Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Arch Virol ; 165(9): 2133-2146, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32533329

ABSTRACT

Parvoviridae, a diverse family of small single-stranded DNA viruses was established in 1975. It was divided into two subfamilies, Parvovirinae and Densovirinae, in 1993 to accommodate parvoviruses that infect vertebrate and invertebrate animals, respectively. This relatively straightforward segregation, using host association as the prime criterion for subfamily-level classification, has recently been challenged by the discovery of divergent, vertebrate-infecting parvoviruses, dubbed "chapparvoviruses", which have proven to be more closely related to viruses in certain Densovirinae genera than to members of the Parvovirinae. Viruses belonging to these genera, namely Brevi-, Hepan- and Penstyldensovirus, are responsible for the unmatched heterogeneity of the subfamily Densovirinae when compared to the Parvovirinae in matters of genome organization, protein sequence homology, and phylogeny. Another genus of Densovirinae, Ambidensovirus, has challenged traditional parvovirus classification, as it includes all newly discovered densoviruses with an ambisense genome organization, which introduces genus-level paraphyly. Lastly, current taxon definition and virus inclusion criteria have significantly limited the classification of certain long-discovered parvoviruses and impedes the classification of some potential family members discovered using high-throughput sequencing methods. Here, we present a new and updated system for parvovirus classification, which includes the introduction of a third subfamily, Hamaparvovirinae, resolves the paraphyly within genus Ambidensovirus, and introduces new genera and species into the subfamily Parvovirinae. These proposals were accepted by the ICTV in 2020 March.


Subject(s)
Parvoviridae Infections/veterinary , Parvoviridae Infections/virology , Parvoviridae/classification , Parvoviridae/physiology , Phylogeny , Animals , Host Specificity , Humans , Parvoviridae/genetics , Parvoviridae/isolation & purification , Viral Proteins/genetics
2.
Viruses ; 11(5)2019 05 04.
Article in English | MEDLINE | ID: mdl-31060205

ABSTRACT

Rodent protoparvoviruses (PVs), parvovirus H-1 (H-1PV) in particular, are naturally endowed with oncolytic properties. While being historically described as agents that selectively replicate in and kill cancer cells, recent yet growing evidence demonstrates that these viruses are able to reverse tumor-driven immune suppression through induction of immunogenic tumor cell death, and the establishment of antitumorigenic, proinflammatory milieu within the tumor microenvironment. This review summarizes the most important preclinical proofs of the interplay and the cooperation between PVs and the host immune system. The molecular mechanisms of PV-induced immunostimulation are also discussed. Furthermore, initial encouraging in-human observations from clinical trials and compassionate virus uses are presented, and speak in favor of further H-1PV clinical development as partner drug in combined immunotherapeutic protocols.


Subject(s)
Neoplasms/virology , Oncolytic Viruses/physiology , Parvoviridae/physiology , Animals , Humans , Immune System , Neoplasms/immunology , Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Parvoviridae/genetics
3.
Viruses ; 6(7): 2899-937, 2014 Jul 23.
Article in English | MEDLINE | ID: mdl-25055856

ABSTRACT

To get access to the replication site, small non-enveloped DNA viruses have to cross the cell membrane using a limited number of capsid proteins, which also protect the viral genome in the extracellular environment. Most of DNA viruses have to reach the nucleus to replicate. The capsid proteins involved in transmembrane penetration are exposed or released during endosomal trafficking of the virus. Subsequently, the conserved domains of capsid proteins interact with cellular membranes and ensure their efficient permeabilization. This review summarizes our current knowledge concerning the role of capsid proteins of small non-enveloped DNA viruses in intracellular membrane perturbation in the early stages of infection.


