Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters










Publication year range
1.
Ann ICRP ; 43(2): 5-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25915837

ABSTRACT

The use of technologies to provide security screening for individuals and objects has been increasing rapidly, in keeping with the significant increase in security concerns worldwide. Within the spectrum of technologies, the use of ionizing radiation to provide backscatter and transmission screening capabilities has also increased. The Commission has previously made a number of statements related to the general topic of deliberate exposures of individuals in non-medical settings. This report provides advice on how the radiological protection principles recommended by the Commission should be applied within the context of security screening. More specifically, the principles of justification, optimisation of protection, and dose limitation for planned exposure situations are directly applicable to the use of ionising radiation in security screening. In addition, several specific topics are considered in this report, including the situation in which individuals may be exposed because they are concealed ('stowaways') in a cargo container or conveyance that may be subject to screening. The Commission continues to recommend that careful justification of screening should be considered before decisions are made to employ the technology. If a decision is made that its use is justified, the framework for protection as a planned exposure situation should be employed, including optimization of protection with the use of dose constraints and the appropriate provisions for authorisation and inspection.


Subject(s)
Environmental Exposure/prevention & control , Radiation Monitoring , Radiation Protection/methods , Security Measures/standards , Humans , Occupational Exposure/prevention & control , Radiation Protection/standards
2.
Public Health ; 124(6): 313-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20542303

ABSTRACT

OBJECTIVES: Mr Alexander Litvinenko died in a London hospital on 23 November 2006, allegedly from poisoning with the radionuclide polonium-210 ((210)Po). Associated circumstances required an integrated response to investigate the potential risk of internal contamination for individuals exposed to contaminated environments. STUDY DESIGN: Descriptive study. METHODS: Contaminated locations presenting a potential risk to health were identified through environmental assessment by radiation protection specialists. Individuals connected with these locations were identified and assessed for internal contamination with (210)Po. RESULTS: In total, 1029 UK residents were identified, associated with the 11 most contaminated locations. Of these, 974 were personally interviewed and 787 were offered urine tests for (210)Po excretion. Overall, 139 individuals (18%) showed evidence of probable internal contamination with (210)Po arising from the incident, but only 53 (7%) had assessed radiation doses of 1mSv or more. The highest assessed radiation dose was approximately 100mSv. CONCLUSIONS: Although internal contamination with (210)Po was relatively frequent and was most extensive among individuals associated with locations judged a priori to pose the greatest risk, a high degree of assurance could be given to UK and international communities that the level of health risk from exposure to the radionuclide in this incident was low.


Subject(s)
Air Pollution, Radioactive/adverse effects , Polonium/poisoning , Public Health/methods , Radiation Injuries/etiology , Radioactive Hazard Release , Cluster Analysis , Humans , London , Polonium/urine , Public Health/standards , Radiation Injuries/urine , Risk Assessment/methods
3.
J Mol Biol ; 314(5): 1029-39, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11743720

ABSTRACT

Parvovirus initiation factor (PIF), identified in HeLa cells as a host factor essential for parvoviral DNA replication, is a ubiquitous heterodimeric cellular transcription factor. This protein complex was simultaneously identified as glucocorticoid modulatory element binding protein (GMEB) by its ability to bind to the glucocorticoid modulating element (GME) upstream of the tyrosine transaminase promoter. Here, we show that the two PIF/GMEB subunits form site-specific DNA-binding heterodimers when co-expressed from recombinant baculoviruses and homodimers when expressed separately. Degenerate oligonucleotide selection experiments, combined with analysis of dissociation rates, established that the three complexes bind to flexibly spaced tetranucleotide half-sites that conform to the consensus ACGPy N(1-9) PuCGPy, with an optimum of N=6. Binding of all three complexes is extremely sensitive to methylation of the cytosine residues in the invariant CpG half-site core, suggesting a means by which PIF/GMEB binding could be regulated in vivo. Because CpG dinucleotides are suppressed in eukaryotic genomes, such binding sites would be expected to be very rare. However, analysis of 100 human promoters showed that over half of them contained at least one site conforming to the consensus, a significant deviation from the expected random distribution. In many of these, the binding site is within 100 nucleotides of the transcriptional start site, indicating that PIF/GMEB may be involved in regulation of these genes. Oligonucleotides corresponding to five of these sequences, chosen to represent the range of half-site separations identified by the consensus, were tested for PIF/GMEB binding by mobility shift assay. All five probes bound the heterodimer efficiently and, in each case, binding was completely abrogated by 5-methylation of the C residues in the CpGs of the putative half-sites.


