Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Vet Ital ; 59(4)2023 Dec 31.
Article in English | MEDLINE | ID: mdl-38685825

ABSTRACT

Fowl Pox Viruses (FPV) infect chickens and turkeys giving rise to pock lesions on various body parts like combs, wattles, legs, shanks, eyes, mouth etc. The birds, affected with FPV, also show anemia and ruffled appearance which are clinical symptoms of Reticuloendotheliosis. Interestingly, the field strains of FPV are integrated with the provirus of Reticuloendotheliosis Virus (REV). Due to this integration, the infected birds, upon replication of FPV, give rise to free REV virions, causing severe immunosuppression and anemia. Pox scabs, collected from the infected birds, not only show positive PCR results upon performing FPV-specific 4b core protein gene PCR but also show positive results for the PCR of REV-specific env gene and FPV-REV 5'LTR junction. Homogenized suspension of the pock lesions, upon inoculating to the Chorio-allantoic Membrane (CAM) of 10 days old specific pathogen-free embryonated chicken eggs, produces characteristic pock lesions in serial passages. But the lesions also harbor REV mRNA or free virion, which can be identified by performing REV-specific env gene PCR using REV RNA from FPV-infected CAMs. The study suggests successful replication and availability of REV mRNA and free virion alongside the FPV virus, although the CAM is an ill-suited medium for any retroviral (like REV) growth and replication.


Subject(s)
Chickens , Fowlpox virus , Poultry Diseases , Reticuloendotheliosis virus , Animals , Reticuloendotheliosis virus/isolation & purification , Chickens/virology , Poultry Diseases/virology , Fowlpox virus/genetics , Fowlpox virus/isolation & purification , Specific Pathogen-Free Organisms , Chick Embryo , Fowlpox/virology , Chorioallantoic Membrane/virology , Retroviridae Infections/veterinary , Retroviridae Infections/virology
2.
PLoS One ; 16(12): e0261122, 2021.
Article in English | MEDLINE | ID: mdl-34914770

ABSTRACT

Fowlpox (FP) is an economically important viral disease of commercial poultry. The fowlpox virus (FPV) is primarily characterised by immunoblotting, restriction enzyme analysis in combination with PCR, and/or nucleotide sequencing of amplicons. Whole-genome sequencing (WGS) of FPV directly from clinical specimens prevents the risk of potential genome modifications associated with in vitro culturing of the virus. Only one study has sequenced FPV genomes directly from clinical samples using Nanopore sequencing, however, the study didn't compare the sequences against Illumina sequencing or laboratory propagated sequences. Here, the suitability of WGS for strain identification of FPV directly from cutaneous tissue was evaluated, using a combination of Illumina and Nanopore sequencing technologies. Sequencing results were compared with the sequence obtained from FPV grown in chorioallantoic membranes (CAMs) of chicken embryos. Complete genome sequence of FPV was obtained directly from affected comb tissue using a map to reference approach. FPV sequence from cutaneous tissue was highly similar to that of the virus grown in CAMs with a nucleotide identity of 99.8%. Detailed polymorphism analysis revealed the presence of a highly comparable number of single nucleotide polymorphisms (SNPs) in the two sequences when compared to the reference genome, providing essentially the same strain identification information. Comparative genome analysis of the map to reference consensus sequences from the two genomes revealed that this field isolate had the highest nucleotide identity of 99.5% with an FPV strain from the USA (Fowlpox virus isolate, FWPV-MN00.2, MH709124) and 98.8% identity with the Australian FPV vaccine strain (FWPV-S, MW142017). Sequencing results showed that WGS directly from cutaneous tissues is not only rapid and cost-effective but also provides essentially the same strain identification information as in-vitro grown virus, thus circumventing in vitro culturing.


