Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Ned Tijdschr Geneeskd ; 1622018 09 06.
Article in Dutch | MEDLINE | ID: mdl-30306764

ABSTRACT

The 2017/2018 influenza season was severe and lasted twice as long as usual. Hospitals struggled to meet the demand for care. In addition to a high number of patients with flu and its complications, other factors played a role. These included absenteeism of informal caretakers and professional home care staff due to having flu themselves, and added strain on hospital capacity due to flu-related sick leave of hospital staff. A minority of the latter group is vaccinated annually against influenza. The authors of this article argue that all healthcare providers should take the yearly influenza vaccination. This will prove beneficial to the employer and employees, since non-attendance among employees will be reduced during peak demand and thus ensure continuity of care capacity. It will also have a positive impact in terms of patient safety and professionalism through improved protection of vulnerable patients against nosocomial influenza infection.


Subject(s)
Influenza Vaccines , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Vaccination , Absenteeism , Caregivers , Cross Infection/prevention & control , Female , Hospitals/statistics & numerical data , Humans , Netherlands/epidemiology , Personnel, Hospital , Seasons , Sick Leave , Vaccination/statistics & numerical data
2.
Arch Virol ; 158(1): 251-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23053517

ABSTRACT

Novel viruses might be responsible for numerous disease cases with unknown etiology. In this study, we screened 1800 nasopharyngeal samples from adult outpatients with respiratory disease symptoms and healthy individuals. We employed a reverse transcription (RT)-PCR assay and CODEHOP-based primers (CT12-mCODEHOP) previously developed to recognize known and unknown corona- and toroviruses. The CT12-mCODEHOP assay detected 42.0 % (29/69) of samples positive for human coronaviruses (HCoV), including HCoV-229 (1/16), HCoV-NL63 (9/17), and HCoV-OC43 (19/36), and additionally HCoV-HKU1 (3), which was not targeted by the diagnostic real-time PCR assays. No other coronaviruses were identified in the analyzed samples.


Subject(s)
Coronavirus/isolation & purification , DNA Primers/genetics , Nasopharynx/virology , Respiratory Tract Infections/virology , Reverse Transcriptase Polymerase Chain Reaction/methods , Coronavirus/classification , Coronavirus/genetics , Humans , Respiratory Tract Infections/diagnosis
3.
J Gen Virol ; 93(Pt 9): 1918-1923, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22673933

ABSTRACT

Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is the sole known receptor of murine hepatitis virus (MHV) A59, but the available, often qualitative, data about CEACAM1 expression does not explain MHV organ tropism. Ceacam1 transcripts undergo alternative splicing resulting in multiple isoforms, including secreted CEACAM1 isoforms that can neutralize the virus. We determined the quantities of Ceacam1 transcripts encoding membrane-bound and secreted isoforms in mouse organs and a set of cell lines. In vivo, the lowest receptor mRNA levels were found in brain and muscle and these were similar to those in easily infectable cultured cells. While the quantities of the receptor transcripts varied between mouse organs, their abundance did not correlate with susceptibility to MHV infection. The proportion of transcripts encoding secreted isoforms also could not explain the selection of sites for virus replication, as it was constant in all organs. Our data suggest that neither of the two CEACAM1 isoforms defines MHV organ tropism.


Subject(s)
Carcinoembryonic Antigen/genetics , Cell Membrane/genetics , Coronavirus Infections/veterinary , Murine hepatitis virus/physiology , Muscles/metabolism , Receptors, Virus/genetics , Rodent Diseases/genetics , Viral Tropism , Animals , Brain , Carcinoembryonic Antigen/metabolism , Cell Membrane/metabolism , Coronavirus Infections/genetics , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Murine hepatitis virus/genetics , Muscles/virology , Organ Specificity , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Receptors, Virus/metabolism , Rodent Diseases/metabolism , Rodent Diseases/virology
4.
J Virol Methods ; 177(2): 174-83, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21864579

