Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
J Virol ; 93(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31315994

ABSTRACT

As many tumor cells synthetize vascular endothelial growth factors (VEGF) that promote neo-vascularization and metastasis, frontline cancer therapies often administer anti-VEGF (α-VEGF) antibodies. To target the oncolytic parvovirus minute virus of mice (MVM) to the tumor vasculature, we studied the functional tolerance, evasion of neutralization, and induction of α-VEGF antibodies of chimeric viruses in which the footprint of a neutralizing monoclonal antibody within the 3-fold capsid spike was replaced by VEGF-blocking peptides: P6L (PQPRPL) and A7R (ATWLPPR). Both peptides allowed viral genome replication and nuclear translocation of chimeric capsid subunits. MVM-P6L efficiently propagated in culture, exposing the heterologous peptide on the capsid surface, and evaded neutralization by the anti-spike monoclonal antibody. In contrast, MVM-A7R yielded low infectious titers and was poorly recognized by an α-A7R monoclonal antibody. MVM-A7R showed a deficient assembly pattern, suggesting that A7R impaired a transitional configuration that the subunits must undergo in the 3-fold axis to close up the capsid shell. The MVM-A7R chimeric virus consistently evolved in culture into a mutant carrying the P6Q amino acid substitution within the A7R sequence, which restored normal capsid assembly and infectivity. Consistent with this finding, anti-native VEGF antibodies were induced in mice by a single injection of MVM-A7R empty capsids, but not by MVM-A7R virions. This fundamental study provides insights to endow an infectious parvovirus with immune antineovascularization and evasion capacities by replacing an antibody footprint in the capsid 3-fold axis with VEGF-blocking peptides, and it also illustrates the evolutionary capacity of single-stranded DNA (ssDNA) viruses to overcome engineered capsid structural restrictions.IMPORTANCE Targeting the VEGF signaling required for neovascularization by vaccination with chimeric capsids of oncolytic viruses may boost therapy for solid tumors. VEGF-blocking peptides (VEbp) engineered in the capsid 3-fold axis endowed the infectious parvovirus MVM with the ability to induce α-VEGF antibodies without adjuvant and to evade neutralization by MVM-specific antibodies. However, these properties may be compromised by structural restraints that the capsid imposes on the peptide configuration and by misassembly caused by the heterologous peptides. Significantly, chimeric MVM-VEbp resolved the structural restrictions by selecting mutations within the engineered peptides that restored efficient capsid assembly. These data show the promise of antineovascularization vaccines using chimeric VEbp-icosahedral capsids of oncolytic viruses but also raise safety concerns regarding the genetic stability of manipulated infectious parvoviruses in cancer and gene therapies.


Subject(s)
Cancer Vaccines/immunology , Capsid Proteins/immunology , Capsid Proteins/metabolism , Minute Virus of Mice/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/genetics , Capsid Proteins/genetics , Mice, Inbred BALB C , Minute Virus of Mice/genetics , Minute Virus of Mice/growth & development , Oncolytic Viruses/genetics , Oncolytic Viruses/growth & development , Oncolytic Viruses/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Load , Virus Assembly , Virus Attachment , Virus Internalization
2.
Vaccine ; 37(7): 1001-1005, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30642729

ABSTRACT

We used a combination of DOP-PCR with high throughput sequencing (HTS) to study infected cell cultures over time to assess the feasibility of using this technique to provide a read-out other than cytopathic effect in cell culture infectivity assays. Because DOP-PCR primers feature a short constant sequence at their 3' terminus, the procedure yields a reproducible representational library of products from a given PCR template, including viral nucleic acids. Using SV40- and MVM-infected cultures harvested at different times, we show that the number of viral matches among DOP-PCR products parallels the quantity of virus as shown by real-time PCR, and further show that HTS analysis of specific DOP-PCR products that increase in quantity over time could be used to identify the infecting virus with a sensitivity similar to that of typical cell-culture assays that rely on cytopathic effect.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Minute Virus of Mice/growth & development , Polymerase Chain Reaction/methods , Simian virus 40/growth & development , Virus Cultivation/methods , Animals , CHO Cells , Chlorocebus aethiops , Cricetulus , Minute Virus of Mice/genetics , Simian virus 40/genetics , Vero Cells , Viral Load
3.
J Virol ; 88(17): 10189-99, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24965470