Subject(s)
Consensus Sequence/genetics , CpG Islands/genetics , DNA Methylation , Promoter Regions, Genetic/genetics , Response Elements/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism , Amino Acid Motifs , Base Sequence , Binding Sites , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dimerization , Electrophoretic Mobility Shift Assay , HeLa Cells , Humans , Kinetics , Molecular Weight , Protein Binding , Protein Subunits , Substrate Specificity
4.
Virology ; 288(1): 154-63, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11543668

ABSTRACT

Previously it was shown that a 53-nucleotide viral replication origin, derived from the left-end (3') telomere of minute virus of mice (MVM) DNA, directed integration of infecting MVM genomes into an Epstein-Barr virus (EBV)-based episome in cell culture. Integration depended upon the presence, in the episome, of a functional origin sequence which could be nicked by NS1, the viral initiator protein. Here we extend our studies to the genomic right-end (5') origin and report that three 131- to 135-nucleotide right-end origin sequences failed to target MVM episomal integration even though the same sequences were functional in NS1-driven DNA replication assays in vitro. Additionally, we observed amplification of episomal DNA in response to MVM infection in cell lines harboring episomes which directed integration, but not in cell lines containing episomes which did not direct integration, including those with inserts of the MVM right-end origin.


Subject(s)
DNA Replication , DNA, Viral/genetics , DNA, Viral/metabolism , Minute Virus of Mice/physiology , Plasmids/genetics , Virus Integration , Virus Replication/genetics , 5' Untranslated Regions/genetics , Animals , Cell Line , DNA, Viral/chemistry , HeLa Cells , Humans , Mice , Minute Virus of Mice/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
5.
J Virol ; 75(15): 7009-17, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11435581

ABSTRACT

Parvoviral rolling hairpin replication generates palindromic genomic concatemers whose junctions are resolved to give unit-length genomes by a process involving DNA replication initiated at origins derived from each viral telomere. The left-end origin of minute virus of mice (MVM), oriL, contains binding sites for the viral initiator nickase, NS1, and parvovirus initiation factor (PIF), a member of the emerging KDWK family of transcription factors. oriL is generated as an active form, oriL(TC), and as an inactive form, oriL(GAA), which contains a single additional nucleotide inserted between the NS1 and PIF sites. Here we examined the interactions on oriL(TC) which lead to activation of NS1 by PIF. The two subunits of PIF, p79 and p96, cooperatively bind two ACGT half-sites, which can be flexibly spaced. When coexpressed from recombinant baculoviruses, the PIF subunits preferentially form heterodimers which, in the presence of ATP, show cooperative binding with NS1 on oriL, but this interaction is preferentially enhanced on oriL(TC) compared to oriL(GAA). Without ATP, NS1 is unable to bind stably to its cognate site, but PIF facilitates this interaction, rendering the NS1 binding site, but not the nick site, resistant to DNase I. Varying the spacing of the PIF half-sites shows that the distance between the NS1 binding site and the NS1-proximal half-site is critical for nickase activation, whereas the position of the distal half-site is unimportant. When expressed separately, both PIF subunits form homodimers that bind site specifically to oriL, but only complexes containing p79 activate the NS1 nickase function.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Minute Virus of Mice/genetics , Replication Origin , Transcription Factors/metabolism , Viral Nonstructural Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Base Sequence , Binding Sites , DNA Replication , DNA-Binding Proteins/genetics , Dimerization , HeLa Cells , Humans , Mice , Minute Virus of Mice/metabolism , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics , Viral Nonstructural Proteins/genetics
8.
J Virol ; 74(3): 1332-41, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10627544