ABSTRACT

The ssRNA+ family Coronaviridae includes two subfamilies prototyped by coronaviruses and toroviruses that cause respiratory and enteric infections. To facilitate the identification of new distantly related members of the family Coronaviridae, we have developed a molecular assay with broad specificity. The consensus-degenerated hybrid oligonucleotide primer (CODEHOP) strategy was modified to design primers targeting the most conserved motifs in the RNA-dependent RNA polymerase locus. They were evaluated initially on RNA templates from virus-infected cells using a two-step RT-PCR protocol that was further advanced to a one-step assay. The sensitivity of the assay ranged from 10(2) to 10(6) and from 10(5) to 10(9) RNA copy numbers for individual corona-/torovirus templates when tested, respectively, with and without an excess of RNA from human cells. This primer set compared to that designed according to the original CODEHOP rules showed 10-10(3) folds greater sensitivity for 5 of the 6 evaluated corona-/torovirus templates. It detected 57% (32 of 56) of the respiratory specimens positive for 4 human coronaviruses, as well as stool specimens positive for a bovine torovirus. The high sensitivity and broad virus range of this assay makes it suitable for screening biological specimens in search for new viruses of the family Coronaviridae.


Subject(s)
Coronavirus/isolation & purification , DNA Primers/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Torovirus/isolation & purification , Amino Acid Sequence , Animals , Cattle , Cell Line , Conserved Sequence , Coronavirus/classification , Coronavirus/genetics , Coronavirus Infections/diagnosis , Coronavirus Infections/virology , DNA Primers/metabolism , Humans , Molecular Sequence Data , Phylogeny , RNA, Viral/genetics , Sensitivity and Specificity , Sequence Alignment , Torovirus/classification , Torovirus/genetics , Torovirus Infections/diagnosis , Torovirus Infections/virology , Virus Cultivation
5.
J Clin Virol ; 51(1): 25-30, 2011 May.
Article in English | MEDLINE | ID: mdl-21388868

ABSTRACT

BACKGROUND: Persistent infections with herpesviruses such as human cytomegalovirus (HCMV) frequently occur after solid organ or stem cell transplantation, and are due to either failure of the host to immunologically control the virus or emerging resistance of the virus to the antiviral drug(s) used. Antiviral therapy can be guided by viral drug susceptibility testing based on screening for known resistance-inducing mutations in the viral genome. Mass spectrometry-based comparative sequence analysis (MSCSA) might be advantageous for this purpose because of its suitability for semi-automation. OBJECTIVES: The applicability of MSCSA to detect sequence polymorphisms and drug resistance-inducing mutations in the HCMV genome was investigated. STUDY DESIGN: We analyzed the 3' part of the HCMV UL97 gene, which encodes the kinase that is activated by the commonly used anti-HCMV drug ganciclovir. Sequences obtained by MSCSA of material from HCMV-infected patients (43 samples) and the HCMV type strain were compared to conventional cycle sequencing results. RESULTS: In 94.1% of all samples the results obtained by MSCSA of the UL97 gene were identical to those from conventional cycle sequencing. The threshold to detect mutant sequences in a mixture with wild-type material was 20% using either technique. Furthermore, MSCSA was successfully applied to study the development of drug resistance in a patient who developed encephalitis due to ganciclovir-resistant HCMV. CONCLUSIONS: MSCSA was found to be equally accurate compared to conventional cycle sequencing in the analysis of UL97 of HCMV.


Subject(s)
Antiviral Agents/pharmacology , Cytomegalovirus Infections/virology , Cytomegalovirus/drug effects , Ganciclovir/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Antiviral Agents/therapeutic use , Base Sequence , Cytomegalovirus/genetics , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/drug therapy , DNA Mutational Analysis/methods , DNA, Viral/analysis , DNA, Viral/genetics , Drug Resistance, Viral/genetics , Female , Ganciclovir/therapeutic use , Genotype , Humans , Middle Aged , Molecular Sequence Data , Molecular Typing , Mutation , Polymorphism, Single Nucleotide , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
6.
J Virol ; 84(21): 11395-406, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739539