Subject(s)
Chorioallantoic Membrane/virology , Fowlpox virus/isolation & purification , Fowlpox/diagnosis , Genome, Viral , High-Throughput Nucleotide Sequencing/methods , Skin/virology , Whole Genome Sequencing/methods , Animals , Australia , Chick Embryo , Chickens , Fowlpox/virology , Fowlpox virus/classification , Fowlpox virus/genetics , Fowlpox virus/growth & development , Polymorphism, Genetic
3.
Avian Pathol ; 50(1): 61-77, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33034512

ABSTRACT

Inoculation of embryonated chicken eggs has been widely used during the past decades; however, inoculation success rates have not been investigated systematically. In this study named success rates were assessed in brown eggs incubated between 5 and 19 days, which were inoculated with 0.2 ml methylene blue per egg. Inoculations were performed in a simple and fully standardized way. Five embryonic compartments were targeted blindly (amniotic cavity, embryo, allantoic cavity, albumen and yolk) with needles of four different lengths; albumen and yolk were targeted with eggs in upside down position. Three compartments were inoculated within sight (air chamber, chorioallantoic membrane and blood vessel). Twenty embryos were used per incubation day, intended deposition site and needle length. Success rates were assessed by visual inspection after breaking the eggs. The inoculations targeting albumen, yolk, amniotic cavity and embryo yielded low scores. Magnetic resonance imaging was performed to elucidate the reason(s) for these low success rates: needles used were of appropriate length, but embryo and amniotic cavity had variable positions in the eggs, while albumen and yolk rapidly changed position after turning the eggs upside down. The latter led to adjustment of the inoculation method for albumen and yolk. Failures to inoculate compartments within sight were immediately visible; therefore, these eggs could be discarded. Except for the amniotic cavity, full scores (20/20) were obtained for all compartments although not always on every day of incubation. In conclusion, the present study may serve as a guide to more accurately inoculate the various chicken embryo compartments. RESEARCH HIGHLIGHTS Blind inoculation of embryonated egg compartments was successful, except for the amniotic cavity. MRI showed rapid position change of albumen and yolk after turning eggs upside down. In ovo vaccination against Marek's disease might be improved by using 38 mm needles.


Subject(s)
Marek Disease/virology , Ovum/ultrastructure , Allantois/ultrastructure , Allantois/virology , Amnion/ultrastructure , Amnion/virology , Animals , Chick Embryo , Chorioallantoic Membrane/ultrastructure , Chorioallantoic Membrane/virology , Female , Injections , Magnetic Resonance Imaging/veterinary , Male , Methylene Blue , Ovum/virology
4.
Methods Mol Biol ; 2203: 67-74, 2020.
Article in English | MEDLINE | ID: mdl-32833204

ABSTRACT

This chapter reports the high-throughput sequencing protocol for sequencing Coronaviruses and other positive strand viruses to produce a dataset of significant depth of coverage. The protocol describes sequencing of infectious bronchitis virus propagated in embryonated eggs and harvested in the allantoic fluid. The protocol is composed of three main steps-enrichment of the allantoic fluid using ultracentrifugation, extraction of total RNA from allantoic fluid, and library preparation from total RNA to DNA sequencing libraries. The workflow will be suitable for all coronaviruses using high-throughput sequencing platforms.


Subject(s)
Coronavirus/genetics , Whole Genome Sequencing/methods , Animals , Chorioallantoic Membrane/virology , Genome, Viral , High-Throughput Nucleotide Sequencing/methods , Infectious bronchitis virus/genetics , Infectious bronchitis virus/isolation & purification , Workflow
5.
J Virol Methods ; 275: 113752, 2020 01.
Article in English | MEDLINE | ID: mdl-31654683

ABSTRACT

Bovine viral diarrhea virus (BVDV) is a common contaminant of Madin-Darby bovine kidney (MDBK) cells as well as fetal calf serum (FCS). It is pathogenic to cattle and regulatory authorities require that veterinary vaccine stocks are free from BVDV. MDBK cells are used in the generation of recombinant lumpy skin disease virus (LSDV) and have been used for the growth of LSDV vaccines. This paper describes how vaccine stocks can be cleared of BVDV by passage through an avian host, nonpermissive to BVDV, but permissive to LSDV. LSDV vaccine stocks were shown to be cleared of BVDV after passage on the chorioallantoic membranes (CAMs) of fertilized 7-day old hens' eggs. Vaccines were passaged a second time on CAMs before being grown in primary lamb testes (LT) cells. Vaccines retained BVDV-negative status after passage on LT cells.


