Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Pathog Dis ; 77(5)2019 07 01.
Article in English | MEDLINE | ID: mdl-31603501

ABSTRACT

H1 parvovirus (H1PV) infection in rats is of concern to the research community as infection may compromise rodent-based experiments. The aim of this study was to evaluate the influence of H1PV infection on rat gut microbiota. Inbred Wistar rats were infected with H1PV by routine gavage and clinical signs were recorded. Gross anatomical and histopathological examination of the gut was performed, as was immune cytokine analysis. The cecal contents were also collected for 16S rRNA sequencing. Gross anatomical examination showed distention of the ileum associated with flatulence after infection, while histopathological examination showed hyperemia and inflammatory cell infiltration in the ileum. Upregulation of the interleukin-6 in sera in H1PV infected rats was also detected. The gut microbiota had been significantly changed in H1PV infected rats: there was a reduction in several bacteria species including probiotic bacteria from the genera Parabacteroides and Butyricicoccus, while others were increased, including those from the genera Methanobrevibacter and Syntrophococcus. Taken together, these results demonstrate that chronic H1PV infection in rats leads to gastrointestinal inflammation with flatulence. The gut microbiota alterations were associated with decreased polymorphisms, reduced abundance of probiotic bacteria and increased abundance of methane-producing bacteria.


Subject(s)
Bacteria/classification , Dysbiosis/veterinary , Gastrointestinal Microbiome , Parvoviridae Infections/veterinary , Parvovirus/growth & development , Rodent Diseases/pathology , Animals , Bacteria/genetics , Cluster Analysis , Cytokines/blood , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Dysbiosis/pathology , Ileum/pathology , Parvoviridae Infections/complications , Parvoviridae Infections/pathology , Phylogeny , RNA, Ribosomal, 16S/genetics , Rats, Wistar , Rodent Diseases/microbiology , Rodent Diseases/virology , Sequence Analysis, DNA
2.
Arch Virol ; 164(10): 2479-2491, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31321584

ABSTRACT

Codon usage is biased in most species, and the pattern of codon usage bias is specific to each species or group of closely related species. Although viruses use the host translational machinery for synthesis of their proteins, their codon usage patterns do not match those of their host. Viral codon usage is determined by a complex interplay of mutational bias, genome composition constraints, translational adaptation to the host, and host cellular innate defense. The codon usage of parvoviruses was previously shown not to be strongly biased and selective pressure was found to be a dominating factor driving codon usage. The family Parvoviridae includes the genus Dependoparvovirus, some of the members of which require a helper virus to complete their replication cycle, whereas the rest of the family can replicate without the need for helper viruses. Here, we show that difference in the replication strategy of these viruses may be an important factor determining viral codon usage. Hierarchical clustering and principal component analysis revealed that the codon usage pattern of adeno-associated viruses (AAVs) of the genus Dependoparvovirus is distinct from that of members of the other genera of vertebrate parvoviruses, and even from that of independent viruses of the genus Dependoparvovirus. Furthermore, the codon usage of human AAVs was found to be similar to that of some human adenoviruses in hierarchical clustering and principal component analysis. This suggests that the codon usage of AAVs is different from that of other parvoviruses because of their distinctive replication strategy and that their codon usage is probably driven by forces similar to those that shaped the codon usage pattern of their helper viruses.


Subject(s)
Codon , Parvovirus/growth & development , Parvovirus/genetics , Virus Replication , Animals , Humans
3.
Int J Hyg Environ Health ; 221(3): 479-488, 2018 04.
Article in English | MEDLINE | ID: mdl-29402695

ABSTRACT

Sewage contains a mixed ecosystem of diverse sets of microorganisms, including human pathogenic viruses. Little is known about how conventional as well as advanced treatments of sewage, such as ozonation, reduce the environmental spread of viruses. Analyses for viruses were therefore conducted for three weeks in influent, after conventional treatment, after additional ozonation, and after passing an open dam system at a full-scale treatment plant in Knivsta, Sweden. Viruses were concentrated by adsorption to a positively charged filter, from which they were eluted and pelleted by ultracentrifugation, with a recovery of about 10%. Ion Torrent sequencing was used to analyze influent, leading to the identification of at least 327 viral species, most of which belonged to 25 families with some having unclear classification. Real-time PCR was used to test for 21 human-related viruses in inlet, conventionally treated, and ozone-treated sewage and outlet waters. The viruses identified in influent and further analyzed were adenovirus, norovirus, sapovirus, parechovirus, hepatitis E virus, astrovirus, pecovirus, picobirnavirus, parvovirus, and gokushovirus. Conventional treatment reduced viral concentrations by one to four log10, with the exception of adenovirus and parvovirus, for which the removal was less efficient. Ozone treatment led to a further reduction by one to two log10, but less for adenovirus. This study showed that the amount of all viruses was reduced by conventional sewage treatment. Further ozonation reduced the amounts of several viruses to undetectable levels, indicating that this is a promising technique for reducing the transmission of many pathogenic human viruses.