ABSTRACT

Cells and mice infected with arthropod-borne flaviviruses produce a small subgenomic RNA that is colinear with the distal part of the viral 3'-untranslated region (UTR). This small subgenomic flavivirus RNA (sfRNA) results from the incomplete degradation of the viral genome by the host 5'-3' exonuclease XRN1. Production of the sfRNA is important for the pathogenicity of the virus. This study not only presents a detailed description of the yellow fever virus (YFV) sfRNA but, more importantly, describes for the first time the molecular characteristics of the stalling site for XRN1 in the flavivirus genome. Similar to the case for West Nile virus, the YFV sfRNA was produced by XRN1. However, in contrast to the case for other arthropod-borne flaviviruses, not one but two sfRNAs were detected in YFV-infected mammalian cells. The smaller of these two sfRNAs was not observed in infected mosquito cells. The larger sfRNA could also be produced in vitro by incubation with purified XRN1. These two YFV sfRNAs formed a 5'-nested set. The 5' ends of the YFV sfRNAs were found to be just upstream of the previously predicted RNA pseudoknot PSK3. RNA structure probing and mutagenesis studies provided strong evidence that this pseudoknot structure was formed and served as the molecular signal to stall XRN1. The sequence involved in PSK3 formation was cloned into the Sinrep5 expression vector and shown to direct the production of an sfRNA-like RNA. These results underscore the importance of the RNA pseudoknot in stalling XRN1 and also demonstrate that it is the sole viral requirement for sfRNA production.


Subject(s)
DNA-Binding Proteins/genetics , Exoribonucleases/genetics , RNA, Viral/biosynthesis , Yellow fever virus/genetics , Animals , Culicidae , Genome, Viral , Humans , Mice , Molecular Probes , Mutagenesis, Site-Directed , Nucleic Acid Conformation , Yellow fever virus/pathogenicity
7.
BMC Cell Biol ; 11: 33, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20470363

ABSTRACT

BACKGROUND: Glycosyl transferases transfer glycosyl groups onto their substrate. Localization partially defines their function. Glycosyl transferase 25 domain 1 (GLT25D1) was recently shown to have galactosyltransferase activity towards collagens and another well known substrate, mannose binding lectin (MBL). To gain more insight in the role of galactosylation of lysines in the Gly-X-Lys repeats of collagenous proteins, we investigated the subcellular localization of GLT25D1. RESULTS: Immunofluorescence analysis of GLT25D1 expressed in the human hepatoma cell line (Huh7), revealed a perinuclear lattice like staining, resembling localization to the endoplasmic reticulum (ER). Possible targeting signals, an N-terminal signal sequence and a C-terminal ER-retention signal, were identified using prediction programs. These signals were then investigated by constructing a series of epitope-tagged forms of GLT25D1 that were analyzed by immunofluorescence and western blotting. In agreement with the predictions our results show that GLT25D1 is directed to the ER lumen as a soluble protein and retained there. Moreover, using two endoglycosidase enzymes EndoH and EndoF, we demonstrate that the putative bi-functional glycosyl transferase itself is a glycoprotein. Additionally we examined co-localization of GLT25D1 with MBL and lysyl hydroxylase 3 (LH3, PLOD3), which is a protein able to catalyze hydroxylation of lysine residues before they can be glycosylated. We demonstrate overlapping localization patterns of GLT25D1, MBL and LH3. CONCLUSIONS: Taken together our data indicate that galactosylation of collagenous proteins by the soluble GLT25D1 occurs in the early secretory pathway.


Subject(s)
Endoplasmic Reticulum/chemistry , Galactosyltransferases/analysis , Cell Line, Tumor , Collagen/metabolism , Endoplasmic Reticulum/metabolism , Galactosyltransferases/metabolism , Glycosylation , Humans , Mannose-Binding Lectin/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism
8.
J Gen Virol ; 91(Pt 4): 1013-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19923258