Subject(s)
Chorioallantoic Membrane/virology , Diarrhea Viruses, Bovine Viral/isolation & purification , Lumpy skin disease virus , Ovum/cytology , Viral Vaccines/analysis , Virus Cultivation/methods , Animals , Cattle , Cell Line , Chickens , Female , Fertilization , Kidney/cytology , Kidney/virology , Ovum/virology , Serum Albumin, Bovine , Viral Vaccines/standards
6.
Br Poult Sci ; 60(6): 729-735, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31328539

ABSTRACT

1. The purpose of this study was to investigate ATP levels and the activities of important enzymes involved in glycolysis and TCA cycle in livers of embryonated chicken eggs infected by infectious bursal disease virus (IBDV).2. Embryonated chicken eggs (9 days) were randomly divided into two groups (50 eggs per group). The first group was inoculated with a very virulent IBDV (vvIBDV) isolate into the chorioallantoic membrane. The second group was maintained as uninfected control eggs and inoculated with physiological saline. Embryo survival was assessed daily, and six embryos were sacrificed at 24, 48, 72, 96, and 120 hpi for examining livers. Viral loads in the livers were evaluated by qRT-PCR. A comparative analysis of markers associated with the regulation of energy metabolism across several functional classes (ATP, pyruvic and lactic acids, mitochondrial protein, NAD+/NADH ratios, and enolase, lactic acid dehydrogenase and the respiratory chain complex I activities) were examined in the context of IBDV infection.3. The results indicated that increases in the enzymatic activities associated with glycolytic metabolism in turn affected the synthesis and cytoplasmic concentrations of ATP at early timepoints in infected chicken embryos. Subsequently, energy metabolism was inhibited through the pathological perturbations of metabolic enzymes and mitochondrial damage, as inferred from reduced ATP generation.4. These results suggested impaired bioenergetics, which may lead to liver dysfunction consequent to IBDV infection, contributing to the disease pathogenesis.


Subject(s)
Energy Metabolism , Infectious bursal disease virus/physiology , Liver/virology , Adenosine Triphosphate/analysis , Adenosine Triphosphate/metabolism , Animals , Chick Embryo , Chorioallantoic Membrane/virology , Cytosol/chemistry , Electron Transport Complex I/analysis , Glycolysis , Infectious bursal disease virus/genetics , Infectious bursal disease virus/pathogenicity , L-Lactate Dehydrogenase/analysis , Lactic Acid/analysis , Liver/embryology , Liver/enzymology , Liver/metabolism , Luminescent Measurements , Mitochondria/chemistry , NAD/analysis , Phosphopyruvate Hydratase/analysis , Proteins/analysis , Proteins/isolation & purification , Pyruvic Acid/analysis , RNA, Viral/analysis , Random Allocation , Specific Pathogen-Free Organisms , Virulence , Virus Replication/physiology
7.
Immunol Lett ; 200: 1-4, 2018 08.
Article in English | MEDLINE | ID: mdl-29886119

ABSTRACT

Sir Mac Farlane Burnet was the most honored of all Australian scientists. In 1960, Burnet shared the Nobel Prize for Medicine with Peter Medawar of Britain for the discovery of acquired immunological tolerance. He developed techniques for growing influenza viruses in the chorioallantoic membrane of the chick embryo. This became a standard laboratory practice. He continued to work with chick embryos long after the use of cell cultures had become general. His virology research resulted in significant discoveries concerning the nature and replication of viruses and their interaction with the immune system.


Subject(s)
Chorioallantoic Membrane/virology , Host-Pathogen Interactions/immunology , Virus Diseases/immunology , Virus Diseases/virology , Virus Physiological Phenomena , Virus Replication , Animals , Australia , Chick Embryo , History, 20th Century , Humans , Immune Tolerance , Immunity , Models, Theoretical , Nobel Prize
8.
Virol J ; 14(1): 120, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28637468