ABSTRACT

Minute virus of mice (MVM) replicates via a linearized form of rolling-circle replication in which the viral nickase, NS1, initiates DNA synthesis by introducing a site-specific nick into either of two distinct origin sequences. In vitro nicking and replication assays with substrates that had deletions or mutations were used to explore the sequences and structural elements essential for activity of one of these origins, located in the right-end (5') viral telomere. This structure contains 248 nucleotides, most-favorably arranged as a simple hairpin with six unpaired bases. However, a pair of opposing NS1 binding sites, located near its outboard end, create a 33-bp palindrome that could potentially assume an alternate cruciform configuration and hence directly bind HMG1, the essential cofactor for this origin. The palindromic nature of this sequence, and thus its ability to fold into a cruciform, was dispensable for origin function, as was the NS1 binding site occupying the inboard arm of the palindrome. In contrast, the NS1 site in the outboard arm was essential for initiation, even though positioned 120 bp from the nick site. The specific sequence of the nick site and an additional NS1 binding site which directly orients NS1 over the initiation site were also essential and delimited the inboard border of the minimal right-end origin. DNase I and hydroxyl radical footprints defined sequences protected by NS1 and suggest that HMG1 allows the NS1 molecules positioned at each end of the origin to interact, creating a distortion characteristic of a double helical loop.


Subject(s)
DNA Replication , DNA, Viral/metabolism , High Mobility Group Proteins/metabolism , Minute Virus of Mice/genetics , Replication Origin/genetics , Animals , Base Sequence , DNA Footprinting , DNA, Viral/chemistry , HeLa Cells , High Mobility Group Proteins/genetics , Humans , Mice , Minute Virus of Mice/metabolism , Minute Virus of Mice/physiology , Molecular Sequence Data , Nucleic Acid Conformation , Telomere , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
9.
Mol Cell Biol ; 19(11): 7741-50, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10523663

ABSTRACT

Initially recognized as a HeLa factor essential for parvovirus DNA replication, parvovirus initiation factor (PIF) is a site-specific DNA-binding complex consisting of p96 and p79 subunits. We have cloned and sequenced the human cDNAs encoding each subunit and characterized their products expressed from recombinant baculoviruses. The p96 and p79 polypeptides have 40% amino acid identity, focused particularly within a 94-residue region containing the sequence KDWK. This motif, first described for the Drosophila homeobox activator DEAF-1, identifies an emerging group of metazoan transcriptional modulators. During viral replication, PIF critically regulates the viral nickase, but in the host cell it probably modulates transcription, since each subunit is active in promoter activation assays and the complex binds to previously described regulatory elements in the tyrosine aminotransferase and transferrin receptor promoters. Within its recognition site, PIF binds coordinately to two copies of the tetranucleotide PuCGPy, which, remarkably, can be spaced from 1 to 15 nucleotides apart, a novel flexibility that we suggest may be characteristic of the KDWK family. Such tetranucleotides are common in promoter regions, particularly in activating transcription factor/cyclic AMP response element-binding protein (ATF/CREB) and E-box motifs, suggesting that PIF may modulate the transcription of many genes.


Subject(s)
Amino Acid Motifs , DNA-Binding Proteins/metabolism , GC Rich Sequence , Multigene Family , Transcription Factors/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Dimerization , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Parvovirus/genetics , Promoter Regions, Genetic , Protein Binding , Receptors, Transferrin/genetics , Recombinant Proteins/metabolism , Replication Origin , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/isolation & purification , Tyrosine Transaminase/genetics
10.
Hum Gene Ther ; 10(10): 1619-32, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10428207

ABSTRACT

The replication of viral genomes and the production of recombinant viral vectors from infectious molecular clones of parvoviruses MVMp and H1 were greatly improved by the introduction of a consensus NS-1 nick site at the junction between the left-hand viral terminus and the plasmid DNA. Progressive deletions of up to 1600 bp in the region encoding the structural genes as well as insertions of foreign DNA in replacement of those sequences did not appreciably affect the replication ability of the recombinant H1 virus genomes. In contrast, the incorporation of these genomes into recombinant particles appeared to depend on in cis-provided structural gene sequences. Indeed, the production of H1 viral vectors by cotransfection of recombinant clones and helper plasmids providing the structural proteins (VPs) in trans, drastically decreased when more than 800 bp was removed from the VP transcription unit. Furthermore, titers of viral vectors, in which most of the VP-coding region was replaced by an equivalent-length sequence consisting of reporter cDNA and stuffer DNA, were reduced more than 50 times in comparison with recombinant vectors in which stuffer DNA was not substituted for the residual VP sequence. In addition, viral vector production was restricted by the overall size of the genome, with a mere 6% increase in DNA length leading to an approximately 10 times lower encapsidation yield. Under conditions fulfilling the above-mentioned requirements for efficient packaging, titers of virus vectors from improved recombinant molecular DNA clones amounted to 5 x 10(7) infectious units per milliliter of crude extract. These titers should allow the assessment of the therapeutic effect of recombinant parvoviruses expressing small transgenes in laboratory animals.


