of EAV RNA synthesis, whereas in the case of the SARS-CoV RdRp elongation was inhibited and template binding reduced. By chelating Zn(2+) with MgEDTA, the inhibitory effect of the divalent cation could be reversed, which provides a novel experimental tool for in vitro studies of the molecular details of nidovirus replication and transcription.


Subject(s)
Arterivirus/enzymology , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Severe acute respiratory syndrome-related coronavirus/enzymology , Virus Replication/drug effects , Zinc Compounds/pharmacology , Animals , Arterivirus/drug effects , Arterivirus Infections/drug therapy , Arterivirus Infections/pathology , Arterivirus Infections/virology , Blotting, Western , Chlorocebus aethiops , Electrophoretic Mobility Shift Assay , Escherichia coli/enzymology , Escherichia coli/genetics , In Vitro Techniques , Ionophores/pharmacology , RNA, Messenger/genetics , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe Acute Respiratory Syndrome/drug therapy , Severe Acute Respiratory Syndrome/pathology , Severe Acute Respiratory Syndrome/virology , Vero Cells
5.
J Virol ; 84(2): 833-46, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19889777

ABSTRACT

To accommodate its RNA synthesis in the infected cell, severe acute respiratory syndrome coronavirus (SARS-CoV) induces a cytoplasmic reticulovesicular network (RVN) that is derived from endoplasmic reticulum (ER) membranes. We set out to investigate how the early secretory pathway interacts with the RVN and the viral replication/transcription complex (RTC) that is anchored to it. When the secretory pathway was disrupted by brefeldin A (BFA) treatment at the start of infection, RVN formation and viral RTC activity were not blocked and continued up to 11 h postinfection, although RNA synthesis was reduced by ca. 80%. In vitro RTC assays, using membrane fractions from infected cells, demonstrated that BFA does not directly interfere with the activity of the viral RNA-synthesizing enzymes. Confocal microscopy studies showed that early secretory pathway components are not associated with SARS-CoV-induced replication sites, although our studies revealed that infection induces a remarkable redistribution of the translocon subunit Sec61alpha. Ultrastructural studies, including electron tomography, revealed that the formation of the RVN and all its previously documented features can occur in the presence of BFA, despite differences in the volume and morphology of the network. We therefore conclude that early secretory pathway proteins do not play a direct role in RVN morphogenesis or the functionality of the SARS-CoV RTC. The BFA-induced disruption of ER integrity and functionality probably affects the overall quality of the membrane scaffold that is needed to support the viral RTC and/or the availability of specific host factors, which in turn compromises viral RNA synthesis.


Subject(s)
Endoplasmic Reticulum , Host-Pathogen Interactions , Intracellular Membranes , Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Animals , Brefeldin A/pharmacology , Chlorocebus aethiops , Electron Microscope Tomography , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Intracellular Membranes/virology , Microscopy, Confocal , Proteins/drug effects , Proteins/genetics , RNA, Viral/metabolism , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/metabolism , Vero Cells/ultrastructure , Vero Cells/virology , Viral Proteins/metabolism , Virus Replication
6.
Nucleic Acids Res ; 38(1): 203-14, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19875418

ABSTRACT

An RNA-dependent RNA polymerase (RdRp) is the central catalytic subunit of the RNA-synthesizing machinery of all positive-strand RNA viruses. Usually, RdRp domains are readily identifiable by comparative sequence analysis, but biochemical confirmation and characterization can be hampered by intrinsic protein properties and technical complications. It is presumed that replication and transcription of the approximately 30-kb severe acute respiratory syndrome (SARS) coronavirus (SARS-CoV) RNA genome are catalyzed by an RdRp domain in the C-terminal part of nonstructural protein 12 (nsp12), one of 16 replicase subunits. However, thus far full-length nsp12 has proven refractory to expression in bacterial systems, which has hindered both the biochemical characterization of coronavirus RNA synthesis and RdRp-targeted antiviral drug design. Here, we describe a combined strategy involving bacterial expression of an nsp12 fusion protein and its in vivo cleavage to generate and purify stable SARS-CoV nsp12 (106 kDa) with a natural N-terminus and C-terminal hexahistidine tag. This recombinant protein possesses robust in vitro RdRp activity, as well as a significant DNA-dependent activity that may facilitate future inhibitor studies. The SARS-CoV nsp12 is primer dependent on both homo- and heteropolymeric templates, supporting the likeliness of a close enzymatic collaboration with the intriguing RNA primase activity that was recently proposed for coronavirus nsp8.