ABSTRACT

BACKGROUND: Influenza virus isolation in embryonated chicken eggs (ECEs) is not applicable for rapid diagnosis, however it allows the recovery and propagation of the viable virus. A low number of infectious virus particles in the swabs, poor quality of samples or individual strain properties can lead to difficulties during the virus isolation process. We propose to utilize chorioallantoic membranes (CAM) of ECEs with the assistance of real-time RT PCR to facilitate equine influenza virus isolation. METHODS: Real-time RT PCR was used to detect influenza virus genetic material in amniotic/allantoic fluids (AF) and CAM of ECEs. Haemagglutination assay was used for AF. We used highly diluted virus as a substitute of clinical specimen for ECEs inoculation. RESULTS: Our study demonstrated that real-time RT PCR testing of CAM homogenates was more useful than testing of AF for EIV detection in ECEs. Positive results from CAM allowed to select the embryos from those with haemagglutination assay (HA) - and real-time RT PCR-negative AF for further passages. Using homogenates of CAM for subsequent passages, we finally obtained HA-positive AF, which confirmed virus replication. CONCLUSION: We postulate that real-time RT PCR testing of CAM homogenates and their subsequent passages may facilitate the isolation of equine influenza viruses.


Subject(s)
Chorioallantoic Membrane/virology , Eggs/virology , Equidae/virology , Orthomyxoviridae/isolation & purification , Virus Cultivation/methods , Animals , Chick Embryo , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
12.
J Virol ; 88(15): 8615-28, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24850732

ABSTRACT

UNLABELLED: Cowpox viruses (CPXV) cause hemorrhagic lesions ("red pocks") on infected chorioallantoic membranes (CAM) of embryonated chicken eggs, while most other members of the genus Orthopoxvirus produce nonhemorrhagic lesions ("white pocks"). Cytokine response modifier A (CrmA) of CPXV strain Brighton Red (BR) is necessary but not sufficient for the induction of red pocks. To identify additional viral proteins involved in the induction of hemorrhagic lesions, a library of single-gene CPXV knockout mutants was screened. We identified 10 proteins that are required for the formation of hemorrhagic lesions, which are encoded by CPXV060, CPXV064, CPXV068, CPXV069, CPXV074, CPXV136, CPXV168, CPXV169, CPXV172, and CPXV199. The genes are the homologues of F12L, F15L, E2L, E3L, E8R, A4L, A33R, A34R, A36R, and B5R of vaccinia virus (VACV). Mutants with deletions in CPXV060, CPXV168, CPXV169, CPXV172, or CPXV199 induced white pocks with a comet-like shape on the CAM. The homologues of these five genes in VACV encode proteins that are involved in the production of extracellular enveloped viruses (EEV) and the repulsion of superinfecting virions by actin tails. The homologue of CPXV068 in VACV is also involved in EEV production but is not related to actin tail induction. The other genes encode immunomodulatory proteins (CPXV069 and crmA) and viral core proteins (CPXV074 and CPXV136), and the function of the product of CPXV064 is unknown. IMPORTANCE: It has been known for a long time that cowpox virus induces hemorrhagic lesions on chicken CAM, while most of the other orthopoxviruses produce nonhemorrhagic lesions. Although cowpox virus CrmA has been proved to be responsible for the hemorrhagic phenotype, other proteins causing this phenotype remain unknown. Recently, we generated a complete single-gene knockout bacterial artificial chromosome (BAC) library of cowpox virus Brighton strain. Out of 183 knockout BAC clones, 109 knockout viruses were reconstituted. The knockout library makes possible high-throughput screening for studying poxvirus replication and pathogenesis. In this study, we screened all 109 single-gene knockout viruses and identified 10 proteins necessary for inducing hemorrhagic lesions. The identification of these genes gives a new perspective for studying the hemorrhagic phenotype and may give a better understanding of poxvirus virulence.