Subject(s)
DNA, Viral , Genetic Vectors , Parvovirus , Capsid/genetics , Capsid Proteins , Cell Line, Transformed , Chemokine CCL2/genetics , DNA Replication , Gene Expression , HeLa Cells , Humans , Mutagenesis , Parvovirus/genetics , Parvovirus/physiology , Recombination, Genetic , Virus Replication
11.
Virology ; 254(1): 169-81, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9927584

ABSTRACT

Antisera were raised against peptides corresponding to the N-termini of capsid proteins VP1 and VP2 from the parvovirus minute virus of mice. Epitopes in the 142-amino-acid VP1-specific region were not accessible in the great majority of newly released viral particles, and sera directed against them failed to neutralize virus directly or deplete stocks of infectious virions. However, brief exposure to temperatures of 45 degreesC or more induced a conformational transition in a population of full virions, but not in empty viral particles, in which VP1-specific sequences became externally accessible. In contrast, the VP2 N-terminus was antibody-accessible in all full, but not empty, particles without prior treatment. An electrophoretic mobility shift assay, in which particles were heat-treated and/or preincubated with antibodies prior to electrophoresis, confirmed this pattern of epitope accessibility, showing that the heat-induced conformational transition produces a retarded form of virion that can be supershifted by incubation with VP1-specific sera. The proportion of virions undergoing transition increased with temperature, but at all temperatures up to 70 degreesC viral particles retained structure-specific antigenic determinants and remained essentially intact, without shedding individual polypeptide species or subunits. However, despite the apparent integrity of its protective coat, the genome became accessible to externally applied enzymes in an increasing proportion of virions through this temperature range, suggesting that the conformational transitions that expose VP1 likely also allow access to the genome. Heating particles to 80 degreesC or above finally induced disassembly to polypeptide monomers.


Subject(s)
Capsid/chemistry , Genome, Viral , Minute Virus of Mice/physiology , Protein Conformation , Virus Assembly , Amino Acid Sequence , Animals , Antigens, Viral/immunology , Capsid/immunology , Capsid Proteins , DNA, Viral , Epitopes, B-Lymphocyte/immunology , Heating , Mice , Minute Virus of Mice/genetics , Minute Virus of Mice/immunology , Molecular Sequence Data , Virion
12.
Structure ; 6(11): 1369-81, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9817841

ABSTRACT

BACKGROUND: Minute virus of mice (MVM) is a single-stranded (ss) DNA-containing, murine parvovirus with a capsid built up of 60 icosahedrally related polypeptide chains, each of which consists of the C-terminal region common to two structural proteins, VP1 and VP2. In infectious virions, most VP2 molecules are cleaved to VP3 by the removal of about 20 amino acids from the N terminus. Of the 587 amino acids in VP2, approximately half are identical to those in the analogous capsid protein of the antigenically distinct canine parvovirus (CPV), the crystal structure of which has previously been determined. The three-dimensional structure determination of MVMi (the immunosuppressive strain of MVM) was previously reported to 3.5 A resolution. RESULTS: We report here an analysis of the MVMi virus structure and provide insights into tissue tropism, antigenicity and DNA packaging. Amino acids determining MVM tissue tropism were found to cluster on, or near, the viral surface. A conserved, glycine-rich, N-terminal peptide was seen to run through a cylindrical channel along each fivefold axis and may have implications for antigenicity. Density within the virion was interpreted as 29 ssDNA nucleotides per icosahedral asymmetric unit, and accounts for over one-third of the viral genome. CONCLUSIONS: The presence of the glycine-rich sequence in the fivefold channels of MVMi provides a possible mechanism to explain how the unique N-terminal region of VP1 becomes externalized in infectious parvovirions. Residues that determine tropism may form an attachment recognition site for a secondary host-cell factor that modulates tissue specificity. The ordering of nucleotides in a similar region of the interior surface in the CPV and MVMi capsids suggests the existence of a genomic DNA-recognition site within the parvoviral capsid.