Subject(s)
Capsid Proteins/metabolism , Cell Membrane/virology , Eukaryotic Cells/virology , Virus Internalization , Adenoviridae/physiology , Capsid Proteins/chemistry , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Nucleus/virology , Endosomes/metabolism , Endosomes/virology , Eukaryotic Cells/metabolism , Host-Pathogen Interactions , Humans , Papillomaviridae/physiology , Parvoviridae/physiology , Polyomaviridae/physiology , Protein Binding , Protein Transport , Receptors, Virus/metabolism , Virus Replication
4.
J Fish Dis ; 37(11): 969-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24117535

ABSTRACT

An attempt was made to determine the replication efficiency of hepatopancreatic parvo-like virus (HPV) of shrimp in different organs of freshwater rice-field crab Paratelphusa hydrodomous (Herbst) using bioassay, PCR, RT-PCR, ELISA, Western blot and q-PCR analyses. Another attempt was made to use this crab as an alternative to penaeid shrimp for the large-scale production of HPV. This crab was found to be highly susceptible to HPV by intramuscular injection. The systemic HPV infection was confirmed by PCR and Western blot analyses in freshwater crab. The expression of capsid protein gene in different organs of infected crab was revealed by RT-PCR analysis. Indirect ELISA was used to quantify the capsid protein in different organs of the crab. The copy number of HPV in different organs of the infected crab was quantified by q-PCR. The results revealed a steady decrease in CT values in different organs of the infected crab during the course of infection. The viral inoculum that was prepared from different organs of the infected crab caused significant mortality in post-larvae of tiger prawn, Penaeus monodon (Fabricius). The results revealed that this rice-field crab could be used as an alternative host for HPV replication and also for large-scale production of HPV.


Subject(s)
Brachyura/virology , Parvoviridae/physiology , Animals , Capsid Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Oryza , Tissue Distribution , Virus Replication
5.
BMC Neurosci ; 7: 46, 2006 Jun 08.
Article in English | MEDLINE | ID: mdl-16762059

ABSTRACT

BACKGROUND: Somatosensation of the mammalian head is mainly mediated by the trigeminal nerve that provides innervation of diverse tissues like the face skin, the conjunctiva of the eyes, blood vessels and the mucouse membranes of the oral and nasal cavities. Trigeminal perception encompasses thermosensation, touch, and pain. Trigeminal chemosensation from the nasal epithelia mainly evokes stinging, burning, or pungent sensations. In vitro characterization of trigeminal primary sensory neurons derives largely from analysis of complete neuronal populations prepared from sensory ganglia. Thus, functional properties of primary trigeminal afferents depending on the area of innervation remain largely unclear. RESULTS: We established a PrV based tracing technique to identify nasal and cutaneous trigeminal neurons in vitro. This approach allowed analysis and comparison of identified primary afferents by means of electrophysiological and imaging measurement techniques. Neurons were challenged with several agonists that were reported to exhibit specificity for known receptors, including TRP channels and purinergic receptors. In addition, TTX sensitivity of sodium currents and IB4 binding was investigated. Compared with cutaneous neurons, a larger fraction of nasal trigeminal neurons showed sensitivity for menthol and capsaicin. These findings pointed to TRPM8 and TRPV1 receptor protein expression largely in nasal neurons whereas for cutaneous neurons these receptors are present only in a smaller fraction. The majority of nasal neurons lacked P2X3 receptor-mediated currents but showed P2X2-mediated responses when stimulated with ATP. Interestingly, cutaneous neurons revealed largely TTX resistant sodium currents. A significantly higher fraction of nasal and cutaneous afferents showed IB4 binding when compared to randomly chosen trigeminal neurons. CONCLUSION: In conclusion, the usability of PrV mediated tracing of primary afferents was demonstrated. Using this technique it could be shown that compared with neurons innervating the skin nasal trigeminal neurons reveal pronounced chemosensitivity for TRPM8 and TRPV1 channel agonists and only partially meet properties typical for nociceptors. In contrast to P2X3 receptors, TRPM8 and TRPV1 receptors seem to be of pronounced physiological relevance for intranasal trigeminal sensation.


Subject(s)
Afferent Pathways/physiology , Chemoreceptor Cells/physiology , Nasal Cavity/innervation , Neurons, Afferent/physiology , Skin/innervation , Trigeminal Nerve/physiology , Afferent Pathways/cytology , Animals , Binding Sites/physiology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Chemoreceptor Cells/cytology , Mice , Neurons, Afferent/cytology , Parvoviridae/physiology , Plant Lectins , Purinergic Agonists , Purinergic P2 Receptor Agonists , Receptors, Purinergic/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X2 , Sodium Channel Agonists , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Staining and Labeling/methods , TRPC Cation Channels/agonists , TRPC Cation Channels/metabolism , TRPM Cation Channels/agonists , TRPM Cation Channels/metabolism , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism , Tetrodotoxin/pharmacology , Trigeminal Nerve/cytology
6.
J Virol ; 79(4): 2287-300, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15681430