ABSTRACT

Post-translational modifications (PTMs) of viral proteins regulate various stages of infection. With only 10 proteins, hepatitis C virus (HCV) can orchestrate its complete viral life cycle. HCV non-structural protein 3 (NS3) has many functions. It has protease and helicase activities, interacts with several host-cell proteins and plays a role in translation, replication and virus-particle formation. Organization of all these functions is necessary and could be regulated by PTMs. We therefore searched for modifications of the NS3 protein in the subgenomic HCV replicon. When performing a tag-capture approach coupled with two-dimensional gel electrophoresis analyses, we observed that isolated His6-NS3 yielded multiple spots. Individual protein spots were digested in gel and analysed by mass spectrometry. Differences observed between the individual peptide mass fingerprints suggested the presence of modified peptides and allowed us to identify N-terminal acetylation and an adaptive mutation of NS3 (Q1067R). Further analysis of other NS3 variants revealed phosphorylation of NS3.


Subject(s)
Protein Processing, Post-Translational , Viral Nonstructural Proteins/metabolism , Virus Replication , Amino Acid Sequence , Electrophoresis, Gel, Two-Dimensional , Hepacivirus/physiology , Humans , Molecular Sequence Data , Phosphorylation
9.
Virol J ; 6: 230, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20034394

ABSTRACT

BACKGROUND: The spike protein (S) of SARS Coronavirus (SARS-CoV) mediates entry of the virus into target cells, including receptor binding and membrane fusion. Close to or in the viral membrane, the S protein contains three distinct motifs: a juxtamembrane aromatic part, a central highly hydrophobic stretch and a cysteine rich motif. Here, we investigate the role of aromatic and hydrophobic parts of S in the entry of SARS CoV and in cell-cell fusion. This was investigated using the previously described SARS pseudotyped particles system (SARSpp) and by fluorescence-based cell-cell fusion assays. RESULTS: Mutagenesis showed that the aromatic domain was crucial for SARSpp entry into cells, with a likely role in pore enlargement.Introduction of lysine residues in the hydrophobic stretch of S also resulted in a block of entry, suggesting the borders of the actual transmembrane domain. Surprisingly, replacement of a glycine residue, situated close to the aromatic domain, with a lysine residue was tolerated, whereas the introduction of a lysine adjacent to the glycine, was not. In a model, we propose that during fusion, the lateral flexibility of the transmembrane domain plays a critical role, as do the tryptophans and the cysteines. CONCLUSIONS: The aromatic domain plays a crucial role in the entry of SARS CoV into target cells. The positioning of the aromatic domain and the hydrophobic domain relative to each other is another essential characteristic of this membrane fusion process.


Subject(s)
Membrane Fusion , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Amino Acid Sequence , Cell Fusion , Cell Line , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Mutagenesis , Severe acute respiratory syndrome-related coronavirus/genetics , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/metabolism
10.
Virol J ; 6: 62, 2009 May 25.
Article in English | MEDLINE | ID: mdl-19467155

ABSTRACT

BACKGROUND: Hepatitis C virus (HCV) induces membrane rearrangements during replication. All HCV proteins are associated to membranes, pointing out the importance of membranes for HCV. Non structural protein 4B (NS4B) has been reported to induce cellular membrane alterations like the membranous web. Four transmembrane segments in the middle of the protein anchor NS4B to membranes. An amphipatic helix at the amino-terminus attaches to membranes as well. The carboxy-terminal domain (CTD) of NS4B is highly conserved in Hepaciviruses, though its function remains unknown. RESULTS: A cytosolic localization is predicted for the NS4B-CTD. However, using membrane floatation assays and immunofluorescence, we now show targeting of the NS4B-CTD to membranes. Furthermore, a profile-profile search, with an HCV NS4B-CTD multiple sequence alignment, indicates sequence similarity to the membrane binding domain of prokaryotic D-lactate dehydrogenase (d-LDH). The crystal structure of E. coli d-LDH suggests that the region similar to NS4B-CTD is located in the membrane binding domain (MBD) of d-LDH, implying analogy in membrane association. Targeting of d-LDH to membranes occurs via electrostatic interactions of positive residues on the outside of the protein with negative head groups of lipids. To verify that anchorage of d-LDH MBD and NS4B-CTD is analogous, NS4B-CTD mutants were designed to disrupt these electrostatic interactions. Membrane association was confirmed by swopping the membrane contacting helix of d-LDH with the corresponding domain of the 4B-CTD. Furthermore, the functionality of these residues was tested in the HCV replicon system. CONCLUSION: Together these data show that NS4B-CTD is associated to membranes, similar to the prokaryotic d-LDH MBD, and is important for replication.