Subject(s)
Capsid Proteins , Capsid/chemistry , Minute Virus of Mice/chemistry , Amino Acid Sequence , Glycine/chemistry , Minute Virus of Mice/physiology , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Tropism
13.
J Virol ; 72(11): 8477-84, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9765384

ABSTRACT

Rolling-circle replication is initiated by a replicon-encoded endonuclease which introduces a single-strand nick into specific origin sequences, becoming covalently attached to the 5' end of the DNA at the nick and providing a 3' hydroxyl to prime unidirectional, leading-strand synthesis. Parvoviruses, such as minute virus of mice (MVM), have adapted this mechanism to amplify their linear single-stranded genomes by using hairpin telomeres which sequentially unfold and refold to shuttle the replication fork back and forth along the genome, creating a continuous, multimeric DNA strand. The viral initiator protein, NS1, then excises individual genomes from this continuum by nicking and reinitiating synthesis at specific origins present within the hairpin sequences. Using in vitro assays to study ATP-dependent initiation within the right-hand (5') MVM hairpin, we have characterized a HeLa cell factor which is absolutely required to allow NS1 to nick this origin. Unlike parvovirus initiation factor (PIF), the cellular complex which activates NS1 endonuclease activity at the left-hand (3') viral origin, the host factor which activates the right-hand hairpin elutes from phosphocellulose in high salt, has a molecular mass of around 25 kDa, and appears to bind preferentially to structured DNA, suggesting that it might be a member of the high-mobility group 1/2 (HMG1/2) protein family. This prediction was confirmed by showing that purified calf thymus HMG1 and recombinant human HMG1 or murine HMG2 could each substitute for the HeLa factor, activating the NS1 endonuclease in an origin-specific nicking reaction.


Subject(s)
DNA Replication/physiology , DNA, Viral/biosynthesis , High Mobility Group Proteins/metabolism , Minute Virus of Mice/metabolism , Animals , Base Sequence , DNA, Viral/chemistry , DNA, Viral/genetics , Endonucleases/genetics , Endonucleases/metabolism , HeLa Cells , Humans , Mice , Minute Virus of Mice/genetics , Nucleic Acid Conformation , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
14.
J Virol ; 72(10): 8002-12, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9733839

ABSTRACT

NS1, the 83-kDa major nonstructural protein of minute virus of mice (MVM), is a multifunctional nuclear phosphoprotein which is required in a variety of steps during progeny virus production, early as well as late during infection. NS1 is the initiator protein for viral DNA replication. It binds specifically to target DNA motifs; has site-specific single-strand nickase, intrinsic ATPase, and helicase activities; trans regulates viral and cellular promoters; and exerts cytotoxic stress on the host cell. To investigate whether these multiple activities of NS1 depend on posttranslational modifications, in particular phosphorylation, we expressed His-tagged NS1 in HeLa cells by using recombinant vaccinia viruses, dephosphorylated it at serine and threonine residues with calf intestine alkaline phosphatase, and compared the biochemical activities of the purified un(der)phosphorylated (NS1(O)) and the native (NS1(P)) polypeptides. Biochemical analyses of replicative functions of NS1(O) revealed a severe reduction of intrinsic helicase activity and, to a minor extent, of ATPase and nickase activities, whereas its affinity for the target DNA sequence [ACCA]2-3 was enhanced compared to that of NS1(P). In the presence of endogenous protein kinases found in replication extracts, NS1(O) showed all functions necessary for resolution and replication of the 3' dimer bridge, indicating reactivation of NS1(O) by rephosphorylation. Partial reactivation of the helicase activity was found as well when NS1(O) was incubated with protein kinase C.