ABSTRACT

The parvovirus minute virus of mice (MVM) packages predominantly negative-sense single strands, while its close relative LuIII encapsidates strands of both polarities with equal efficiency. Using genomic chimeras and mutagenesis, we show that the ability to package positive strands maps not, as originally postulated, to divergent untranslated regions downstream of the capsid gene but to the viral hairpins and predominantly to the nick site of OriR, the right-end replication origin. In MVM, the sequence of this site is 5'-CTAT(black triangle down)TCA-3', while in LuIII a two-base insertion (underlined) changes it to 5'-CTATAT(black triangle down)TCA-3'. Matched LuIII genomes differing only at this position (designated LuIII and LuDelta2) packaged 47 and <8% positive-sense strands, respectively. OriR sequences from these viruses were both able to support NS1-mediated nicking in vitro, but initiation efficiency was consistently two- to threefold higher for LuDelta2 derivatives, suggesting that LuIII's ability to package positive strands is determined by a suboptimal right-end origin rather than by strand-specific packaging sequences. These observations support a mathematical "kinetic hairpin transfer" model, previously described by Chen and colleagues (K. C. Chen, J. J. Tyson, M. Lederman, E. R. Stout, and R. C. Bates, J. Mol. Biol. 208:283-296, 1989), that postulates that preferential excision of particular strands is solely responsible for packaging specificity. By analyzing replicative-form (RF) DNA generated in vivo during LuIII and LuDelta2 infections, we extend this model, showing that positive-sense strands do accumulate in LuDelta2 infections as part of duplex RF DNA, but these do not support packaging. However, replication is biphasic, so that accumulation of positive-sense strands is ultimately suppressed, probably because the onset of packaging removes newly displaced single strands from the replicating pool.


Subject(s)
Genome, Viral , Minute Virus of Mice/physiology , Parvoviridae/genetics , Replication Origin/genetics , Virus Replication/genetics , Animals , Capsid , DNA, Viral/analysis , DNA, Viral/chemistry , Mice , Parvoviridae/physiology
7.
J Virol ; 77(6): 3851-8, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12610161

ABSTRACT

Recent studies demonstrated the ability of the recombinant autonomous parvoviruses MVMp (fibrotropic variant of the minute virus of mice) and H-1 to transduce therapeutic genes in tumor cells. However, recombinant vector stocks are contaminated by replication-competent viruses (RCVs) generated during the production procedure. To reduce the levels of RCVs, chimeric recombinant vector genomes were designed by replacing the right-hand region of H-1 virus DNA with that of the closely related MVMp virus DNA and conversely. Recombinant H-1 and MVMp virus pseudotypes were also produced with this aim. In both cases, the levels of RCVs contaminating the virus stocks were considerably reduced (virus was not detected in pseudotyped virus stocks, even after two amplification steps), while the yields of vector viruses produced were not affected. H-1 virus could be distinguished from MVMp virus by its restriction in mouse cells at an early stage of infection prior to detectable viral DNA replication and gene expression. The analysis of the composite viruses showed that this restriction could be assigned to a specific genomic determinant(s). Unlike MVMp virus, H-1 virus capsids were found to be a major determinant of the greater permissiveness of various human cell lines for this virus.


Subject(s)
Genetic Vectors , Minute Virus of Mice/genetics , Parvoviridae/genetics , Recombinant Fusion Proteins/metabolism , Recombination, Genetic , Virus Replication , Animals , Cell Line , Genetic Engineering/methods , Humans , Mice , Minute Virus of Mice/physiology , Parvoviridae/physiology , Rats , Recombinant Fusion Proteins/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
8.
Curr Mol Med ; 3(1): 61-71, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12558075

ABSTRACT

The goal of oncolytic therapy is to exploit the innate ability of viruses to infect tumor cells, replicate in tumor cells, and produce selective oncolysis while sparing normal cells. Although the concept that viruses can be oncolytic is not new, it is only in the last three decades that efforts have been directed at genetically mutating viruses to specifically target characteristics of cancer cells. Several viruses have the potential to infect, replicate and lyse tumor cells, each taking advantage of different host cancer cell biology. This review will focus on the major viruses under current investigation for oncolytic therapy, the mechanism by which they specifically eradicate tumors, and the clinical strategies currently under investigation.