ABSTRACT

UNLABELLED: The ATR kinase has essential functions in maintenance of genome integrity in response to replication stress. ATR is recruited to RPA-coated single-stranded DNA at DNA damage sites via its interacting partner, ATRIP, which binds to the large subunit of RPA. ATR activation typically leads to activation of the Chk1 kinase among other substrates. We show here that, together with a number of other DNA repair proteins, both ATR and its associated protein, ATRIP, were recruited to viral nuclear replication compartments (autonomous parvovirus-associated replication [APAR] bodies) during replication of the single-stranded parvovirus minute virus of mice (MVM). Chk1, however, was not activated during MVM infection even though viral genomes bearing bound RPA, normally a potent trigger of ATR activation, accumulate in APAR bodies. Failure to activate Chk1 in response to MVM infection was likely due to our observation that Rad9 failed to associate with chromatin at MVM APAR bodies. Additionally, early in infection, prior to the onset of the virus-induced DNA damage response (DDR), stalling of the replication of MVM genomes with hydroxyurea (HU) resulted in Chk1 phosphorylation in a virus dose-dependent manner. However, upon establishment of full viral replication, MVM infection prevented activation of Chk1 in response to HU and various other drug treatments. Finally, ATR phosphorylation became undetectable upon MVM infection, and although virus infection induced RPA32 phosphorylation on serine 33, an ATR-associated phosphorylation site, this phosphorylation event could not be prevented by ATR depletion or inhibition. Together our results suggest that MVM infection disables the ATR signaling pathway. IMPORTANCE: Upon infection, the parvovirus MVM activates a cellular DNA damage response that governs virus-induced cell cycle arrest and is required for efficient virus replication. ATM and ATR are major cellular kinases that coordinate the DNA damage response to diverse DNA damage stimuli. Although a significant amount has been discovered about ATM activation during parvovirus infection, involvement of the ATR pathway has been less studied. During MVM infection, Chk1, a major downstream target of ATR, is not detectably phosphorylated even though viral genomes bearing the bound cellular single-strand binding protein RPA, normally a potent trigger of ATR activation, accumulate in viral replication centers. ATR phosphorylation also became undetectable. In addition, upon establishment of full viral replication, MVM infection prevented activation of Chk1 in response to hydroxyurea and various other drug treatments. Our results suggest that MVM infection disables this important cellular signaling pathway.


Subject(s)
Host-Pathogen Interactions , Minute Virus of Mice/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Humans , Mice , Minute Virus of Mice/growth & development
4.
J Mol Biol ; 357(3): 1026-38, 2006 Mar 31.
Article in English | MEDLINE | ID: mdl-16469332

ABSTRACT

The connection between nuclear transport and morphogenesis of a large macromolecular entity has been investigated using the karyophylic capsid of the parvovirus minute virus of mice (MVM) as a model. The VP1 (82 kDa) and VP2 (63 kDa) proteins forming the T = 1 icosahedral MVM capsid at the respective 1:5 molar ratio of synthesis, could be covalently cross-linked with dimethyl suberimidate into two types of oligomeric assemblies, which were present at stoichiometric amounts in infected cell extracts and purified viral particles. The larger species contained VP1 and corresponded in size (200 kDa) to a heterotrimer of one VP1 and two VP2 subunits. The smaller species contained VP2 only and corresponded in size (180 kDa) to a homotrimer. The introduction of bulky residues or the truncation of side-chains involved in multiple interactions at the interfaces between trimers of VPs in the MVM capsid, produced the accumulation of trimeric intermediates that were competent in nuclear translocation but not in capsid assembly. These results indicate that MVM maturation proceeds by cytoplasmic oligomerization of the capsid subunits into two types of trimers, which are the assembly intermediates competent to translocate across the nuclear membrane. Consistent with this conclusion, mutations at basic residues that inactivate a previously identified beta-stranded nuclear localization motif, which notably are not involved in inter or intra-subunit contacts, led to cytoplasmic retention of the two types of trimers, with no evidence for other assembly intermediates. Although a fraction of the VP1-containing trimers were translocated into the nucleus driven by the conventional nuclear transport signal of VP1 N terminus, their further assembly in the absence of the VP2-only trimers yielded large molecular mass amorphous aggregates. Therefore, the nuclear transport stoichiometry of assembly intermediates may exert a morphogenetic quality control on macromolecular complexes like the MVM capsid.