Subject(s)
DNA Helicases/metabolism , Minute Virus of Mice/metabolism , Viral Nonstructural Proteins/metabolism , Base Sequence , DNA Primers , Enzyme Activation , HeLa Cells , Humans , Phosphorylation
15.
J Virol ; 71(8): 5733-41, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9223459

ABSTRACT

A novel human site-specific DNA-binding factor has been partially purified from extracts of HeLa S3 cells. This factor, designated PIF, for parvovirus initiation factor, binds to the minimal origin of DNA replication at the 3' end of the minute virus of mice (MVM) genome and functions as an essential cofactor in the replication initiation process. Here we show that PIF is required for the viral replicator protein NS1 to nick and become covalently attached to a specific site in the origin sequence in a reaction which requires ATP hydrolysis. DNase I and copper ortho-phenanthroline degradation of the PIF-DNA complexes showed that PIF protects a stretch of some 20 nucleotides, covering the entire region in the minimal left-end origin not already known to be occupied by NS1. Methylation and carboxy-ethylation interference analysis identified two ACGT motifs, spaced by five nucleotides, as the sequences responsible for this binding. A series of mutant oligonucleotides was then used as competitive inhibitors in gel mobility shift assays to confirm that PIF recognizes both of these ACGT sequences and to demonstrate that the two motifs comprise a single binding site rather than two separate sites. Competitive inhibition of the origin nicking assay, using the same group of oligonucleotides, confirmed that the same cellular factor is responsible for both mobility shift and nicking activities. UV cross-linking and relative mobility assays suggest that PIF binds DNA as a heterodimer or higher-order multimer with subunits in the 80- to 100-kDa range.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Minute Virus of Mice/genetics , Animals , Base Sequence , Binding Sites , HeLa Cells , Humans , Molecular Sequence Data , Molecular Weight , Oligonucleotides/metabolism , Spodoptera , Viral Nonstructural Proteins/physiology
16.
Virology ; 231(2): 267-80, 1997 May 12.
Article in English | MEDLINE | ID: mdl-9168889

ABSTRACT

Mutants of minute virus of mice (MVM) which express truncated forms of the NS2 polypeptide are known to exhibit a host range defect, replicating productively in transformed human cells but not in cells from their normal murine host. To explore this deficiency we generated viruses with translation termination codons at various positions in the second exon of NS2. In human cells these mutants were viable, but showed a late defect in progeny virion release which put them at a selective disadvantage compared to the wildtype. In murine cells, however, duplex viral DNA amplification was reduced to 5% of wildtype levels and single-strand DNA synthesis was undetectable. These deficiencies could not be attributed to a failure to initiate infection or to a generalized defect in viral gene expression, since the viral replicator protein NS1 was expressed to normal or elevated levels early in infection. In contrast, truncated NS2 gene products failed to accumulate, so that each mutant exhibited a similar NS2-null phenotype. Expression of the capsid polypeptides VP1 and VP2 and their subsequent assembly into intact particles were examined in detail. Synchronized infected cell populations labeled under pulse-chase conditions were analyzed by differential immunoprecipitation of native or denatured extracts using antibodies which discriminated between intact particles and isolated polypeptide chains. These analyses showed that at early times in infection, capsid protein synthesis and stability were normal, but particle assembly was impaired. Unassembled VP proteins were retained in the cell for several hours, but as the unprocessed material accumulated, capsid protein synthesis progressively diminished, so that at later times relatively few VP molecules were synthesized. Thus in NS2-null infections of mouse cells there is a major primary defect in the folding or assembly processes required for effective capsid production.


Subject(s)
Capsid/ultrastructure , Minute Virus of Mice/growth & development , Viral Nonstructural Proteins/physiology , Amino Acid Sequence , Animals , Antigens, Viral/chemistry , Base Sequence , DNA, Viral/biosynthesis , DNA, Viral/genetics , Humans , Mice , Molecular Sequence Data , Morphogenesis , Mutagenesis, Site-Directed , Species Specificity , Transfection , Virus Replication
17.
J Virol ; 71(2): 1405-16, 1997 Feb.
Article in English | MEDLINE | ID: mdl-8995666