Subject(s)
Ozone , Sewage/virology , Virus Diseases/virology , Viruses/growth & development , Waste Disposal, Fluid/methods , Adenoviridae/growth & development , Animals , Humans , Parvovirus/growth & development , Sweden , Virus Diseases/transmission , Wastewater/virology , Water , Water Supply
4.
Arch Virol ; 162(12): 3869-3874, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28884224

ABSTRACT

For members of the family Parvoviridae, rescue of infectious virus from recombinant plasmid is usually done in cultured cells. In this study, the whole genome of the pathogenic Muscovy duck parvovirus (MDPV) strain YY was cloned into the pBluescript II (SK) vector, generating recombinant plasmid pYY. With the aid of a transfection reagent, pYY plasmid was inoculated into 11-day-old embryonated Muscovy duck eggs via the chorioallantoic membrane route, resulting in the successful rescue of infectious virus and death of the embryos. The rescued virus exhibited pathogenicity in Muscovy ducklings similar to that of its parental strain, as evaluated based on the mortality rate. The results demonstrate that plasmid transfection in embryonated Muscovy duck eggs is a convenient and efficacious method for rescue of infectious MDPV in comparison to transfection of primary cells, which is somewhat time-consuming and laborious.


Subject(s)
Ducks/virology , Parvovirus/growth & development , Parvovirus/genetics , Plasmids , Transfection , Animals , Cloning, Molecular , Ducks/growth & development , Embryo, Nonmammalian/virology
5.
Virol J ; 13: 104, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27329377

ABSTRACT

BACKGROUND: Muscovy duck parvovirus (MDPV) is the etiological agent of Muscovy duckling parvoviral disease, which is characterized by diarrhea, locomotive dysfunction, stunting, and death in young ducklings, and causes substantial economic losses in the Muscovy duck industry worldwide. FZ91-30 is an attenuated vaccine strain that is safe and immunogenic to ducklings, but the genomic information and molecular mechanism underlining the attenuation are not understood. METHODS: The FZ91-30 strain was propagated in 11-day-old embryonated goose eggs, and viral particles were purified from the pooled allantoic fluid by differential centrifugation and ultracentrifugation. Single-stranded genomic DNA was extracted and annealed to form double-stranded DNA. The dsDNA digested with NcoI resulted two sub-genomic fragments, which were then cloned into the modified plasmid pBluescript II SK, respectively, generating plasmid pBSKNL and pBSKNR. The sub-genomic plasmid clones were sequenced and further combined to construct the plasmid pFZ that contained the entire genome of strain FZ91-30. The complete genome sequences of strain FM and YY and partial genome sequences of other strains were retrieved from GenBank for sequence comparison. The plasmid pFZ containing the entire genome of FZ91-30 was transfected in 11-day-old embryonated goose eggs via the chorioallantoic membranes route to rescue infectious virus. A genetic marker was introduced into the rescued virus to discriminate from its parental virus. RESULTS: The genome of FZ91-30 consists of 5,131 nucleotides and has 98.9 % similarity to the FM strain. The inverted terminal repeats (ITR) are 456 nucleotides in length, 14 nucleotides longer than that of Goose parvovirus (GPV). The exterior 415 nucleotides of the ITR form a hairpin structure, and the interior 41 nucleotides constitute the D sequence, a reverse complement of the D' sequence at the 3' ITR. Amino acid sequence alignment of the VP1 proteins between FZ91-30 and five pathogenic MDPV strains revealed that FZ91-30 had five mutations; two in the unique region of the VP1 protein (VP1u) and three in VP3. Sequence alignment of the Rep1 proteins revealed two amino acid alterations for FZ91-30, both of which were conserved for two pathogenic strains YY and P. Transfection of the plasmid pFZ in 11-day-old embryonated goose eggs resulted in generation of infectious virus with similar biological properties as compared with the parental strain. CONCLUSIONS: The amino acid mutations identified in the VP1 and Rep1 protein may contribute to the attenuation of FZ91-30 in Muscovy ducklings. Plasmid transfection in embryonated goose eggs was suitable for rescue of infectious MDPV.