Subject(s)
Capsid/metabolism , Cell Nucleus/metabolism , Minute Virus of Mice/physiology , Virus Assembly , Active Transport, Cell Nucleus/genetics , Capsid/chemistry , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line, Transformed , Cell Nucleus/genetics , Cross-Linking Reagents/metabolism , Humans , Minute Virus of Mice/genetics , Minute Virus of Mice/growth & development , Mutagenesis, Site-Directed , Nuclear Envelope/chemistry , Nuclear Envelope/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Virus Assembly/genetics
5.
J Virol ; 79(6): 3517-24, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15731246

ABSTRACT

Due to their oncolytic properties and apathogenicity, autonomous parvoviruses have attracted significant interest as possible anticancer agents. Recent preclinical studies provided evidence of the therapeutic potential of minute virus of mice prototype strain (MVMp) and its recombinant derivatives. In a murine model of hemangiosarcoma, positive therapeutic outcome correlated with high intratumoral expression of MVMp-encoded genes in tumors and lymphoid organs, especially in tumor-draining lymph nodes. The source and relevance of this extratumoral expression, which came as a surprise because of the known fibrotropism of MVMp, remained unclear. In the present study, we investigated (i) whether the observed expression pattern occurs in different tumor models, (ii) which cell population is targeted by the virus, and (iii) the immunological consequences of this infection. Significant MVMp gene expression was detected in lymphoid tissues from infected tumor-free as well as melanoma-, lymphoma-, and hemangiosarcoma-bearing mice. This expression was especially marked in lymph nodes draining virus-injected tumors. Fluorescent in situ hybridization analysis, multicolor fluorescence-activated cell sorting, and quantitative reverse transcription-PCR revealed that MVMp was expressed in rare subpopulations of CD11b (Mac1)-positive cells displaying CD11c+ (myeloid dendritic cells [MDC]) or CD45B (B220+ [B1 lymphocytes]) markers. Apart from the late deletion of cytotoxic memory cells (CD8+ CD44+ CD62L-), this infection did not lead to significant alteration of the immunological profile of cells populating lymphoid organs. However, subtle changes were detected in the production of specific proinflammatory cytokines in lymph nodes from virus-treated animals. Considering the role of B1 lymphocytes and MDC in cancer and immunological surveillance, the specific ability of these cell types to sustain parvovirus-driven gene expression may be exploited in gene therapy protocols.


Subject(s)
B-Lymphocytes/virology , Dendritic Cells/virology , Lymphoid Tissue/virology , Minute Virus of Mice/genetics , Parvoviridae Infections/virology , Animals , B-Lymphocytes/immunology , CD11b Antigen/analysis , CD11c Antigen/analysis , Dendritic Cells/immunology , Female , Gene Expression , Genes, Viral , Hemangiosarcoma/virology , Leukocyte Common Antigens/analysis , Lymph Nodes/immunology , Lymph Nodes/virology , Lymphocyte Subsets/immunology , Lymphoid Tissue/immunology , Lymphoma/virology , Melanoma, Experimental/virology , Mice , Minute Virus of Mice/growth & development , Parvoviridae Infections/immunology , Spleen/immunology , Spleen/virology , Transcription, Genetic
6.
J Virol ; 78(19): 10685-94, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15367635

ABSTRACT

It is uncertain whether nonenveloped karyophilic virus particles may actively traffic from the nucleus outward. The unordered amino-terminal domain of the VP2 major structural protein (2Nt) of the icosahedral parvovirus minute virus of mice (MVM) is internal in empty capsids, but it is exposed outside of the shell through the fivefold axis of symmetry in virions with an encapsidated single-stranded DNA genome, as well as in empty capsids subjected to a heat-induced structural transition. In productive infections of transformed and normal fibroblasts, mature MVM virions were found to efficiently exit from the nucleus prior to cell lysis, in contrast to the extended nuclear accumulation of empty capsids. Newly formed mutant viruses lacking the three phosphorylated serine residues of 2Nt were hampered in their exit from the human transformed NB324K nucleus, in correspondence with the capacity of 2Nt to drive microinjected phosphorylated heated capsids out of the nucleus. However, in normal mouse A9 fibroblasts, in which the MVM capsid was phosphorylated at similar sites but with a much lower rate, the nuclear exit of virions and microinjected capsids harboring exposed 2Nt required the infection process and was highly sensitive to inhibition of the exportin CRM1 in the absence of a demonstrable interaction. Thus, the MVM virion exits the nucleus by accessing nonconventional export pathways relying on cell physiology that can be intensified by infection but in which the exposure of 2Nt remains essential for transport. The flexible 2Nt nuclear transport signal may illustrate a common structural solution used by nonenveloped spherical viruses to propagate in undamaged host tissues.