Subject(s)
Adenoviridae/physiology , Neoplasms/therapy , Reoviridae/physiology , Simplexvirus/physiology , Adenoviridae/genetics , Animals , Cell Death/genetics , Genetic Therapy , Humans , Mutation , Newcastle disease virus/physiology , Parvoviridae/physiology , Simplexvirus/genetics , Vesicular stomatitis Indiana virus/physiology
9.
Rev Sci Tech ; 16(1): 146-60, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9329114

ABSTRACT

Within the past decade, viral diseases have emerged as serious economic impediments to successful shrimp farming in many of the shrimp-farming countries of the world. In the western hemisphere, the viral agents of Taura syndrome (TS) and infectious hypodermal and haematopoietic necrosis have caused serious disease epizootics throughout the shrimp-growing regions of the Americas and Hawaii, while in Asia the viral agents of white spot syndrome (WSS) and yellow head (YH) have caused pandemics with catastrophic losses. The international transfer of live shrimp for aquaculture purposes is an obvious mechanism by which the viruses have spread within and between regions in which they have occurred. Shrimp-eating gulls, other seabirds and aquatic insects may also be factors in the spread of shrimp viruses between and within regions. Another potentially important mechanism for the international spread of these pathogens is the trade in frozen commodity shrimp, which may contain viruses exotic to the importing countries. The viral agents of WSS, YH and TS have been found, and demonstrated to be infectious, in frozen shrimp imported into the United States market. Mechanisms identified for the potential transfer of virus in imported frozen products to domestic populations of cultured or wild penaeid shrimp stocks include: the release of untreated liquid or solid wastes from shrimp importing and processing plants directly into coastal waters, improper disposal of solid waste from shrimp importing and processing plants in landfills so that the waste is accessible to gulls and other seabirds, and the use of imported shrimp as bait by sports fishermen.


Subject(s)
Aquaculture , Cryopreservation , Decapoda/virology , Seafood/virology , Americas , Animals , Baculoviridae/classification , Baculoviridae/physiology , Mononegavirales/classification , Mononegavirales/physiology , Parvoviridae/classification , Parvoviridae/physiology , Picornaviridae/classification , Picornaviridae/physiology , Transportation
10.
Rev Sci Tech ; 15(2): 579-601, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8890382

ABSTRACT

Marine penaeid shrimp are effected by approximately twenty viruses, the majority of which were discovered as a result of their negative effects on aquaculture. In the Americas, infectious hypodermal and haematopoietic necrosis (IHHN) virus and Taura syndrome (TS) virus have had a significant negative impact on aquaculture industries and, in one instance, on a commercial fishery. Both viruses have become widely distributed as a consequence of the movement of host stocks for aquaculture. IHHN virus (IHHNV) causes catastrophic losses in cultured and wild Penaeus stylirostris. In marked contrast, P. vannamei is relatively resistant to IHHN but infection results, nonetheless, in poor culture performance. TS virus (TSV) is the 'mirror image' of IHHNV in its effect on P. stylirostris and P. vannamei. TSV causes catastrophic losses in P. vannamei, whereas P. stylirostris is highly resistant to TS. In the less than three years since the discovery of TSV in Ecuador in 1992, the virus has spread rapidly and caused massive production losses in most shrimp-growing countries in the Americas.


Subject(s)
Decapoda/virology , Parvoviridae/physiology , Picornaviridae/physiology , Animals , Commerce , International Cooperation , Specific Pathogen-Free Organisms
11.
J Virol ; 69(6): 3915-9, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7745746

ABSTRACT

Infection of young adult BALB/cByJ mice with mouse parvovirus-1, a newly recognized, lymphocytotropic, nonpathogenic parvovirus, was examined by in situ hybridization. Virus appeared to enter through the small intestine and was disseminated to the liver and lymphoid tissues. Strand-specific probes detected virion DNA in a consistently larger number of cells than replicative forms of viral DNA and/or viral mRNA. The number of signal-positive cells in the intestinal mucosa, lymph nodes, spleen, and thymus increased through day 10 after oral inoculation but decreased after seroconversion. Positive cells were still detected, however, in peripheral lymphoid tissues of mice examined at 9 weeks postinoculation. The results underscore the need to assess potential effects of persistent mouse parvovirus-1 infection on immune function in mice.