Subject(s)
Cell Membrane/virology , Hepacivirus/physiology , Viral Nonstructural Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Cell Line , Escherichia coli Proteins/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Sequence Homology, Amino Acid
11.
J Virol ; 82(1): 451-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17959675

ABSTRACT

Virus infection in vitro can either result in a cytopathic effect (CPE) or proceed without visible changes in infected cells (noncytopathic infection). We are interested in understanding the mechanisms controlling the impact of coronavirus infection on host cells. To this end, we compared a productive, noncytopathic infection of murine hepatitis virus (MHV) strain A59 in the fibroblastlike cell line NIH 3T3 with cytopathic MHV infections. Infected NIH 3T3 cells could be cultured for up to 4 weeks without apparent CPE and yet produce virus at 10(7) to 10(8) PFU/ml. Using flow cytometry, we demonstrated that NIH 3T3 cells expressed as much MHV receptor CEACAM1 as other cell lines which die from MHV infection. In contrast, using quantitative reverse transcription-PCR and metabolic labeling of RNA, we found that the rate of viral RNA amplification in NIH 3T3 cells was lower than the rate in cells in which MHV induces a CPE. The rate of cellular RNA synthesis in contact-inhibited confluent NIH 3T3 cells was also lower than in cells permissive to cytopathic MHV infection. However, the induction of cellular RNA synthesis in growing NIH 3T3 cells did not result in an increase of either viral RNA amplification or CPE. Our results suggest that a specific, receptor CEACAM1-independent mechanism restricting coronaviral RNA synthesis and CPE is present in NIH 3T3 and, possibly, other cells with preserved contact inhibition.


Subject(s)
Murine hepatitis virus/growth & development , Virus Replication , Animals , Carcinoembryonic Antigen/analysis , Cell Membrane/chemistry , Cytopathogenic Effect, Viral , Cytosol/chemistry , Flow Cytometry , Mice , Murine hepatitis virus/physiology , NIH 3T3 Cells , RNA, Viral/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
12.
J Virol ; 81(20): 10981-90, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17670839

ABSTRACT

Murine hepatitis virus (MHV) and severe acute respiratory syndrome (SARS) coronavirus (CoV) are two of the best-studied representatives of the family Coronaviridae. During CoV infection, numerous cytokines and chemokines are induced in vitro and in vivo. Human interleukin 8 and its mouse functional counterpart, CXCL2, are early-expressed chemokines. Here we show that SARS-CoV and MHV induce endoplasmic reticulum (ER) stress and Cxcl2 mRNA transcription during infection in vitro. Expression of the viral spike protein significantly induced ER stress and Cxcl2 mRNA upregulation, while expression of the other structural genes did not. Additional experiments with UV-inactivated virus, cell-cell fusion-blocking antibodies, and an MHV mutant with a defect in spike protein maturation demonstrated that spike-host interactions in the ER are responsible for the induction of ER stress and subsequent Cxcl2 mRNA transcription. Despite significant increases in levels of Cxcl2 mRNA and functional nucleus-to-cytoplasm RNA transport, no CXCL2 protein was released into the medium from MHV-infected cells. Yet Sendai virus-infected cells showed substantial Cxcl2 mRNA induction and a simultaneous increase in levels of secreted CXCL2 protein. Our results demonstrate that expression of CoV spike proteins induces ER stress, which could subsequently trigger innate immune responses. However, at that point in infection, translation of host mRNA is already severely reduced in infected cells, preventing the synthesis of CXCL2 and ER stress proteins despite their increased mRNA concentrations.