Subject(s)
Geese/virology , Parvoviridae Infections/veterinary , Parvovirus/growth & development , Parvovirus/immunology , Poultry Diseases/virology , Amino Acid Sequence , Animals , Base Sequence , Ducks/virology , Geese/embryology , Parvoviridae Infections/embryology , Parvoviridae Infections/immunology , Parvoviridae Infections/virology , Parvovirus/genetics , Parvovirus/isolation & purification , Poultry Diseases/embryology , Poultry Diseases/immunology , Poultry Diseases/pathology , Sequence Alignment , Sequence Analysis, DNA , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/genetics , Viral Vaccines/immunology
6.
Arch Virol ; 160(3): 711-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25559668

ABSTRACT

In this study, the complete genome of the virulent strain LH of goose parvovirus (GPV) was sequenced and cloned into the pBluescript II (SK) plasmid vector. Sequence alignments of the inverted terminal repeats (ITR) of GPV strains revealed a common 14-nt-pair deletion in the stem of the palindromic structure in the LH strain and three other strains isolated after 1982 when compared to three GPV strains isolated earlier than that time. Transfection of 11-day-old embryonated goose eggs with the plasmid pLH, which contains the entire genome of strain LH, resulted in successful rescue of the infectious virus. Death of embryos after transfection via the chorioallantoic membrane infiltration route occurred earlier than when transfection was done via the allantoic cavity inoculation route. The rescued virus exhibited virulence similar to that of its parental virus, as evaluated by the mortality rate in goslings. Generation of the pathogenic infectious clone provides us with a powerful tool to elucidate the molecular pathogenesis of GPV in the future.


Subject(s)
DNA, Viral/chemistry , DNA, Viral/genetics , Genome, Viral , Parvovirus/genetics , Parvovirus/pathogenicity , Animals , Base Sequence , Cloning, Molecular , Embryo, Mammalian/virology , Geese/virology , Molecular Sequence Data , Parvovirus/growth & development , Parvovirus/isolation & purification , Sequence Alignment , Sequence Analysis, DNA , Survival Analysis , Transfection , Virulence
7.
J Vet Med Sci ; 76(9): 1277-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24871643

ABSTRACT

Scallop shell powder produced by calcination process - the average diameter of the powder particles being 20 µm (SSP) - was further ground into nano-sized particles, with average diameter of 500 nm, here designated CaO-Nano. Solution of CaO-Nano could inactivate avian influenza virus within 5 sec, whereas the solution of SSP could not even after 1 hr incubation. CaO-Nano solution could also inactivate Newcastle disease virus and goose parvovirus within 5 sec and 30 sec, respectively. The virus-inactivating capacity (neutralizing index: NI>3) of the solution was not reduced by the presence of 20% fetal bovine serum. CaO-Nano solution seems to be a good candidate of materials for enhancement of biosecurity in farms.


Subject(s)
Calcium/pharmacology , Influenza A virus/growth & development , Nanoparticles/administration & dosage , Newcastle disease virus/growth & development , Parvovirus/growth & development , Poultry Diseases/virology , Virus Inactivation , Animal Shells , Animals , Neutralization Tests/veterinary , Pectinidae , Poultry Diseases/prevention & control
8.
J Virol ; 88(10): 5263-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24574398