ABSTRACT

Replication of linear single-stranded parvovirus DNA proceeds by a rolling-hairpin mechanism which generates long, palindromic, duplex concatamers. Processing to monomer length requires initiation from origins of DNA replication located at the 3' and 5' ends of each embedded monomer, reactions which can be recapitulated in vitro for minute virus of mice (MVM). To determine which cellular proteins were essential for replication from these origins, S100 extracts from 293S cells were fractionated on phosphocellulose. When recombined, these fractions were able to support replication in vitro, dependent on the viral initiator protein NS1, using plasmid forms of the 5' origin or the minimal 3' origin as templates. Fraction P-cell 1 contains two factors, replication protein A (RPA) and proliferating-cell nuclear antigen (PCNA), known to be essential for simian virus 40 replication in vitro. When P-cell 1 was replaced with purified recombinant RPA and PCNA, NS1-mediated MVM replication initiated from the 5' origin but not from the 3' origin. The 3' origin is a 50-bp sequence containing three distinct recognition elements, an NS1 binding site, a site at which NS1 nicks the DNA to generate the priming 3' OH, and a region containing a consensus activated transcription factor (ATF) binding site. To identify the missing factor(s) for 3' origin replication, P-cell 1 was fractionated by further chromatography and active fractions were identified by their ability to complement RPA, PCNA, and P-cell 2 for NS1-mediated, origin-specific replication. Gel shift and UV cross-linking analysis of the replication-competent fractions revealed a novel 110-kDa sequence-specific DNA binding protein which recognized the consensus ATF binding site region of the origin and which we have termed parvovirus initiation factor, or PIF. Binding of PIF appears to activate the endonuclease function of NS1, allowing efficient and specific nicking of the 3' minimal origin under stringent conditions in vitro.


Subject(s)
DNA, Viral/genetics , DNA-Binding Proteins/genetics , Parvovirus/physiology , Viral Nonstructural Proteins/genetics , Virus Replication , Animals , Base Sequence , Cell Line , DNA Replication , DNA-Binding Proteins/isolation & purification , Gene Expression Regulation, Viral , Humans , Mice , Molecular Sequence Data
18.
Acta Crystallogr D Biol Crystallogr ; 53(Pt 1): 93-102, 1997 Jan 01.
Article in English | MEDLINE | ID: mdl-15299974

ABSTRACT

The three-dimensional crystal structure of the single-stranded DNA-containing ('full') parvovirus, minute virus of mice (MVM), has been determined to 3.5 A resolution. Both full and empty particles of MVM were crystallized in the monoclinic space group C2 with cell dimensions of a = 448.7, b = 416.7, c = 305.3 A and beta = 95.8 degrees. Diffraction data were collected at the Cornell High Energy Synchrotron Source using an oscillation camera. The crystals have a pseudo higher R32 space group in which the particles are situated at two special positions with 32 point symmetry, separated by (1/2)c in the hexagonal setting. The self-rotation function showed that the particles are rotated with respect to each other by 60 degrees around the pseudo threefold axis. Subsequently, a more detailed analysis of the structure amplitudes demonstrated that the correct space-group symmetry is C2 as given above. Only one of the three twofold axes perpendicular to the threefold axis in the pseudo R32 space group is a 'true' crystallographic twofold axis; the other two are only 'local' non-crystallographic symmetry axes. The known canine parvovirus (CPV) structure was used as a phasing model to initiate real-space electron-density averaging phase improvement. The electron density was easily interpretable and clearly showed the amino-acid differences between MVM and CPV, although the final overall correlation coefficient was only 0.63. The structure of MVM has a large amount of icosahedrally ordered DNA, amounting to 22% of the viral genome, which is significantly more than that found in CPV.