Subject(s)
Chemokine CXCL2/genetics , Endoplasmic Reticulum/virology , Membrane Glycoproteins/physiology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Viral Envelope Proteins/physiology , Animals , Cell Line , Chemokines/genetics , Endoplasmic Reticulum/pathology , Mice , Murine hepatitis virus/pathogenicity , RNA, Messenger/analysis , Spike Glycoprotein, Coronavirus , Up-Regulation
13.
J Virol ; 81(15): 8352-5, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17507478

ABSTRACT

Recently, a paper was published in which it was proposed that the GxxxG motif of the severe acute respiratory syndrome (SARS) coronavirus spike (S) protein transmembrane domain plays a vital role in oligomerization of the protein (E. Arbely, Z. Granot, I. Kass, J. Orly, and I. T. Arkin, Biochemistry 45:11349-11356, 2006). Here, we show that the GxxxG motif is not involved in SARS S oligomerization by trimerization analysis of S GxxxG mutant proteins. In addition, the capability of S to mediate entry of SARS S-pseudotyped particles overall was affected moderately in the mutant proteins, also arguing for a nonvital role for the GxxxG motif in SARS coronavirus entry.


Subject(s)
Amino Acid Motifs , Membrane Glycoproteins , Protein Structure, Quaternary , Severe Acute Respiratory Syndrome , Severe acute respiratory syndrome-related coronavirus , Viral Envelope Proteins , Virus Internalization , Animals , Cells, Cultured , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/metabolism , Sequence Alignment , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
14.
J Gen Virol ; 88(Pt 6): 1738-1747, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17485534

ABSTRACT

The pentanucleotide (PN) sequence 5'-CACAG-3' at the top of the 3' stem-loop structure of the flavivirus genome is well conserved in the arthropod-borne viruses but is more variable in flaviviruses with no known vector. In this study, the sequence requirements of the PN motif for yellow fever virus 17D (YFV) replication were determined. In general, individual mutations at either the second, third or fourth positions were tolerated and resulted in replication-competent virus. Mutations at the fifth position were lethal. Base pairing of the nucleotide at the first position of the PN motif and a nucleotide four positions downstream of the PN (ninth position) was a major determinant for replication. Despite the fact that the majority of the PN mutants were able to replicate efficiently, they were outcompeted by parental YFV-17D virus following repeated passages in double-infected cell cultures. Surprisingly, some of the virus mutants at the first and/or the ninth position that maintained the possibility of forming a base pair were found to have a similar fitness to YFV-17D under these conditions. Overall, these experiments suggest that YFV is less dependent on sequence conservation of the PN motif for replication in animal cells than West Nile virus. However, in animal cell culture, YFV has a preference for the wt CACAG PN sequence. The molecular mechanisms behind this preference remain to be elucidated.


Subject(s)
3' Untranslated Regions , Conserved Sequence , Genome, Viral , RNA, Viral/genetics , Virus Replication , Yellow fever virus/genetics , Yellow fever virus/physiology , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , Cricetinae , Models, Molecular , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , RNA, Viral/physiology , West Nile virus/genetics
15.
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
16.
Hepatology ; 45(3): 602-13, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17326154

ABSTRACT

UNLABELLED: Broad T cell and B cell responses to multiple HCV antigens are observed early in individuals who control or clear HCV infection. The prevailing hypothesis has been that similar immune responses induced by prophylactic immunization would reduce acute virus replication and protect exposed individuals from chronic infection. Here, we demonstrate that immunization of naïve chimpanzees with a multicomponent HCV vaccine induced robust HCV-specific immune responses, and that all vaccinees exposed to heterologous chimpanzee-adapted HCV 1b J4 significantly reduced viral RNA in serum by 84%, and in liver by 99% as compared to controls (P=0.024 and 0.028, respectively). However, despite control of HCV in plasma and liver in the acute period, in the chronic phase, 3 of 4 vaccinated animals developed persistent infection. Analysis of expression levels of proinflammatory cytokines in serial hepatic biopsies failed to reveal an association with vaccine outcome. However, expression of IDO, CTLA-4 [corrected] and PD-1 levels in liver correlated with clearance or chronicity. CONCLUSION: Despite early control of virus load, a virus-associated tolerogenic-like state can develop in certain individuals independent of vaccination history.