ABSTRACT

UNLABELLED: Novel therapies employing oncolytic viruses have emerged as promising anticancer modalities. The cure of particularly aggressive malignancies requires induction of immunogenic cell death (ICD), coupling oncolysis with immune responses via calreticulin, ATP, and high-mobility group box protein B1 (HMGB1) release from dying tumor cells. The present study shows that in human pancreatic cancer cells (pancreatic ductal adenocarcinoma [PDAC] cells n=4), oncolytic parvovirus H-1 (H-1PV) activated multiple interconnected death pathways but failed to induce calreticulin exposure or ATP release. In contrast, H-1PV elevated extracellular HMGB1 levels by 4.0±0.5 times (58%±9% of total content; up to 100 ng/ml) in all infected cultures, whether nondying, necrotic, or apoptotic. An alternative secretory route allowed H-1PV to overcome the failure of gemcitabine to trigger HMGB1 release, without impeding cytotoxicity or other ICD activities of the standard PDAC medication. Such broad resistance of H-1PV-induced HMGB1 release to apoptotic blockage coincided with but was uncoupled from an autocrine interleukin-1ß (IL-1ß) loop. That and the pattern of viral determinants maintained in gemcitabine-treated cells suggested the activation of an inflammasome/caspase 1 (CASP1) platform alongside DNA detachment and/or nuclear exclusion of HMGB1 during early stages of the viral life cycle. We concluded that H-1PV infection of PDAC cells is signaled through secretion of the alarmin HMGB1 and, besides its own oncolytic effect, might convert drug-induced apoptosis into an ICD process. A transient arrest of cells in the cyclin A1-rich S phase would suffice to support compatibility of proliferation-dependent H-1PV with cytotoxic regimens. These properties warrant incorporation of the oncolytic virus H-1PV, which is not pathogenic in humans, into multimodal anticancer treatments. IMPORTANCE: The current therapeutic concepts targeting aggressive malignancies require an induction of immunogenic cell death characterized by exposure of calreticulin (CRT) as well as release of ATP and HMGB1 from dying cells. In pancreatic tumor cells (PDAC cells) infected with the oncolytic parvovirus H-1PV, only HMGB1 was released by all infected cells, whether nondying, necrotic, or succumbing to one of the programmed death pathways, including contraproductive apoptosis. Our data suggest that active secretion of HMGB1 from PDAC cells is a sentinel reaction emerging during early stages of the viral life cycle, irrespective of cell death, that is compatible with and complements cytotoxic regimens. Consistent induction of HMGB1 secretion raised the possibility that this reaction might be a general "alarming" phenomenon characteristic of H-1PV's interaction with the host cell; release of IL-1ß points to the possible involvement of a danger-sensing inflammasome platform. Both provide a basis for further virus-oriented studies.


Subject(s)
Antineoplastic Agents/metabolism , Cell Death , Deoxycytidine/analogs & derivatives , Epithelial Cells/physiology , Oncolytic Viruses/growth & development , Parvovirus/growth & development , Cell Line, Tumor , Deoxycytidine/metabolism , Epithelial Cells/drug effects , Epithelial Cells/virology , HMGB1 Protein/metabolism , Humans , Signal Transduction , Gemcitabine
9.
J Virol Methods ; 200: 41-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24565999

ABSTRACT

The SYG61v is an attenuated goose parvovirus (GPV) that has been used as a vaccine strain in China. The genome of SYG61v was sequenced to attempt to identify the genetic basis for the attenuation of this strain. The entire genome consists of 5102 nucleotides (nts), with four nt deletions compared to that of virulent strain B. The inverted terminal repeats (ITR) are 442 nts in length, of which 360 nts form a stem region, and 43 nts constitute the bubble region. Although mutations were observed throughout the ITR, no mismatch was found in the stem. Alignment with other pathogenic GPV strains (B, 82-0321, 06-0329, and YZ99-5) indicated that there are 10 and 11 amino acid mutations in the Rep1 and VP1 proteins of SYG61v, respectively. The complete genome of SYG61v was cloned into the pBluescript II vector and an infectious plasmid pSYG61v was generated. Infectious progeny virus was successfully rescued through transfection of the plasmid pSYG61v in embryonated goose eggs and yielded viral titers similar to its parental virus, as evaluated by ELD50.


Subject(s)
Cloning, Molecular , Parvovirus/growth & development , Parvovirus/genetics , Viral Vaccines/genetics , Virion/isolation & purification , Zygote/virology , Animals , China , DNA, Viral/chemistry , DNA, Viral/genetics , Geese , Molecular Sequence Data , Mutation , Polymorphism, Genetic , Sequence Alignment , Sequence Analysis, DNA , Vaccines, Attenuated/genetics , Virion/genetics , Virulence
10.
Virology ; 446(1-2): 37-48, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24074565

ABSTRACT

The rodent parvoviruses are known to be oncoselective, and lytically infect many transformed human cells. Because current therapeutic regimens for metastatic melanoma have low response rates and have little effect on improving survival, this disease is a prime candidate for novel approaches to therapy, including oncolytic parvoviruses. Screening of low-passage, patient-derived melanoma cell lines for multiplicity-dependent killing by a panel of five rodent parvoviruses identified LuIII as the most melanoma-lytic. This property was mapped to the LuIII capsid gene, and an efficiently melanoma tropic chimeric virus shown to undergo three types of interaction with primary human melanoma cells: (1) complete lysis of cultures infected at very low multiplicities; (2) acute killing resulting from viral protein synthesis and DNA replication, without concomitant expansion of the infection, due to failure to export progeny virions efficiently; or (3) complete resistance that operates at an intracellular step following virion uptake, but preceding viral transcription.