Subject(s)
Capsid Proteins/metabolism , Cell Nucleus/virology , Minute Virus of Mice/metabolism , Receptors, Cytoplasmic and Nuclear , Active Transport, Cell Nucleus , Animals , Capsid Proteins/chemistry , Cell Line , Cell Nucleus/metabolism , Cell Transformation, Viral , Humans , Karyopherins/metabolism , Mice , Minute Virus of Mice/growth & development , Phosphorylation , Virion/metabolism , Exportin 1 Protein
7.
Biologicals ; 31(3): 181-5, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12935806

ABSTRACT

Treatment with steam and/or dilute NaOH are commonly used techniques to disinfect manufacturing vessels and tools in the pharmaceutical industry. The aim of this procedure is sanitisation and inactivation of microbiological and viral contaminants. Here we describe the inactivation of the mouse parvovirus Minute Virus of Mice (MVM) under these conditions. Parvoviruses are known to be resistant to physico-chemical treatment and one representative of this family, the human parvovirus B19, is a potential contaminant of blood plasma. We show inactivation kinetics for MVM treated with wet-heat (70, 80, 90 degrees C) and with 0.01-1 M NaOH solutions (pH >/=11.9). Robust inactivation was only achieved at 90 degrees C for at least 10 min and in NaOH solutions of pH >/=12.8 (0.1 M NaOH). It was observed, that aggregation of viruses might protect viral particles from inactivation by NaOH. Therefore, appropriate sample preparation of spiking material is important for accurate simulation of the naturally occurring situation. The observed stability at pH 11.8 exceeds the previously reported upper limit of pH 9. Inactivation was due to disintegration of the viral capsid as assessed by accessibility of viral DNA for endonucleases.


Subject(s)
Hot Temperature , Minute Virus of Mice/physiology , Sodium Hydroxide/pharmacology , Virus Inactivation , Animals , Capsid/metabolism , Cell Line , DNA, Viral/metabolism , Humans , Hydrogen-Ion Concentration , Kinetics , Mice , Minute Virus of Mice/drug effects , Minute Virus of Mice/growth & development , Minute Virus of Mice/isolation & purification , Polymerase Chain Reaction
8.
Virology ; 306(2): 268-79, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12642100

ABSTRACT

Activation of the minute virus of mice (MVM) P4 promoter is a key step in the life cycle of the virus and is completely dependent on host transcription factors. Since transcription-factor composition varies widely in different cell types, there is the possibility that only some cell types in the host organism have the capacity to initiate expression from the P4 promoter and therefore that the promoter may be a factor in determining the tropism of MVM. In this study, the ability of various cell types to activate P4, independent of the other virus-host interactions, was examined in transgenic mouse lines bearing a beta-galactosidase reporter sequence driven by the P4 promoter. It was found that lacZ was expressed during embryogenesis and in the adult in a cell-type-specific and differentiation-dependent pattern. The data are consistent with cell-type and stage-specific activation of the P4 promoter having a role in determining the host cell-type range of MVM. The ability of some parvoviruses to replicate in, and kill oncogenically transformed cells, and to destroy induced tumors in laboratory animals is the basis of recent approaches to use MVM-based vectors in cancer gene therapy. Since these vectors rely on the activation of the P4 promoter by the target tissues, understanding the promoter dependence on cell-type and differentiation status is important for their design and potential use.


Subject(s)
Genes, Viral , Minute Virus of Mice/genetics , Promoter Regions, Genetic , Animals , Cell Line , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Viral , Genes, Reporter , Genetic Vectors , Lac Operon , Mice , Mice, Inbred BALB C , Mice, Transgenic , Minute Virus of Mice/growth & development , Minute Virus of Mice/pathogenicity , Minute Virus of Mice/physiology , Parvoviridae Infections/virology , Pregnancy , Virulence , Virus Replication , beta-Galactosidase/genetics
9.
Antiviral Res ; 52(1): 33-41, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11530186

ABSTRACT

Virus inactivation by ethyleneimines was first introduced more than 30 years ago. Selective targeting of nucleic acids was reported for oligomeric ethyleneimines. In this study, trimeric ethyleneimine (TEI) was used to inactivate minute virus of mice (MVM; Parvoviridae) and Semliki forest virus (SFV; Togaviridae). The pH-dependency of the inactivation kinetics observed with MVM was different compared to the kinetics reported for other viruses. The higher inactivation rate at higher pH favoured the idea of a mechanism involving protein modifications. Alteration of the isoelectric point and changes in mass could be observed after treatment of soluble proteins with TEI. The uptake of MVM by host cells was reduced or completely blocked by TEI treatment, as shown by monitoring viral internalisation of DNA into target cells. The observed loss of virus infectivity coincided with the inhibition of virus uptake. Thus, virus inactivation by TEI is most likely also a result of chemical modifications of viral surface proteins.