Subject(s)
Chorioallantoic Membrane/pathology , Chorioallantoic Membrane/virology , Cowpox virus/physiology , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Chick Embryo , Cowpox virus/genetics , Gene Knockout Techniques , Hemorrhage/pathology , Hemorrhage/virology , Viral Proteins/genetics , Virulence Factors/genetics
13.
PLoS One ; 7(5): e36531, 2012.
Article in English | MEDLINE | ID: mdl-22606269

ABSTRACT

The lack of affordable techniques for gene transfer in birds has inhibited the advancement of molecular studies in avian species. Here we demonstrate a new approach for introducing genes into chicken somatic tissues by administration of a lentiviral vector, derived from the feline immunodeficiency virus (FIV), into the chorioallantoic membrane (CAM) of chick embryos on embryonic day 11. The FIV-derived vectors carried yellow fluorescent protein (YFP) or recombinant alpha-melanocyte-stimulating hormone (α-MSH) genes, driven by the cytomegalovirus (CMV) promoter. Transgene expression, detected in chicks 2 days after hatch by quantitative real-time PCR, was mostly observed in the liver and spleen. Lower expression levels were also detected in the brain, kidney, heart and breast muscle. Immunofluorescence and flow cytometry analyses confirmed transgene expression in chick tissues at the protein level, demonstrating a transduction efficiency of ∼0.46% of liver cells. Integration of the viral vector into the chicken genome was demonstrated using genomic repetitive (CR1)-PCR amplification. Viability and stability of the transduced cells was confirmed using terminal deoxynucleotidyl transferase (dUTP) nick end labeling (TUNEL) assay, immunostaining with anti-proliferating cell nuclear antigen (anti-PCNA), and detection of transgene expression 51 days post transduction. Our approach led to only 9% drop in hatching efficiency compared to non-injected embryos, and all of the hatched chicks expressed the transgenes. We suggest that the transduction efficiency of FIV vectors combined with the accessibility of the CAM vasculature as a delivery route comprise a new powerful and practical approach for gene delivery into somatic tissues of chickens. Most relevant is the efficient transduction of the liver, which specializes in the production and secretion of proteins, thereby providing an optimal target for prolonged study of secreted hormones and peptides.


Subject(s)
Chick Embryo , Gene Transfer Techniques , Genetic Vectors , Immunodeficiency Virus, Feline/genetics , Animals , Animals, Genetically Modified , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cells, Cultured , Chick Embryo/metabolism , Chick Embryo/virology , Chickens/genetics , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/virology , DNA Primers/genetics , Liver/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tissue Distribution , Transduction, Genetic/methods , alpha-MSH/genetics , alpha-MSH/metabolism
14.
J Comp Pathol ; 146(2-3): 223-9, 2012.
Article in English | MEDLINE | ID: mdl-21705014

ABSTRACT

Inclusion body hepatitis (IBH) associated with fowl adenovirus (FAdV) infection has a worldwide distribution. The aim of the present study was to determine the pathogenicity of Malaysian FAdV serotype 9 (UPM04217) in specific pathogen free (SPF) embryonated chicken embryos. FAdV (titre 10(5.8)/ml) was inoculated into SPF embryonated chicken eggs (0.1 ml per egg) via the chorioallantoic membrane (CAM). There was 100% embryo mortality within 4-11 days post infection (dpi). The gross and microscopical lesions of the embryo were confined to the liver and were noted at 5, 7, 9 and 11 dpi. The liver was pale with multifocal areas of necrosis, fibrosis and haemorrhage. Microscopically, there was moderate to severe congestion and haemorrhage and severe and diffuse hepatocyte degeneration and necrosis, with intranuclear inclusion bodies (INIBs) and associated inflammation. Haemorrhage, congestion, degeneration, necrosis and hyperplasia of the CAM with INIBs were observed at 5, 7, 9 and 11 dpi. Varying degrees of congestion, haemorrhage, degeneration and necrosis were also observed in the yolk sac, kidney, spleen, heart and bursa of Fabricius. Ultrastructurally, numerous viral particles in the nucleus of hepatocytes were recorded at 7, 9 and 11 dpi, whereas at 5 dpi, fine granular and filamentous INIBs were observed. The INIBs in the CAM were present either as fine granular filamentous structures or as large viral inclusions. FAdV (UPM04217) is therefore highly pathogenic to SPF chicken embryos and the embryonic liver should be used for isolation and propagation of the virus.