Subject(s)
Antigens, CD/metabolism , Hepatitis C/immunology , Viral Hepatitis Vaccines/therapeutic use , Animals , Antigens, Viral/immunology , Apoptosis Regulatory Proteins/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease/prevention & control , Cytokines/metabolism , DNA, Viral/genetics , Hepacivirus/genetics , Hepacivirus/immunology , Hepatitis C/prevention & control , Pan troglodytes , Programmed Cell Death 1 Receptor , Viral Load
17.
J Gen Virol ; 87(Pt 7): 1961-1975, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16760398

ABSTRACT

Understanding the orchestrated genome-wide cellular responses is critical for comprehending the early events of coronavirus infection. Microarray analysis was applied to assess changes in cellular expression profiles during different stages of two independent, highly controlled murine hepatitis virus (MHV) infections in vitro. Fibroblast-like L cells were infected at high multiplicity in order to study the direct effects of a synchronized lytic coronavirus infection. Total RNA was harvested from MHV- or mock-infected L cells at 3, 5 and 6 h post-infection and hybridized to Affymetrix microarrays representing approximately 12,500 murine genes and expressed sequences. The expression data were compared to their respective mock-infected controls. Quantitative RT-PCR of selected transcripts was used to validate the differential expression of transcripts and inter-experiment reproducibility of microarray analysis. It was concluded that MHV-A59 infection in fibroblast-like cells triggers very few transcriptional cellular responses in the first 3 h of infection. Later, after having established a productive infection, a chemokine response is induced together with other cellular changes associated with RNA and protein metabolism, cell cycle and apoptosis. Interferon responses are not triggered during infection, although the L cells can be readily stimulated to produce interferon by dsRNA, a known potent inducer of interferon. Possibly, the interferon response is actively counteracted by a virus-encoded antagonist as has been described previously for other RNA viruses.


Subject(s)
Murine hepatitis virus/genetics , Murine hepatitis virus/pathogenicity , Animals , Apoptosis , Cell Cycle , Cytopathogenic Effect, Viral , DNA Repair , Gene Expression Profiling , Immunity, Innate , Inflammation Mediators/metabolism , L Cells , Mice , Murine hepatitis virus/immunology , Murine hepatitis virus/physiology , Oligonucleotide Array Sequence Analysis , Oxidative Stress , RNA, Viral/biosynthesis , RNA, Viral/genetics , Signal Transduction , Transcription, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication
18.
J Gen Virol ; 87(Pt 6): 1403-1421, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16690906

ABSTRACT

Many positive-stranded RNA viruses use subgenomic mRNAs to express part of their genetic information. To produce structural and accessory proteins, members of the order Nidovirales (corona-, toro-, arteri- and roniviruses) generate a 3' co-terminal nested set of at least three and often seven to nine mRNAs. Coronavirus and arterivirus subgenomic transcripts are not only 3' co-terminal but also contain a common 5' leader sequence, which is derived from the genomic 5' end. Their synthesis involves a process of discontinuous RNA synthesis that resembles similarity-assisted RNA recombination. Most models proposed over the past 25 years assume co-transcriptional fusion of subgenomic RNA leader and body sequences, but there has been controversy over the question of whether this occurs during plus- or minus-strand synthesis. In the latter model, which has now gained considerable support, subgenomic mRNA synthesis takes place from a complementary set of subgenome-size minus-strand RNAs, produced by discontinuous minus-strand synthesis. Sense-antisense base-pairing interactions between short conserved sequences play a key regulatory role in this process. In view of the presumed common ancestry of nidoviruses, the recent finding that ronivirus and torovirus mRNAs do not contain a common 5' leader sequence is surprising. Apparently, major mechanistic differences must exist between nidoviruses, which raises questions about the functions of the common leader sequence and nidovirus transcriptase proteins and the evolution of nidovirus transcription. In this review, nidovirus transcription mechanisms are compared, the experimental systems used are critically assessed and, in particular, the impact of recently developed reverse genetic systems is discussed.