Subject(s)
Parvoviridae Infections/virology , Parvoviridae/physiology , Animals , DNA, Viral/analysis , In Situ Hybridization , Intestine, Small/virology , Liver/virology , Lymphoid Tissue/virology , Mice , Mice, Inbred BALB C , Parvoviridae/genetics , Parvoviridae/isolation & purification , Parvoviridae Infections/immunology
12.
Mol Gen Mikrobiol Virusol ; (1): 27-31, 1993.
Article in Russian | MEDLINE | ID: mdl-8510680

ABSTRACT

The possibility of suppression of porcine parvovirus (PPV) reproduction in the culture of thyroid gland cells of a swine that contain the integrated genes for asRNA against the nonstructural proteins of the virus has been studied. 10 cell lines with the asRNA genes have been obtained. The line with the maximal number of integrated gene copies was used to inflict with the parvovirus. The expression of asRNA in this cell line was shown to lead to 95% suppression of PPV replication as compared with the control cell line.


Subject(s)
Parvoviridae/physiology , RNA, Antisense/pharmacology , Thyroid Gland/microbiology , Virus Replication/genetics , Animals , Cell Line , Cells, Cultured , Genetic Vectors , Swine , Thyroid Gland/cytology , Virus Replication/drug effects
13.
Arch Virol ; 130(3-4): 227-36, 1993.
Article in English | MEDLINE | ID: mdl-8517788

ABSTRACT

We have cloned genome fragments of canine parvovirus strain Y1 from replicative-form DNA and double-stranded DNA synthesized from virion DNA in vitro, and constructed a recombinant plasmid containing a full-length Y1 genome (pCPVY 1). When this recombinant plasmid was transfected into cell cultures, an infectious virus could be recovered. To characterize this pCPVY 1-derived virus, its biological properties were compared with those of the parental strain. No difference was observed between them in antigen expression, viral DNA replication, hemagglutination ability, and virus multiplication, indicating that the virus derived from the infectious plasmid inherited the biological properties of the authentic Y1 strain. Therefore, this recombinant plasmid appears to be useful for reverse genetics of canine parvovirus.


Subject(s)
DNA, Viral/genetics , Parvoviridae/genetics , Animals , Cats , Cells, Cultured , Cloning, Molecular , Dogs/microbiology , Genome, Viral , Hemagglutination Tests , Immunoblotting , Parvoviridae/physiology , Plasmids , Viral Plaque Assay
15.
J Virol ; 66(8): 4686-92, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1385833

ABSTRACT

Vertebrate parvoviruses share a similar genomic organization, with the capsid proteins encoded by genes on the right side and nonstructural proteins encoded by genes on the left side. The temporal and cell-specific appearances of these two types of gene products are regulated by a variety of genetic mechanisms. Rodent parvovirus structural proteins, for example, are encoded by a separate promoter which is positively regulated by nonstructural-gene products. In contrast, for the human B19 parvovirus, the analogous structural-gene promoter is nonfunctional, and both left- and right-side transcripts originate from a single promoter and are highly processed. Using a combination of sensitive RNA analyses of wild-type and mutant templates, we have found that the relative abundance of these alternatively processed transcripts appears to be governed by unique postinitiation events. In permissive cells, the steady-state level of right-side structural-gene transcripts predominates over that of left-side nonstructural-gene transcripts. In nonpermissive cells transfected with the B19 virus genome, nonstructural-gene transcripts predominate. Removal of 3' processing signals located in the middle of the viral genome increases transcription of the far right side. Disruption of a polyadenylation signal in this region makes readthrough of full-length right-side transcripts possible. These results suggest that the abundance of B19 virus RNAs is determined by active 3' processing and is coupled to DNA template replication.