Subject(s)
Adenoviridae Infections/veterinary , Aviadenovirus/pathogenicity , Chickens/virology , Hepatitis, Viral, Animal/virology , Liver/virology , Poultry Diseases/virology , Adenoviridae Infections/pathology , Adenoviridae Infections/virology , Animals , Chick Embryo , Chorioallantoic Membrane/pathology , Chorioallantoic Membrane/virology , Hepatitis, Viral, Animal/pathology , Inclusion Bodies, Viral/pathology , Inclusion Bodies, Viral/virology , Liver/pathology , Poultry Diseases/pathology
15.
Cancer Gene Ther ; 19(1): 58-68, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22015640

ABSTRACT

Oncolytic adenoviruses are promising anticancer agents. To study and optimize their tumor-killing potency, genuine tumor models are required. Here we describe the use of the chicken chorioallantoic membrane (CAM) tumor model in studies on oncolytic adenoviral vectors. Suspensions of human melanoma, colorectal carcinoma and glioblastoma multiforme cell lines were grafted on the CAM of embryonated chicken eggs. All cell lines tested formed 5-10 mm size tumors, which recapitulated hallmarks of corresponding human specimens. Furthermore, melanoma tumors were injected with adenoviral vector-carrying gene encoding the fusion protein of parainfluenza virus type 5. This led to the induction of cell fusion and syncytia formation in the infected cells. At 6 days post-injection, histological and immunohistochemical analyses of tumor sections confirmed adenovirus replication and syncytia formation. These results demonstrate that the CAM model allows rapid assessment of oncolytic viruses in three-dimensional tumors. Hence, this model constitutes an easy and affordable system for preclinical characterization of viral oncolytic agents that may precede the mandatory process of animal testing. Application of this model will help reducing the use of human xenografts in mice for preclinical evaluation of oncolytic viruses and other anticancer agents.


Subject(s)
Chorioallantoic Membrane/virology , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Adenoviridae/genetics , Animals , Cell Line, Tumor , Chickens , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Colorectal Neoplasms/virology , Genetic Vectors/genetics , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Glioblastoma/virology , Humans , Immunohistochemistry , Melanoma/genetics , Melanoma/pathology , Melanoma/therapy , Melanoma/virology
16.
Virus Res ; 163(1): 254-61, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22016035

ABSTRACT

It was previously reported that cowpox virus (CPXV) strain Brighton Red (BR) causes red pocks upon inoculation of chorioallantoic membranes (CAMs) of embryonated chicken eggs. Red pocks are characterized by hemorrhage and reduced numbers of inflammatory cells while white pocks induced by other members of the genus Orthopoxvirus lack hemorrhage and have higher numbers of infiltrating heterophils. Analyses of CPXV BR white pock variants identified the cytokine response modifier A (CrmA) as the factor responsible for the differences in pock phenotype through induction of hemorrhage and inhibition of chemotaxis. In the present study CPXV crmA deletion mutants were generated based on a full-length bacterial artificial chromosome clone of CPXV BR (pBR). Deletion of the first crmA start codon was sufficient to abolish protein expression, whereas modification of a potential second start codon had no impact on CrmA production as shown by Western blot analysis. Immunohistochemistry of CAMs inoculated with crmA-positive BR viruses showed accumulation of viral antigen in endothelial cells, which was consistent with the red pock phenotype. On the other hand, crmA-negative mutants were characterized by the induction of white pocks and the absence of CPXV antigen in endothelia. The introduction of the complete CPXV BR crmA gene into the homologous genome region of the attenuated vaccinia virus strain MVA (modified vaccinia virus Ankara), however, resulted in CrmA production but not the red pock phenotype. We therefore conclude that (i) CPXV CrmA is associated with increased accumulation of virus in endothelial cells and (ii) the poxvirus-encoded serpin is necessary but not sufficient for the red pock phenotype and the anti-chemotactic capabilities on CAMs.