Subject(s)
Aziridines/pharmacology , Minute Virus of Mice/drug effects , Semliki forest virus/drug effects , Aedes , Animals , Capsid/drug effects , Cells, Cultured , Chlorocebus aethiops , Cytopathogenic Effect, Viral , DNA, Viral/drug effects , Hydrogen-Ion Concentration , Isoelectric Point , Kinetics , Mice , Minute Virus of Mice/growth & development , Myoglobin/metabolism , Ovalbumin/metabolism , Protein Processing, Post-Translational/drug effects , Semliki forest virus/growth & development , Spectrometry, Mass, Electrospray Ionization , Time Factors , Vero Cells , Viral Envelope Proteins/drug effects , Virus Latency/drug effects
10.
J Virol ; 73(5): 3877-85, 1999 May.
Article in English | MEDLINE | ID: mdl-10196282

ABSTRACT

Autonomous parvoviruses are tightly dependent on host cell factors for various steps of their life cycle. In particular, DNA replication and gene expression of the prototype strain of the minute virus of mice (MVMp) are closely linked to the onset of host cell DNA replication, pointing to the involvement of an S-phase-specific cellular factor(s) in parvovirus multiplication. The viral nonstructural protein NS-1 is absolutely required for parvovirus DNA replication and is able to transcriptionally regulate parvoviral and heterologous promoters. We previously showed that the promoter P4, which directs the transcription unit encoding the NS proteins, is activated at the onset of S phase. This activation is dependent on an E2F motif in the proximal region of promoter P4. An infectious MVM DNA clone was mutated in the E2F motif of P4. The wild type and the E2F mutant derivative were tested for their ability to produce progeny viruses after transfection of permissive cells. In the context of the whole MVMp genome, the E2F mutation abolished P4 induction in S phase and inactivated the infectious molecular clone, which failed to become amplified and generate progeny particles. The virus could be rescued when NS proteins were supplied in trans, showing that P4 hyperactivity in S is needed to reach a level of NS-1 expression that is sufficient to drive the viral replication cycle. These data show that E2F-mediated P4 activation at the early S phase is a limiting factor for parvovirus production. The primary barrier to parvovirus gene expression in G1 is thought to be promoter formation rather than activation, due to the poor conversion of the parental single-strand genome to a duplex form. The S dependence of P4 activation may therefore be a sign of the virus adaptation to life in the S-phase host cell. If the conversion block in G1 were to be leaky, the S induction of promoter P4 could be envisioned as a safeguard against the production of toxic NS proteins until cells reach the S phase and provide the full machinery for parvovirus replication.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Gene Expression Regulation, Viral , Minute Virus of Mice/genetics , Promoter Regions, Genetic , Viral Nonstructural Proteins/genetics , Animals , Binding Sites , Cell Line , DNA Replication , DNA, Viral , E2F Transcription Factors , G1 Phase , Genetic Complementation Test , Humans , Mice , Minute Virus of Mice/growth & development , Minute Virus of Mice/physiology , Plasmids , Retinoblastoma-Binding Protein 1 , S Phase , Transcription Factor DP1 , Transcription Factors/metabolism , Virus Replication
11.
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
12.
J Virol ; 70(11): 8109-16, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8892936