Subject(s)
Capsid/genetics , DNA Replication , DNA, Viral/genetics , Parvoviridae/genetics , RNA, Viral/genetics , Viral Core Proteins/genetics , Animals , Base Sequence , Blotting, Northern , Capsid Proteins , Cell Line , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Oligonucleotides, Antisense , Parvoviridae/physiology , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , Restriction Mapping , Templates, Genetic , Transcription, Genetic , Transfection , Viral Nonstructural Proteins
16.
J Virol ; 66(2): 715-22, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1370555

ABSTRACT

Swine testicle (ST) cells and Madin-Darby canine kidney (MDCK) cells differ in their ability to support replication of porcine parvovirus (PPV). Viral replication events in ST cells, a permissive cell type, and MDCK cells, a nonpermissive cell type, were compared in an attempt to elucidate putative mechanisms of restrictive virus replication. Radiolabeled PPV bound to the cell surface of both cell types equally well and the binding was shown to be PPV specific, indicating that the restriction was not at the cell surface level. In contrast, profound differences in intracellular events in PPV replication were observed between these two cell types. Synthesis of viral DNA was limited in MDCK cells in that the percentage of cells with replicative-form DNA as determined by strand-specific probe in situ hybridization was approximately 100-fold lower in MDCK cells than in ST cells at the same multiplicity of infection. Northern (RNA) blot analysis, using oligonucleotide probes derived from both structural and nonstructural protein-coding regions of the PPV genome, revealed four PPV mRNA transcripts from infected ST cells. Comparatively, RNA species from the structural protein coding region were actively transcribed in MDCK cells, but synthesis of RNA species from the nonstructural protein coding region was negligible. Immunoprecipitation of viral polypeptides revealed the three characteristic structural polypeptides, VP1, VP2, and VP3, along with the nonstructural polypeptide, NS-1 from ST cells. In contrast, neither viral structural or nonstructural polypeptides nor progeny virions were produced from MDCK cells. The data suggest that mechanisms controlling permissiveness of cells to PPV infection are associated with the level of viral DNA replication, RNA transcription, and viral antigen expression but not absorption to the cell surface.


Subject(s)
Parvoviridae/physiology , Virus Replication , Animals , Antigens, Viral/analysis , Base Sequence , Blotting, Northern , Cell Line , Dogs , Kidney , Male , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotide Probes , Parvoviridae/genetics , RNA/genetics , RNA/isolation & purification , RNA, Viral/genetics , RNA, Viral/isolation & purification , Swine , Testis
17.
Virology ; 186(2): 368-76, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1733094

ABSTRACT

We have investigated whether canine parvovirus (CPV) infection involves a low pH-dependent entry pathway. The effects of two lysosomotropic bases, NH4Cl and chloroquine, on CPV entry were studied by immunofluorescence and ultrastructural and biochemical methods. In the presence of these reagents, input virions appear to accumulate in large vacuoles. Ultrastructural studies indicated that uptake of virions takes place predominantly in small uncoated vesicles that appear to fuse with larger vesicles. In the presence of NH4Cl, virions accumulate in the latter structures and their uncoating appears to be prevented. Viral DNA as well as antigen synthesis were found to be significantly inhibited in the presence of these reagents. In addition, inhibition of viral DNA and antigen synthesis appeared to be most extensive when NH4Cl was present from 30 min preinfection, whereas no significant inhibition was observed when the cells were treated after 2 hr postinfection. Thus, the results indicate that CPV requires exposure to low pH in an endosomal compartment to initiate a productive infection.


Subject(s)
Parvoviridae/physiology , Ammonium Chloride/pharmacology , Animals , Antigens, Viral/biosynthesis , Cell Line , Chloroquine/pharmacology , DNA, Viral/biosynthesis , Dogs , Endocytosis , Fluorescent Antibody Technique , Hydrogen-Ion Concentration , Microscopy, Electron , Parvoviridae/genetics , Parvoviridae/immunology , Parvoviridae/ultrastructure , Virus Replication
18.
Med Microbiol Immunol ; 181(3): 153-64, 1992.
Article in English | MEDLINE | ID: mdl-1522825

ABSTRACT

To investigate a possible role of cytokines in parvovirus-mediated suppression of tumorigenesis, we tested in cell culture whether parvoviruses are able to induce interferon (IFN)-beta, tumor necrosis factor (TNF)-alpha or interleukin-6 (IL-6). Infection of rodent or human cells with the parvoviruses minute virus of mice (MVM), H-1 or adeno-associated virus (AAV) types 2 or 5 failed to induce expression of the luciferase or beta-galactosidase reporter genes transfected into these cells as constructs containing an IFN-beta promoter. Parvoviruses did weakly induce synthesis of TNF-alpha and of IL-6 in cell culture and could slightly enhance synthesis of these cytokines when induced by other agents. These in vitro data suggest that the rather unspecific tumor-suppressive properties of parvoviruses are unlikely to be attributable to stimulation of the synthesis of IFN, TNF or IL-6.