Subject(s)
Chorioallantoic Membrane/pathology , Chorioallantoic Membrane/virology , Cowpox virus/pathogenicity , Serpins/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Blotting, Western , Chick Embryo , Chickens , Cowpox virus/genetics , Endothelial Cells/virology , Immunohistochemistry , Sequence Deletion , Serpins/genetics , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viral Proteins/genetics , Virulence Factors/genetics
17.
Proc Natl Acad Sci U S A ; 108(35): 14389-96, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21813762

ABSTRACT

One hundred years ago Peyton Rous recovered a virus, now known as the Rous sarcoma virus (RSV), from a chicken sarcoma, which reproduced all aspects of the tumor on injection into closely related chickens. There followed recovery of causal viruses of tumors of different morphology from 4 more of 60 chicken tumors. Subsequent studies in chickens of the biology of the first RSV isolated moved slowly for 45 y until an assay of ectodermal pocks of the chorioallantoic membrane of chicken embryos was introduced. The inadequacies of that assay were resolved with the production of transformed foci in cultures of chicken fibroblasts. There followed a productive period on the dynamics of RSV infection. An avian leukosis virus (ALV) was found in some chicken embryos and named resistance-inducing factor (RIF) because it interferes with RSV. Its epidemiology in chickens is described. Another ALV was found in stocks of RSV and called Rous-associated virus (RAV). Cells preinfected with RAV interfere with RSV infection, but RSV does not produce infectious virus unless RAV is added during or after RSV infection. Intracellular RAV provides the infectious coat for the otherwise defective RSV. The coat determines the antigenicity, host range, and maturation rate of RSV. RSV particles carry reverse transcriptase, an enzyme that converts their RNA into DNA and allows integration into the cell's DNA, where it functions as a cellular gene. This was the bridge that joined the biological era to the molecular era. Its relation to oncogenes and human cancer is discussed.


Subject(s)
Avian Leukosis Virus/isolation & purification , Rous sarcoma virus/isolation & purification , Animals , Chick Embryo , Chickens , Chorioallantoic Membrane/virology , Genetic Predisposition to Disease , Helper Viruses/isolation & purification , Humans , Mice , Proviruses/isolation & purification , Sarcoma, Avian/epidemiology , Sarcoma, Avian/virology , Virion/isolation & purification
18.
Virol J ; 8: 190, 2011 Apr 25.
Article in English | MEDLINE | ID: mdl-21518442

ABSTRACT

BACKGROUND: Marek's disease virus (MDV), which is widely considered to be a natural model of virus-induced lymphoma, has the potential to cause tremendous losses in the poultry industry. To investigate the structural basis of MDV membrane fusion and to identify new viral targets for inhibition, we examined the domains of the MDV glycoproteins gH and gB. RESULTS: Four peptides derived from the MDV glycoprotein gH (gHH1, gHH2, gHH3, and gHH5) and one peptide derived from gB (gBH1) could efficiently inhibit plaque formation in primary chicken embryo fibroblast cells (CEFs) with 50% inhibitory concentrations (IC50) of below 12 µM. These peptides were also significantly able to reduce lesion formation on chorioallantoic membranes (CAMs) of infected chicken embryos at a concentration of 0.5 mM in 60 µl of solution. The HR2 peptide from Newcastle disease virus (NDVHR2) exerted effects on MDV specifically at the stage of virus entry (i.e., in a cell pre-treatment assay and an embryo co-treatment assay), suggesting cross-inhibitory effects of NDV HR2 on MDV infection. None of the peptides exhibited cytotoxic effects at the concentrations tested. Structural characteristics of the five peptides were examined further. CONCLUSIONS: The five MDV-derived peptides demonstrated potent antiviral activity, not only in plaque formation assays in vitro, but also in lesion formation assays in vivo. The present study examining the antiviral activity of these MDV peptides, which are useful as small-molecule antiviral inhibitors, provides information about the MDV entry mechanism.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , Glycoproteins/pharmacology , Mardivirus/growth & development , Marek Disease/prevention & control , Peptides/pharmacology , Viral Structural Proteins/pharmacology , Animals , Biological Products/genetics , Cells, Cultured , Chick Embryo , Chorioallantoic Membrane/pathology , Chorioallantoic Membrane/virology , Fibroblasts/virology , Glycoproteins/genetics , Inhibitory Concentration 50 , Mardivirus/drug effects , Mardivirus/genetics , Marek Disease/virology , Microbial Sensitivity Tests/methods , Newcastle disease virus/genetics , Peptides/genetics , Viral Plaque Assay , Viral Structural Proteins/genetics
19.
Mol Ther ; 17(6): 1012-21, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19277014