Subject(s)
RNA-Dependent RNA Polymerase/metabolism , RNA/metabolism , Severe acute respiratory syndrome-related coronavirus/enzymology , Viral Proteins/metabolism , Nucleotides/metabolism , RNA/biosynthesis , RNA-Dependent RNA Polymerase/isolation & purification , Templates, Genetic , Viral Proteins/isolation & purification
7.
PLoS Pathog ; 4(5): e1000054, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18451981

ABSTRACT

SARS-coronavirus (SARS-CoV) replication and transcription are mediated by a replication/transcription complex (RTC) of which virus-encoded, non-structural proteins (nsps) are the primary constituents. The 16 SARS-CoV nsps are produced by autoprocessing of two large precursor polyproteins. The RTC is believed to be associated with characteristic virus-induced double-membrane structures in the cytoplasm of SARS-CoV-infected cells. To investigate the link between these structures and viral RNA synthesis, and to dissect RTC organization and function, we isolated active RTCs from infected cells and used them to develop the first robust assay for their in vitro activity. The synthesis of genomic RNA and all eight subgenomic mRNAs was faithfully reproduced by the RTC in this in vitro system. Mainly positive-strand RNAs were synthesized and protein synthesis was not required for RTC activity in vitro. All RTC activity, enzymatic and putative membrane-spanning nsps, and viral RNA cosedimented with heavy membrane structures. Furthermore, the pelleted RTC required the addition of a cytoplasmic host factor for reconstitution of its in vitro activity. Newly synthesized subgenomic RNA appeared to be released, while genomic RNA remained predominantly associated with the RTC-containing fraction. RTC activity was destroyed by detergent treatment, suggesting an important role for membranes. The RTC appeared to be protected by membranes, as newly synthesized viral RNA and several replicase/transcriptase subunits were protease- and nuclease-resistant and became susceptible to degradation only upon addition of a non-ionic detergent. Our data establish a vital functional dependence of SARS-CoV RNA synthesis on virus-induced membrane structures.


Subject(s)
Host-Pathogen Interactions , Severe acute respiratory syndrome-related coronavirus/physiology , Transcription, Genetic/genetics , Virus Replication/physiology , Animals , Chlorocebus aethiops , Cytoplasm/metabolism , Dactinomycin/pharmacology , Gene Expression Regulation, Viral , Genome, Viral , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA, Viral/biosynthesis , Rabbits , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/ultrastructure , Transcription, Genetic/drug effects , Vero Cells , Viral Matrix Proteins/metabolism
8.
J Virol ; 81(12): 6771-4, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17428856

ABSTRACT

A 190-nucleotide (nt) packaging signal (PS) located in the 3' end of open reading frame 1b in the mouse hepatitis virus, a group IIa coronavirus, was previously postulated to direct genome RNA packaging. Based on phylogenetic data and structure probing, we have identified a 95-nt hairpin within the 190-nt PS domain which is conserved in all group IIa coronaviruses but not in the severe acute respiratory syndrome coronavirus (group IIb), group I coronaviruses, or group III coronaviruses. The hairpin is composed of six copies of a repeating structural subunit that consists of 2-nt bulges and 5-bp stems. We propose that repeating AA bulges are characteristic features of group IIa PSs.