Subject(s)
Melanoma/virology , Oncolytic Viruses/growth & development , Parvovirus/growth & development , Cell Line, Tumor , Cell Survival , Humans
11.
Avian Dis ; 57(1): 123-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23678740

ABSTRACT

Previously we identified a novel parvovirus from enteric contents of chickens that were affected by enteric diseases. Comparative sequence analysis showed that the chicken parvovirus (ChPV) represented a new member in the Parvoviridae family. Here, we describe some of the pathogenic characteristics of ChPV in young broilers. Following experimental infection, 2-day-old broiler chickens showed characteristic signs of enteric disease. Runting-stunting syndrome (RSS) was observed in four of five experimental groups with significant growth retardation between 7 and 28 days postinoculation (DPI). Viral growth in small intestine and shedding was detected at early times postinoculation, which was followed by viremia and generalization of infection. ChPV could be detected in most of the major tissues for 3 to 4 wk postinoculation. Immunohistochemistry staining revealed parvovirus-positive cells in the duodenum of inoculated birds at 7 and 14 DPI. Our data indicate that ChPV alone induces RSS in broilers and is important determinant in the complex etiology of enteric diseases of poultry.


Subject(s)
Chickens , Parvoviridae Infections/veterinary , Parvovirus/pathogenicity , Poultry Diseases/virology , Animals , Antibodies, Viral/blood , Diarrhea/veterinary , Diarrhea/virology , Duodenum/pathology , Enzyme-Linked Immunosorbent Assay/veterinary , Growth Disorders/veterinary , Growth Disorders/virology , Parvoviridae Infections/pathology , Parvoviridae Infections/physiopathology , Parvoviridae Infections/virology , Parvovirus/growth & development , Polymerase Chain Reaction/veterinary , Poultry Diseases/pathology , Poultry Diseases/physiopathology , Virus Shedding , Weight Gain
12.
J Virol ; 86(13): 7280-91, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22553327

ABSTRACT

Because productive infection by parvoviruses requires cell division and is enhanced by oncogenic transformation, some parvoviruses may have potential utility in killing cancer cells. To identify the parvovirus(es) with the optimal oncolytic effect against human glioblastomas, we screened 12 parvoviruses at a high multiplicity of infection (MOI). MVMi, MVMc, MVM-G17, tumor virus X (TVX), canine parvovirus (CPV), porcine parvovirus (PPV), rat parvovirus 1A (RPV1A), and H-3 were relatively ineffective. The four viruses with the greatest oncolytic activity, LuIII, H-1, MVMp, and MVM-G52, were tested for the ability, at a low MOI, to progressively infect the culture over time, causing cell death at a rate higher than that of cell proliferation. LuIII alone was effective in all five human glioblastomas tested. H-1 progressively infected only two of five; MVMp and MVM-G52 were ineffective in all five. To investigate the underlying mechanism of LuIII's phenotype, we used recombinant parvoviruses with the LuIII capsid replacing the MVMp capsid or with molecular alteration of the P4 promoter. The LuIII capsid enhanced efficient replication and oncolysis in MO59J gliomas cells; other gliomas tested required the entire LuIII genome to exhibit enhanced infection. LuIII selectively infected glioma cells over normal glial cells in vitro. In mouse models, human glioblastoma xenografts were selectively infected by LuIII when administered intratumorally; LuIII reduced tumor growth by 75%. LuIII also had the capacity to selectively infect subcutaneous or intracranial gliomas after intravenous inoculation. Intravenous or intracranial LuIII caused no adverse effects. Intracranial LuIII caused no infection of mature mouse neurons or glia in vivo but showed a modest infection of developing neurons.