Subject(s)
Interferon-beta/biosynthesis , Interleukin-6/biosynthesis , Parvoviridae/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Transformation, Viral , Fibroblasts/microbiology , HeLa Cells/microbiology , Humans , Mice , Transfection , Tumor Cells, Cultured
19.
Arch Virol ; 123(3-4): 323-33, 1992.
Article in English | MEDLINE | ID: mdl-1562235

ABSTRACT

Monoclonal antibody technologies were applied to the study of early events in porcine parvovirus (PPV) infections in vitro. Balb/c mice were immunized with whole swine testicle cells and hybridomas were produced following fusion with myeloma cells. Resultant clones were screened firstly in an ELISA system, to detect monoclonal antibody recognition of swine testicle cells, and secondly, in a fluorescent antibody test to detect monoclonal antibody which inhibited production of PPV antigen. One clone, 1H11, which satisfied these screening requirements, recognized proteins present in cell lines both permissive and non-permissive for porcine parvovirus replication and inhibited the production of virus progeny of several PPV isolates. A linear staining pattern of cross-linked plasma membranes, indicative of monoclonal antibody binding at the cell membrane, was demonstrated by indirect immunofluorescence assays. In immunoblotting experiments, 1H11 recognized a polypeptide of approximately 40 kDa in size, present in both permissive and non-permissive cell lines.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Surface/immunology , Parvoviridae/physiology , Receptors, Virus/immunology , Animals , Antibody Specificity , Cell Line , Dogs , Enzyme-Linked Immunosorbent Assay , HeLa Cells , Humans , Immunoblotting , Male , Parvoviridae/immunology , Swine , Virus Replication
20.
Virology ; 184(1): 117-30, 1991 Sep.
Article in English | MEDLINE | ID: mdl-1831309

ABSTRACT

We generated a mutation in the gene for the nonstructural protein NS2 of parvovirus H-1 in which the highly conserved dinucleotide AG at the 3' splice acceptor site of NS2 intron 1 was mutated to CG. The mutation does not change the amino acid sequence for NS1. The splice acceptor (SA) mutant gene was introduced into the H-1 virus (H-1SA) and an infectious clone of LuIII (pLuH1SA). The R2 transcripts encoding NS2 were absent by both Northern blot and primer extension analysis in the LuH1SA or H-1SA virus-infected cells and the NS2 protein was undetectable in the infected cell lysate by immunoprecipitation. These NS2 null mutant viruses were capable of lytic growth in cell lines that were derived from human, hamster, and dog, but they produced lower virus titers than wild-type H-1. The H-1SA virus nonproductively infected Rat2 rat fibroblasts and transformed Rat2 cell lines. Analysis of synchronized infections of rat fibroblasts demonstrated that H-1SA viral duplex replicative form DNA replication was reduced and that single-stranded progeny DNA was deficient compared to wild-type H-1. In addition, H-1SA viral protein synthesis was about 10% of wild-type virus and virions were not detectable in rat fibroblasts. However, H-1SA mRNAs R1 and R3 accumulated to wild-type levels. NS2 was also required for productive infection in newborn rats but not in newborn hamsters. These results indicate that NS2 plays an important role in the regulation of viral protein synthesis in rat cells in vivo and in vitro.


Subject(s)
Capsid/metabolism , DNA Replication , Mutagenesis, Site-Directed , Parvoviridae/physiology , Viral Core Proteins/metabolism , Viral Proteins/biosynthesis , Virus Replication , Animals , Base Sequence , Blotting, Southern , Capsid/genetics , Cell Line , DNA, Viral/genetics , Kinetics , Microscopy, Electron , Molecular Sequence Data , Oligonucleotide Probes , Parvoviridae/genetics , Parvoviridae/metabolism , Parvoviridae/ultrastructure , Polymerase Chain Reaction , Rats , Restriction Mapping , Viral Core Proteins/genetics , Viral Nonstructural Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...