Subject(s)
Nidovirales/metabolism , Transcription, Genetic , Animals , Gene Expression Regulation, Viral , Humans , Nidovirales/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
19.
J Virol ; 80(9): 4336-43, 2006 May.
Article in English | MEDLINE | ID: mdl-16611892

ABSTRACT

The 3' nontranslated region (NTR) of the hepatitis C virus (HCV) genome is highly conserved and contains specific cis-acting RNA motifs that are essential in directing the viral replication machinery to initiate at the correct 3' end of the viral genome. Since the ends of viral genomes may be damaged by cellular RNases, preventing the initiation of viral RNA replication, stable RNA hairpin structures in the 3' NTR may also be essential in host defense against exoribonucleases. During 3'-terminal sequence analysis of serum samples of a patient with chronic hepatitis related to an HCV1b infection, a number of clones were obtained that were several nucleotides shorter at the extreme 3' end of the genome. These shorter 3' ends were engineered in selectable HCV replicons in order to enable the study of RNA replication in cell culture. When in vitro-transcribed subgenomic RNAs, containing shorter 3' ends, were introduced into Huh-7 cells, a few selectable colonies were obtained, and the 3' terminus of these subgenomic RNAs was sequenced. Interestingly, most genomes recovered from these colonies had regained the wild-type 3' ends, showing that HCV, like several other positive-stranded RNA viruses, has developed a strategy to repair deleted 3' end nucleotides. Furthermore, we found several genomes in these replicon colonies that contained a poly(A) tail and a short linker sequence preceding the poly(A) tail. After recloning and subsequent passage in Huh-7 cells, these poly(A) tails persisted and varied in length. In addition, the connecting linker became highly diverse in sequence and length, suggesting that these tails are actively replicated. The possible terminal repair mechanisms, including roles for the poly(A) tail addition, are discussed.


Subject(s)
3' Untranslated Regions/genetics , Genetic Variation/genetics , Hepacivirus/genetics , Poly A/metabolism , Polyadenylation/genetics , Replicon/genetics , Base Sequence , Cell Line, Tumor , Genome, Viral/genetics , Genotype , Hepacivirus/chemistry , Hepacivirus/isolation & purification , Humans , Molecular Sequence Data , Nucleic Acid Conformation , Poly A/genetics
20.
J Virol ; 80(3): 1302-10, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415007

ABSTRACT

The spike protein (S) of severe acute respiratory syndrome coronavirus (SARS-CoV) is responsible for receptor binding and membrane fusion. It contains a highly conserved transmembrane domain that consists of three parts: an N-terminal tryptophan-rich domain, a central domain, and a cysteine-rich C-terminal domain. The cytoplasmic tail of S has previously been shown to be required for assembly. Here, the roles of the transmembrane and cytoplasmic domains of S in the infectivity and membrane fusion activity of SARS-CoV have been studied. SARS-CoV S-pseudotyped retrovirus (SARSpp) was used to measure S-mediated infectivity. In addition, the cell-cell fusion activity of S was monitored by a Renilla luciferase-based cell-cell fusion assay. S(VSV-Cyt), an S chimera with a cytoplasmic tail derived from vesicular stomatitis virus G protein (VSV-G), and S(MHV-TMDCyt), an S chimera with the cytoplasmic and transmembrane domains of mouse hepatitis virus, displayed wild-type-like activity in both assays. S(VSV-TMDCyt), a chimera with the cytoplasmic and transmembrane domains of VSV-G, was impaired in the SARSpp and cell-cell fusion assays, showing 3 to 25% activity compared to the wild type, depending on the assay and the cells used. Examination of the oligomeric state of the chimeric S proteins in SARSpp revealed that S(VSV-TMDCyt) trimers were less stable than wild-type S trimers, possibly explaining the lowered fusogenicity and infectivity.


Subject(s)
Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , DNA, Viral/genetics , Humans , Luciferases, Renilla/genetics , Membrane Fusion/physiology , Membrane Glycoproteins/genetics , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Virus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Spike Glycoprotein, Coronavirus , Vero Cells , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...