Subject(s)
Cell Death , Glioblastoma/virology , Host Specificity , Neuroglia/virology , Oncolytic Viruses/physiology , Parvovirus/physiology , Virus Replication , Animals , Cell Line, Tumor , Disease Models, Animal , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Mice , Oncolytic Virotherapy/adverse effects , Oncolytic Virotherapy/methods , Oncolytic Viruses/growth & development , Oncolytic Viruses/pathogenicity , Parvovirus/growth & development , Parvovirus/pathogenicity , Treatment Outcome
13.
Curr Gene Ther ; 5(3): 265-71, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15975004

ABSTRACT

The human parvovirus Adeno-Associated virus (AAV-2) has been classified as a Dependovirus because it requires the presence of a helper virus to achieve a productive replication cycle. Several viruses such as Adenovirus (Ad), Herpes Simplex Virus (HSV), Vaccinia virus, and human papillomaviruses (HPV) can provide the helper activities required for AAV growth. The studies on the helper activities provided by adenovirus have provided useful information not only to understand the AAV-2 biology but also to develop tools for the production of recombinant AAV particles (rAAV). This review will focus on the current knowledge about the helper activities provided by the most extensively studied helper viruses, Ad and HSV-1, and also illustrate the methods used to supply the helper functions rAAV assembly.


Subject(s)
Dependovirus/growth & development , Dependovirus/genetics , Genetic Therapy/methods , Helper Viruses/physiology , Humans , Parvovirus/genetics , Parvovirus/growth & development
14.
Biol Pharm Bull ; 28(5): 905-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15863903

ABSTRACT

The present study aimed at investigating the antiviral effects of 2-amino-4,4alpha-dihydro-4alpha-7-dimethyl-3H-phenoxazine-3-one (Phx-1) and 3-amino-1,4alpha-dihydro-4alpha-8-dimethyl-2H-phenoxazine-2-one (Phx-2) on 6 representative viruses: poliovirus, porcine parvovirus, simian virus 40 (SV-40), herpes simplex virus-1 (HSV-1), Sindbis virus, and vesicular stomatitis virus (VSV). Phx-1 and Phx-2 suppressed the proliferation of poliovirus in Vero cells and that of porcine parvovirus in ESK cells at concentrations between 0.25 microg/ml and 2 microg/ml, when the cells were treated with Phx-1 and Phx-2 for 1 h and then inoculated with these viruses. The proliferation of the other viruses, SV-40, HSV-1, Sindbis virus, and VSV, in the host cells was not influenced by Phx-1 or Phx-2 at concentrations less than 20 microg/ml. The results suggest that Phx-1 and Phx-2 may be useful to prevent the proliferation of poliovirus and porcine parvovirus infection and may contribute to developing new antiviral drugs in future.


Subject(s)
Antiviral Agents/pharmacology , Oxazines/pharmacology , Parvovirus/drug effects , Parvovirus/growth & development , Poliovirus/drug effects , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Macaca mulatta , Oxazines/chemistry , Poliovirus/growth & development , Simian virus 40 , Swine , Vero Cells
15.
Vet Microbiol ; 98(2): 159-63, 2004 Feb 04.
Article in English | MEDLINE | ID: mdl-14741128

ABSTRACT

Porcine circovirus-2 (PCV-2) is the necessary cause of post-weaning multisystemic wasting syndrome (PMWS) in swine; however, a variety of co-factors, including other infectious agents, are thought to be necessary in the full expression of disease. Porcine parvovirus (PPV) was found in the inoculum used in the first experiments to reproduce PMWS in gnotobiotic swine. Retrospective and prospective studies in the field and laboratory have demonstrated PCV-2 can act synergistically with PPV to enhance the severity of PMWS. PCV-2 has been shown to play a role in the porcine infectious disease complex (PRDC). Other co-infecting agents with PCV-2 in the lung include, porcine reproductive and respiratory syndrome virus (PRRSV), swine influenza virus (SIV) and Mycoplasma hyopneumoniae. Exposure of pregnant sows to PPV, PRRSV, or encephalomyocarditis virus may interact with PCV-2 infected foetuses. The severity of hepatic lesions in PCV-2 infected pigs may be enhanced by co-infection with agents such as swine hepatitis E virus and Aujezsky's disease virus. Additional studies are required to determine the mechanistic basis for the interaction of PCV-2 with other agents in the pathogenesis of the various clinical syndromes that have been associated with PCV-2 infection.