ABSTRACT

Systemically administered vectors must cross the endothelial lining of tumor blood vessels to access cancer cells. Vectors that interact with markers on the lumenal surface of these endothelial cells might have enhanced tumor localization. Here, we generated oncolytic measles viruses (MVs) displaying alpha(v)beta(3) integrin-binding peptides, cyclic arginine-glycine-aspartate (RGD) or echistatin, on the measles hemagglutinin protein. Both viruses had expanded tropisms, and efficiently entered target cells via binding to integrins, but also retained their native tropisms for CD46 and signaling lymphocyte activation molecule (SLAM). When fluorescently labeled and injected intravascularly into chick chorioallantoic membranes (CAMs), in contrast to unmodified viruses, the integrin-binding viral particles bound to the lumenal surface of the developing chick neovessels and infected the CAM vascular endothelial cells. In a mouse model of VEGF-induced angiogenesis in the ear pinna, the integrin-binding viruses, but not the parental virus, infected cells at sites of new blood vessel formation. When given intravenously to mice bearing tumor xenografts, the integrin-binding virus infected endothelial cells of tumor neovessels in addition to tumor parenchyma. To our knowledge, this is the first report demonstrating that oncolytic MVs can be engineered to target the lumenal endothelial surface of newly formed blood vessels when administered intravenously in living animals.


Subject(s)
Endothelial Cells/virology , Measles virus/physiology , Peptides/metabolism , Adenoviridae/genetics , Animals , Cell Line , Chick Embryo , Chickens , Chlorocebus aethiops , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/virology , Female , Humans , Immunoblotting , Immunohistochemistry , Integrin alphaVbeta3/metabolism , Intercellular Signaling Peptides and Proteins , Measles virus/genetics , Measles virus/metabolism , Mice , Mice, Nude , NIH 3T3 Cells , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology , Vero Cells , Xenograft Model Antitumor Assays
20.
Virol J ; 6: 15, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19196466

ABSTRACT

BACKGROUND: Infectious bronchitis virus primarily induces a disease of the respiratory system, different IBV strains may show variable tissue tropisms and also affect the oviduct and the kidneys. Proventriculitis was also associated with some new IBV strains. Aim of this study was to investigate by immunohistochemistry (IHC) the tissue tropism of avian infectious bronchitis virus (IBV) strain M41 in experimentally infected chicken embryos. RESULTS: To this end chicken embryos were inoculated in the allantoic sac with 10(3) EID(50) of IBV M41 at 10 days of age. At 48, 72, and 120 h postinoculation (PI), embryos and chorioallantoic membranes (CAM) were sampled, fixed, and paraffin-wax embedded. Allantoic fluid was also collected and titrated in chicken embryo kidney cells (CEK). The sensitivity of IHC in detecting IBV antigens in the CAM of inoculated eggs matched the virus reisolation and detection in CEK. Using IHC, antigens of IBV were detected in nasal epithelium, trachea, lung, spleen, myocardial vasculature, liver, gastrointestinal tract, kidney, skin, sclera of the eye, spinal cord, as well as in brain neurons of the inoculated embryos. These results were consistent with virus isolation and denote the wide tissue tropism of IBV M41 in the chicken embryo. Most importantly, we found infection of vasculature and smooth muscle of the proventriculus which has not seen before with IBV strain M41. CONCLUSION: IHC can be an additional useful tool for diagnosis of IBV infection in chickens and allows further studies to foster a deeper understanding of the pathogenesis of infections with IBV strains of different virulence. Moreover, these results underline that embryonic tissues in addition to CAM could be also used as possible source to generate IBV antigens for diagnostic purposes.


Subject(s)
Antigens, Viral/analysis , Chick Embryo/virology , Immunohistochemistry/methods , Infectious bronchitis virus/pathogenicity , Nervous System/virology , Proventriculus/virology , Animals , Cells, Cultured , Chickens , Chorioallantoic Membrane/virology , Infectious bronchitis virus/isolation & purification , Kidney/cytology , Kidney/virology , Nervous System/pathology , Organ Specificity , Poultry Diseases/pathology , Poultry Diseases/virology , Proventriculus/pathology , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...