ABSTRACT

Newborn BALB/c mice intranasally inoculated at birth with a lethal dose of the immunosuppressive strain of the parvovirus minute virus of mice (MVMi) developed motor disabilities and intention tremors with a high incidence by the day 6 postinfection (dpi). These neurological syndromes paralleled the synthesis of virus intermediate DNA replicative forms and yield of infectious particles in the brain, with kinetics that peaked by this time. The preferred virus replicative sites in the brain were established early in the infection (2 dpi) and at the onset of clinical symptoms (6 dpi) and were compared with major regions of cellular proliferative activity found after intraperitoneal injection of bromodeoxyuridine 24 h before encephalons were subjected to immunohistochemistry detection. At 2 dpi, viral capsid antigen was located in the laterodorsal thalamic and the pontine nuclei but not in the extensive proliferative regions of the mouse brain at this postnatal day. At 6 dpi, however, the neurotropism of the MVMi was highlighted by its ability to target the subventricular zone of the ventricles, the subependymal zone of the olfactory bulb, and the dentate gyrus of the hippocampus, which are the three main germinal centers of the cerebrum in mouse postbirth neurogenesis. Unexpectedly, in the cerebellum, the MVMi capsid antigen was confined exclusively to cells that have undergone mitosis and have migrated to the internal granular layer (IGL) and not to the proliferative external granular layer (EGL), which was stained with antiproliferative cell nuclear antigen antibody and is the main target in other parvovirus infections. This result implies temporal or differentiation coupling between MVMi cycle and neuroblast morphogenesis, since proliferative granules of the EGL should primarily be infected but must migrate in a virus carrier state into the IGL in order to express the capsid proteins. During migration, many cells undergo destruction, accounting for the marked hypocellularity specifically found in the IGL and the irregular alignment of Purkinje cell bodies, both consistent histopathological hallmarks of animals developing cerebellar symptoms. We conclude that MVMi impairs postmitotic neuronal migration occurring in the first postnatal week, when, through the natural respiratory route of infection, the virus titer peaks in the encephalon. The results illustrate the intimate connection between MVMi neuropathogenesis and mouse brain morphogenetic stage, underscoring the potential of parvoviruses as markers of host developmental programs.


Subject(s)
Brain/virology , Minute Virus of Mice/metabolism , Neurons/virology , Parvoviridae Infections/virology , Animals , Animals, Newborn , Brain/pathology , Capsid/metabolism , Cell Division , Cell Line, Transformed , Central Nervous System/virology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Minute Virus of Mice/growth & development , Minute Virus of Mice/pathogenicity , Parvoviridae Infections/pathology , Parvoviridae Infections/physiopathology , Tumor Cells, Cultured
13.
J Med Virol ; 49(1): 1-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8732864

ABSTRACT

The transmission of hepatitis A virus (HAV) associated with use of FVIII concentrates has been reported in a number of European countries. All of these cases were associated with products inactivated by use of solvent detergent treatment. These reports have emphasized the necessity of evaluating virus inactivation methodologies for their ability to inactivate HAV. Such studies had previously been hampered by the difficulties associated with titration of HAV, because of the minimal cytopathic effect of most strains of virus on tissue culture cells. We have developed a simple, rapid, TCID50 virus titration system using a cytopathic strain of HAV which allows extensive kinetic studies of HAV inactivation. This has been compared with the standard radioimmunofocus forming (RFF) assay which is presently used for HAV titration. The reproducibility of the TCID50 assay was demonstrated to be equal to that of the RFF assay and the 95% confidence intervals for titres determined by both assays were also equal. The thermal stability of the cytopathic strain was studied and shown to be equivalent to that of a noncytopathic strain. The kinetics of HAV inactivation by heating in aqueous solution were compared to those of HIV-1 and a number of model viruses. It was demonstrated that HAV was highly stable, with 5 hours heat treatment at 60 degrees C in aqueous solution being required to inactivate 5.8 log10 virus. In contrast to heating in aqueous solution, lyophilization followed by 1 hour vapor heating at 60 degrees C was sufficient to inactivate 5.9 log10 HAV.


Subject(s)
Hepatovirus/growth & development , Animals , Antithrombin III , Blood Substitutes , Cell Line , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Encephalitis Viruses, Tick-Borne/growth & development , HIV-1/growth & development , Herpesvirus 1, Suid/growth & development , Hot Temperature , Humans , Kinetics , Minute Virus of Mice/growth & development , Titrimetry , Tumor Cells, Cultured , Viral Plaque Assay
15.
J Virol ; 68(5): 2840-8, 1994 May.
Article in English | MEDLINE | ID: mdl-8151755

ABSTRACT

Previous genetic analysis of the DNA replication of minute virus of mice (MVM) minigenomes suggested that specific elements, A (nucleotides [nt] 4489 to 4636) and B (nt 4636 to 4695), found inboard of the 5' palindrome are required for efficient MVM DNA replication (P. Tam and C. R. Astell, Virology 193:812-824, 1993). In this report, we show that two MVM RsaI restriction fragments (RsaI A [nt 4431 to 4579] and RsaI B [nt 4579 to 4662]) are able to activate DNA replication of an MVM minigenome containing deletions of both elements A and B. We also show that sequences inboard of the right palindrome are able to activate replication of minigenomes containing two left termini. In order to investigate the importance of the RsaI fragments, we demonstrate the presence of a number of sequence-specific DNA-protein interactions by electrophoretic mobility shift assays. After partial fractionation of A9 nuclear extracts, DNase I footprinting analysis was used to determine the binding sites for MVM replication factor (MRF) B5. MRF B5 protects two distinct regions (sites I and II) of the RsaI B probe from DNase I digestion. Competition f electrophoretic mobility shift assays with synthetic oligonucleotides corresponding to sites I and II suggest that MRF B5 is composed of two factors, MRF B3 and MRF B4, which bind DNA independently in a sequence-specific manner. It may be possible that these replication factors are proteins which are able to transactivate MVM DNA replication and hence are accessory replication factors.