Subject(s)
Circoviridae Infections/veterinary , Circovirus/growth & development , Swine Diseases/virology , Wasting Syndrome/veterinary , Animals , Circoviridae Infections/complications , Circoviridae Infections/immunology , Circovirus/immunology , Influenza A virus/growth & development , Influenza A virus/immunology , Mycoplasma hyopneumoniae/growth & development , Mycoplasma hyopneumoniae/immunology , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/veterinary , Parvoviridae Infections/complications , Parvoviridae Infections/immunology , Parvoviridae Infections/veterinary , Parvovirus/growth & development , Parvovirus/immunology , Pneumonia of Swine, Mycoplasmal/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/growth & development , Swine , Wasting Syndrome/complications , Wasting Syndrome/immunology
16.
Curr Opin Microbiol ; 6(4): 392-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12941411

ABSTRACT

The past few years have seen major advances in our understanding of the controls of evolution, host range and cell tropism of parvoviruses. Notable findings have included the identification of the transferrin receptor TfR as the cell surface receptor for canine parvovirus and feline panleukopenia virus, and also the finding that specific binding to the canine TfR led to the emergence of canine parvovirus as a new pathogen in dogs. The structures of the adeno-associated virus-2 and porcine parvovirus capsids, along with those of the minute virus of mice, have also advanced our understanding of parvovirus biology. Structure-function studies have shown that in several different parvoviruses the threefold spikes or peaks of the capsid control several aspects of cell tropism and host range, and that those are subject to selective pressures leading to viral evolution. The cell and tissue tropisms of different adeno-associated virus serotypes were demonstrated to be due, in part, to specific receptor binding.


Subject(s)
Parvoviridae Infections/virology , Parvovirus/growth & development , Receptors, Cell Surface/physiology , Animals , Parvovirus/pathogenicity , Virulence
17.
Cell Stress Chaperones ; 7(3): 258-68, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12482202

ABSTRACT

Molecular chaperone complexes containing heat shock protein (Hsp) 70 and Hsp90 are regulated by cochaperones, including a subclass of regulators, such as Hsp70 interacting protein (Hip), C-terminus of Hsp70 interacting protein (CHIP), and Hsp70-Hsp90 organizing factor (Hop), that contain tetratricopeptide repeats (TPRs), where Hsp70 refers to Hsp70 and its nearly identical constitutive counterpart, Hsc70, together. These proteins interact with the Hsp70 to regulate adenosine triphosphatase (ATPase) and folding activities or to generate the chaperone complex. Here we provide evidence that small glutamine-rich protein/viral protein U-binding protein (SGT/UBP) is a cochaperone that negatively regulates Hsp70. By "Far-Western" and pull-down assays, SGT/UBP was shown to interact directly with Hsp70 and weakly with Hsp90. The interaction of SGT/UBP with both these protein chaperones was mapped to 3 TPRs in SGT/UBP (amino acids 95-195) that are flanked by charged residues. Moreover, SGT/UBP caused an approximately 30% reduction in both the intrinsic ATPase activity of Hsc70 and the ability of Hsc70 to refold denatured luciferase in vitro. This negative effect of SGT/UBP on Hsc70 is similar in magnitude to that observed for the cochaperone CHIP. A role for SGT/UBP in protein folding is also supported by evidence that a yeast strain containing a deletion in the yeast homolog to SGT/UBP (delta SGT/UBP) displays a 50-fold reduction in recovery from heat shock compared with the wild type parent. Together, these results are consistent with a regulatory role for SGT/UBP in the chaperone complex.


Subject(s)
Carrier Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Proteins/metabolism , Adenosine Triphosphatases/metabolism , Carrier Proteins/genetics , DNA Replication , Gene Deletion , Glutamine , HIV/growth & development , HIV/metabolism , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Molecular Chaperones/genetics , Mutagenesis , Parvovirus/growth & development , Parvovirus/metabolism , Protein Folding , Proteins/genetics , Virion/metabolism , Virus Replication
18.
Methods ; 28(2): 168-81, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12413415