Subject(s)
Coronavirus/genetics , Genome, Viral , Base Sequence , Databases, Protein , Molecular Sequence Data , Nucleic Acid Conformation , Open Reading Frames , Protein Structure, Secondary , Protein Structure, Tertiary , RNA Viruses/genetics , Severe acute respiratory syndrome-related coronavirus/genetics , Sequence Analysis, DNA , Species Specificity
9.
Virology ; 361(1): 18-26, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17316733

ABSTRACT

Many viruses encode antagonists to prevent interferon (IFN) induction. Infection of fibroblasts with the murine hepatitis coronavirus (MHV) and SARS-coronavirus (SARS-CoV) did not result in nuclear translocation of interferon-regulatory factor 3 (IRF3), a key transcription factor involved in IFN induction, and induction of IFN mRNA transcription. Furthermore, MHV and SARS-CoV infection could not prevent IFN induction by poly (I:C) or Sendai virus, suggesting that these CoVs do not inactivate IRF3-mediated transcription regulation, but apparently prevent detection of replicative RNA by cellular sensory molecules. Our data indicate that shielding of viral RNA to host cell sensors might be the main general mechanism for coronaviruses to prevent IFN induction.


Subject(s)
Interferon-alpha/metabolism , Severe Acute Respiratory Syndrome/immunology , Severe acute respiratory syndrome-related coronavirus/immunology , Animals , Biological Transport , Chlorocebus aethiops , Interferon Regulatory Factor-3/metabolism , L Cells , Mice , Murine hepatitis virus/immunology , RNA, Viral/physiology , Severe acute respiratory syndrome-related coronavirus/genetics , Sendai virus/immunology , Severe Acute Respiratory Syndrome/virology , Vero Cells
10.
J Mol Biol ; 363(4): 858-65, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16989861

ABSTRACT

We solved the structures of the single-stranded RNA bacteriophages Qbeta, PP7 and AP205 by cryo-electron microscopy. On the outside, the symmetrized electron density maps resemble the previously described cryo-electron microscopy structure of MS2. RNA density is present inside the capsids, suggesting that the genomic RNA of Qbeta, PP7 and AP205, analogous to MS2, contains many coat protein-binding sites in addition to the hairpin on which assembly and packaging are initiated. All four bacteriophages harbour the same overall arrangement of the RNA, which is a unique combination of both triangles and pentagons. This combination has not been found in other icosahedral viruses, in which the RNA structures are either triangular or pentagonal. Strikingly, the unique RNA packing of the Leviviridae appears to deploy the most efficient method of RNA storage by obeying icosahedral symmetry.


Subject(s)
Cryoelectron Microscopy , Leviviridae/genetics , Leviviridae/ultrastructure , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/ultrastructure , Amino Acid Sequence , Genome, Viral/genetics , Molecular Sequence Data , Sequence Alignment , Viral Proteins/chemistry , Viral Proteins/genetics , Virion/genetics , Virion/ultrastructure
11.
Cell ; 112(1): 123-9, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12526799

ABSTRACT

Turnip yellow mosaic virus (TYMV) has a genomic plus-strand RNA with a 5' cap followed by overlapping and different reading frames for the movement protein and polyprotein, while the distal coat protein cistron is translated from a subgenomic RNA. The 3'-untranslated region harbors a tRNA-like structure (TLS) to which a valine moiety can be added and it is indispensable for virus viability. Here, we report about a surprising interaction between TYMV-RNA-programmed ribosomes and 3'-valylated TLS that yields polyprotein with the valine N terminally incorporated by a translation mechanism resistant to regular initiation inhibitors. Disruption of the TLS exclusively abolishes polyprotein synthesis, which can be restored by adding excess TLS in trans. Our observations imply a novel eukaryotic mechanism for internal initiation of mRNA translation.


Subject(s)
Molecular Mimicry , Mosaic Viruses/genetics , Protein Biosynthesis , RNA, Transfer/genetics , RNA, Viral , Ribosomes/metabolism , 3' Untranslated Regions/chemistry , 3' Untranslated Regions/genetics , 3' Untranslated Regions/metabolism , Models, Genetic , Mosaic Viruses/metabolism , Protein Structure, Secondary , RNA, Messenger/metabolism , Ribosomes/drug effects , Substrate Specificity , Triticum , Tymovirus/genetics , Valine/chemistry , Valine/metabolism , Viral Proteins/biosynthesis , Viral Proteins/chemistry , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...