Subject(s)
DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Minute Virus of Mice/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Base Sequence , Cell Nucleus/chemistry , DNA Replication , DNA, Viral/drug effects , Deoxyribonuclease I/pharmacology , Mice , Minute Virus of Mice/growth & development , Molecular Sequence Data , Protein Binding , Subcellular Fractions/chemistry , Virus Replication
16.
J Virol ; 68(3): 1397-406, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8107203

ABSTRACT

In this work, we report the transduction of a chloramphenicol acetyltransferase (CAT) reporter gene into a variety of normal and transformed human cells of various tissue origins. The vector used was MVM/P38cat, a recombinant of the prototype strain of the autonomous parvovirus minute virus of mice (MVMp). The CAT gene was inserted into the capsid-encoding region of the infectious molecular clone of MVMp genome, under the control of the MVM P38 promoter. When used to transfect permissive cells, the MVM/P38cat DNA was efficiently replicated and expressed the foreign CAT gene at high levels. By cotransfecting with a helper plasmid expressing the capsid proteins, it was possible to produce mixed virus stocks containing MVM/P38cat infectious particles and variable amounts of recombinant MVM. MVM/P38cat viral particles were successfully used to transfer the CAT gene and to express it in a variety of human cells. Both viral DNA replication and P38-driven CAT expression were achieved in fibroblasts, epithelial cells, T lymphocytes, and macrophages in a transformation-dependent way, but with an efficiency depending on the cell type. In transformed B lymphocytes, however, the vector was not replicated, nor did it express the CAT gene.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/genetics , Minute Virus of Mice/genetics , Animals , Cell Line, Transformed , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , DNA Replication , Epithelial Cells , Epithelium/microbiology , Fibroblasts/cytology , Fibroblasts/microbiology , Humans , Macrophages/cytology , Macrophages/microbiology , Minute Virus of Mice/growth & development , T-Lymphocytes/cytology , T-Lymphocytes/microbiology , Transduction, Genetic , Transfection , Virus Replication
17.
Virology ; 197(2): 630-41, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8249286

ABSTRACT

The nonstructural protein NS-1 of minute virus of mice (MVMp), an autonomous parvovirus, trans-inhibits the replication of a chimeric plasmid containing the SV40 origin of replication (ori) embedded in the MVMp genome. It appears that a 157-bp 5' proximal sequence of MVMp DNA is sufficient, in the presence of NS-1, to cause the inhibition of DNA replication driven by the SV40 ori placed on the same molecule. This effect is not dependent on the orientation of the MVMp target sequence and results from both a reduced level of utilization of SV40 ori and the blockage of progressing replication forks at the level of the target. Furthermore, replication driven by Epstein-Barr virus origin (oriP) is trans-inhibited by MVMp but this inhibition does not require the presence of parvoviral sequences in cis. On the basis of sequence homologies between EBV oriP and MVMp 5' terminal sequence, it is proposed that the direct or indirect interaction of NS-1 with parvovirus-like sequences present in heterologous viral and possibly also cellular genomes may result in an inhibition of DNA replication.


Subject(s)
DNA Replication/drug effects , DNA, Viral/biosynthesis , Minute Virus of Mice/genetics , Regulatory Sequences, Nucleic Acid/genetics , Viral Nonstructural Proteins/pharmacology , Animals , Base Sequence , Cell Line , DNA, Recombinant/biosynthesis , DNA, Viral/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Humans , Minute Virus of Mice/growth & development , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Simian virus 40/genetics , Simian virus 40/growth & development
18.
Virology ; 197(2): 770-3, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8249299

ABSTRACT

To investigate parvoviral interference with human immunodeficiency virus type 1 (HIV-1) in human cells that are normally susceptible to HIV-1 infection, nonstructural (NS) proteins of the parvoviruses H-1 virus and minute virus of mice were studied for their effect on the activity of the HIV-1 promoter in a variety of CD4+ cells. Transient cotransfection assays revealed a reduced HIV-1 promoter activity in the presence of parvoviral NS proteins. Stimulation of the HIV-1 promoter by phorbol 12-myristate 13-acetate (PMA) led to an increase in its sensitivity to NS-induced suppression. The inhibitory effect of NS polypeptides depended, at least in part, on the presence of the NF kappa B motifs of the HIV-1 long terminal repeat, suggesting an interaction of the parvoviral products with PMA-inducible cellular factors binding to these elements of the HIV-1 promoter.