ABSTRACT

Parvoviruses are small, icosahedral viruses (approximately 25 nm) containing a single-strand DNA genome (approximately 5 kb) with hairpin termini. Autonomous parvoviruses (APVs) are found in many species; they do not require a helper virus for replication but they do require proliferating cells (S-phase functions) and, in some cases, tissue-specific factors. APVs can protect animals from spontaneous or experimental tumors, leading to consideration of these viruses, and vectors derived from them, as anticancer agents. Vector development has focused on three rodent APVs that can infect human cells, namely, LuIII, MVM, and H1. LuIII-based vectors with complete replacement of the viral coding sequences can direct transient or persistent expression of transgenes in cell culture. MVM-based and H1-based vectors with substitution of transgenes for the viral capsid sequences retain viral nonstructural (NS) coding sequences and express the NS1 protein. The latter serves to amplify the vector genome in target cells, potentially contributing to antitumor activity. APV vectors have packaging capacity for foreign DNA of approximately 4.8 kb, a limit that probably cannot be exceeded by more than a few percent. LuIII vectors can be pseudotyped with capsid proteins from related APVs, a promising strategy for controlling tissue tropism and circumventing immune responses to repeated administration. Initial success has been achieved in targeting such a pseudotyped vector by genetic modification of the capsid. Subject to advances in production and purification methods, APV vectors have potential as gene transfer agents for experimental and therapeutic use, particularly for cancer therapy.


Subject(s)
DNA Replication/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Parvovirus/genetics , Animals , Humans , Parvovirus/growth & development
19.
Res Vet Sci ; 70(3): 221-5, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11676617

ABSTRACT

Dogs with clinical signs consistent with parvoviral enteritis and leukopenia (total leukocyte count < 5.0 x 10(9) l(-1)) were included in this randomised double-blind study (treatment group: n = 22; control group: n = 21). The dogs in the treatment group received a subcutaneous daily injection of 10 microg kg(-1) of recombinant human granulocyte colony-stimulating factor (rhG-CSF) for 5 days. Clinical and blood investigations were performed prior to the first injection, daily during the treatment period and on the day after treatment ended, and then once more, 26 days after the first injection. During the study, no significant differences were found between the two groups with respect to survival rate (treatment group: 68 per cent; control group: 71 per cent, P > 0 4, Fisher-Test) and other clinical findings. Similarly the total leukocyte count, neutrophil count and other haematologic and biochemical parameters did not differ significantly between the groups, based on differences from initial values (P > 0 05). Consequently, the use of rhG-CSF in the treatment of dogs with parvoviral enteritis cannot be recommended.


Subject(s)
Dog Diseases/virology , Enteritis/veterinary , Granulocyte Colony-Stimulating Factor/therapeutic use , Parvoviridae Infections/veterinary , Parvovirus/growth & development , Animals , Dog Diseases/drug therapy , Dogs , Double-Blind Method , Enteritis/drug therapy , Enteritis/virology , Erythrocyte Count/veterinary , Hematocrit/veterinary , Hemoglobins/metabolism , Leukocyte Count/veterinary , Leukopenia/drug therapy , Leukopenia/veterinary , Leukopenia/virology , Parvoviridae Infections/drug therapy , Parvoviridae Infections/virology , Recombinant Proteins/therapeutic use , Statistics, Nonparametric
20.
Acta Vet Hung ; 47(3): 379-94, 1999.
Article in English | MEDLINE | ID: mdl-10497831

ABSTRACT

Parvoviruses have small genomes and, consequently, are highly dependent on their host for various functions in their reproduction. Since these viruses generally use ubiquitous receptors, restrictions are usually intracellularly regulated. A lack of mitosis, and hence absence of enzymes required for DNA replication, is a powerful block of virus infection. Allotropic determinants have been identified for several parvoviruses: porcine parvovirus, canine parvovirus (CPV), feline parvovirus (feline panleukopenia virus), minute virus of mice, Aleutian disease virus, and GmDNV (an insect parvovirus). Invariably, these identifications involved the use of infectious clones of these viruses and the exchange of restriction fragments to create chimeric viruses, of which the resulting phenotype was then established by transfection in appropriate cell lines. The tropism of these viruses was found to be governed by minimal changes in the sequence of the capsid proteins and, often, only 2 or 3 critical amino acids are responsible for a given tropism. These amino acids are usually located on the outside of the capsid near or on the spike of the threefold axis for the vertebrate parvoviruses and on loops 2 or 3 for the insect parvoviruses. This tropism is not mediated via specific cellular receptors but by interactions with intracellular factors. The nature of these factors is unknown but most data point to a stage beyond the conversion of the single-stranded DNA genome by host cell DNA polymerase into monomeric duplex intermediates of the replicative form. The sudden and devastating emergence of mink enteritis virus (MEV) and CPV in the last 50 years, and the possibility of more future outbreaks, demonstrates the importance of understanding parvovirus tropism.


Subject(s)
Parvovirus/growth & development , Tropism , Animals , Cats , Computer Simulation , Dogs , Mice , Parvovirus/physiology , Structure-Activity Relationship , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...