19.
J Virol ; 69(9): 5422-30, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7636987

ABSTRACT

The NS1 polypeptide of minute virus of mice (MVM) is a potent transcriptional activator of the MVM P38 promoter. The minimum region of this promoter required for transactivation has been identified and termed the transactivation region (tar). However, the function of tar and the biochemical steps involved in NS1-mediated transactivation are not well understood. Here we provide evidence that NS1 binds directly and specifically to tar in a strictly ATP-dependent manner. A DNA fragment containing tar was specifically coimmunoprecipitated with purified baculovirus-expressed MVM NS1, using antibodies directed against NS1 amino- or carboxy-terminal peptides. Using this immunoprecipitation assay, we found that the NS1-tar interaction was enhanced approximately 10-fold by ATP, but subsequent incubation at elevated temperatures in the presence, but not the absence, of MgCl2 caused rapid loss of tar binding. This finding suggests that the tar-NS1 complex has a short half-life under assay conditions which favor ATP hydrolysis. Specific binding was efficiently inhibited by self-ligated oligonucleotides containing the core DNA sequence (ACCA)3, but the same nonligated 20- and 21-mer oligonucleotides were unable to compete effectively, indicating that NS1 only binds to its cognate site when this site is presented on DNA fragments of sufficient size. DNase I footprinting experiments performed in the presence of gamma S-ATP revealed that NS1 protects a 43-bp sequence extending asymmetrically from the (ACCA)2 sequence toward the TATA box of the promoter. NS1 footprints obtained at other sites in the MVM genome were similarly large and asymmetric, all extending approximately 31 bp 5' from the core (ACCA)2-3 sequence. Surprisingly, no footprints were obtained in the absence of gamma S-ATP even under low-stringency binding conditions. However, ATP could be omitted from the reactions if NS1 was first incubated with antibodies directed against its 16-amino-acid carboxy-terminal peptide. Since these antibodies probably create intermolecular cross-links, this finding suggests that NS1 may only bind its cognate site efficiently, or perhaps at all, if the transactivator is first induced to form oligomers. From these data, we hypothesize that ATP binding may also induce NS1 to oligomerize and that such assembly is required before the protein can bind effectively to the tar sequence. The functional implications of the NS1-tar interaction will be discussed.


Subject(s)
Minute Virus of Mice/genetics , Minute Virus of Mice/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Viral Nonstructural Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , DNA, Viral/chemistry , DNA, Viral/metabolism , Deoxyribonuclease I , Genes, Viral , Genetic Vectors , Genome, Viral , Molecular Sequence Data , Nucleopolyhedroviruses , Plasmids , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spodoptera , Transfection , Viral Nonstructural Proteins/biosynthesis , Viral Nonstructural Proteins/isolation & purification , Viral Proteins/biosynthesis , Viral Proteins/metabolism
20.
Virology ; 209(1): 122-35, 1995 May 10.
Article in English | MEDLINE | ID: mdl-7747462

ABSTRACT

Parvoviral DNA replication has many features in common with prokaryotic rolling circle replication (RCR), including the pivotal role of an initiator protein which introduces a site-specific, single strand nick into a duplex origin sequence. In this process, the protein becomes covalently attached to the new 5' end of the DNA, while making available a 3' hydroxyl to prime de novo synthesis. Sequence comparisons of prokaryotic RCR initiators has revealed a set of three common motifs, two of which, a putative metal coordination site and a downstream active-site tyrosine motif, could be tentatively identified in parvoviral replicator proteins. We have introduced mutations into the NS1 gene of the murine parvovirus minute virus of mice (MVM), in the putative metal coordination site at H129, and into the three candidate tyrosine motifs at Y188, Y197, and Y210. Histidine-tagged mutant proteins were expressed in HeLa cells from recombinant vaccinia virus vectors and partially purified. None of the mutant proteins were able to initiate replication of origin-containing plasmids in vitro, and each showed impaired site-specific binding to the viral origin, with Y188 and Y197 being most severely defective. If this deficiency was minimized using low salt conditions, however, Y188 and Y197 mutant proteins were able to nick and become covalently attached to origin DNA, whereas Y210 and H129 mutant proteins were not, suggesting that the latter residues are part of the catalytic site of the NS1 nickase. Transfer of [32P]phosphate from substrate DNA to NS1, followed by cyanogen bromide cleavage of the complex, gave the single, labeled peptide consistent with Y210 being the linking tyrosine.


Subject(s)
Minute Virus of Mice/genetics , Minute Virus of Mice/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Primers/genetics , DNA Replication , DNA, Viral/genetics , DNA, Viral/metabolism , HeLa Cells , Humans , Mice , Molecular Sequence Data , Mutation , Plasmids/genetics , Polymerase Chain Reaction , Replication Origin , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...