Subject(s)
Gene Expression Regulation, Viral/drug effects , HIV-1/growth & development , Parvovirus/growth & development , Viral Interference/genetics , Viral Nonstructural Proteins/pharmacology , Animals , Cells, Cultured , Genes, Reporter , HIV Long Terminal Repeat/genetics , HIV-1/genetics , Humans , Luciferases/biosynthesis , Luciferases/genetics , Minute Virus of Mice/genetics , Minute Virus of Mice/growth & development , NF-kappa B/metabolism , Parvovirus/genetics , Promoter Regions, Genetic/genetics , Rats , Regulatory Sequences, Nucleic Acid/genetics , Suppression, Genetic , Tetradecanoylphorbol Acetate/pharmacology
19.
J Gen Virol ; 74 ( Pt 6): 1175-9, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8509766

ABSTRACT

We previously constructed a recombinant LuIII parvovirus genome lacking viral coding sequences and used it to generate luciferase-transducing virions, by cotransfection of cells with a helper plasmid expressing LuIII viral proteins. Here, we describe similar cotransfections using alternative, replication-defective helpers encoding the non-structural and capsid proteins of parvovirus H1, or of either the fibrotropic or lymphotropic parvovirus strain of minute virus of mice [MVM(p) or MVM(i)]. Each cotransfection generated transducing virus which directed luciferase expression after infection of HeLa cells. The transducing activity of virus produced using either LuIII or H1 helper plasmids could be specifically neutralized by antiserum raised against the corresponding infectious virus. When the recombinant LuIII parvovirus was pseudotyped with MVM(p) or MVM(i), the resulting virions efficiently expressed luciferase after infection in human or murine cells known to be permissive for both MVM strains. The MVM(p) pseudotyped virus also expressed this reporter efficiently when infected into the murine A9 fibroblast line. In contrast, the recombinant virus generated with an MVM(i) helper gave luciferase expression that was barely detectable after infection of A9 cells which are highly restrictive for MVM(i) productive infection. These results support the notion that the allotropic determinant of these MVM strains functions through their capsid proteins. Pseudotyping of recombinant parvovirus genomes should be useful in controlling their host range as vectors, and in studying mechanisms influencing the permissiveness of parvovirus infections.


Subject(s)
DNA, Viral/genetics , Defective Viruses/growth & development , Helper Viruses/growth & development , Minute Virus of Mice/growth & development , Parvoviridae/growth & development , Capsid/metabolism , Cells, Cultured , DNA, Recombinant/genetics , Defective Viruses/genetics , Genome, Viral , Helper Viruses/genetics , Luciferases/biosynthesis , Luciferases/genetics , Minute Virus of Mice/genetics , Parvoviridae/genetics , Recombinant Proteins/biosynthesis , Species Specificity , Tissue Distribution , Transfection , Virus Replication
20.
J Virol ; 67(1): 131-41, 1993 Jan.
Article in English | MEDLINE | ID: mdl-8416366

ABSTRACT

The two capsid proteins of minute virus of mice, VP1 and VP2, are generated from a single large open reading frame by alternate splicing of the capsid gene mRNA. Examination of the replication of a series of mutants that express only VP1, only VP2, or neither capsid protein demonstrates that VP2 is necessary for the accumulation and encapsidation of virus progeny single-stranded DNA. VP1 is dispensable for these functions but is required to produce an infectious virion. Virus that lacks VP1 binds to cells as efficiently as wild-type minute virus of mice but fails to initiate a productive infection. Because neither capsid protein is required for viral-DNA replication, these results suggest that virus lacking VP1 is blocked at a step during virus entry, subsequent to cell binding and prior to the initiation of DNA replication.


Subject(s)
Capsid/genetics , Minute Virus of Mice/pathogenicity , Amino Acid Sequence , Animals , Biological Transport , Capsid/biosynthesis , Capsid/ultrastructure , Capsid Proteins , Cell Line , DNA Mutational Analysis , DNA, Single-Stranded/metabolism , DNA, Viral/metabolism , Minute Virus of Mice/genetics , Minute Virus of Mice/growth & development , Molecular Sequence Data , Virion/genetics , Virion/growth & development , Virion/pathogenicity , Virulence , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...