Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 295
Filter
1.
Adv Exp Med Biol ; 1451: 55-74, 2024.
Article in English | MEDLINE | ID: mdl-38801571

ABSTRACT

The complex cytoplasmic DNA virus known as the fowlpox virus (FWPV) is a member of the avipoxvirus genus, Subfamily Chordopoxvirinae, and Family Poxviridae. The large genome size of FWPV makes it a potential vector for the creation of vaccines against a range of serious veterinary and human ailments. It also allows for multiple gene insertion and the generation of abortive infection in mammalian cells. The virus, which causes fowlpox in chickens and turkeys, is mainly transmitted to poultry through aerosols or biting insects. Fowlpox is a highly contagious disease that affects both domestic and wild birds, causing cutaneous and/or diphtheritic illnesses. To control the illness, strict hygiene practices and immunization with FWPV attenuated strains or antigenically similar pigeon pox virus vaccines are employed. Recent years have seen an increase in fowlpox outbreaks in chicken flocks, primarily due to the introduction of novel forms of FWPV. It is believed that the pathogenic characteristics of these strains are enhanced by the integration of reticuloendotheliosis virus sequences of variable lengths into the FWPV genome. The standard laboratory diagnosis of FPV involves histopathological analysis, electron microscopy, virus isolation on chorioallantoic membrane (CAM) of embryonated chicken eggs or cell cultures, and serologic techniques. For quick and consistent diagnosis, polymerase chain reaction (PCR) has proven to be the most sensitive method. PCR is used in concert with restriction endonuclease enzyme analysis (REA) to identify, differentiate, and characterize the molecular makeup of isolates of the fowlpox virus. Sequencing of the amplified fragments is then done.


Subject(s)
Fowlpox virus , Fowlpox , Fowlpox virus/genetics , Animals , Fowlpox/virology , Chickens/virology , Genome, Viral
2.
Avian Dis ; 68(1): 33-37, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38687105

ABSTRACT

The aim of this study was to develop a multiplex PCR assay capable of rapidly differentiating two major Avipoxvirus (APV) species, Fowlpox virus (FWPV) and Pigeonpox virus (PGPV), which cause disease in bird species. Despite the importance of a rapid differentiation assay, no such assay exists that can differentiate the APV species without sequencing. To achieve this, species-specific target DNA fragments were selected from the fpv122 gene of FWPV and the HM89_gp120 gene of PGPV, which are unique to each genome. Nine samples collected from unvaccinated chickens, pigeons, and a turkey with typical pox lesions were genetically identified as FWPV and PGPV. The designed primers and target DNA fragments were validated using in silico analyses with the nucleotide Basic Local Alignment Search Tool. The multiplex PCR assay consisted of species-specific primers and previously described PanAPV primers (genus-specific) and was able to differentiate FWPV and PGPV, consistent with the phylogenetic outputs. This study represents the first successful differentiation of FWPV and PGPV genomes using a conventional multiplex PCR test. This assay has the potential to facilitate the rapid diagnosis and control of APV infections.


Desarrollo de un ensayo de PCR múltiple para la diferenciación rápida de los virus de la viruela aviar y la viruela de paloma. El objetivo de este estudio fue desarrollar un ensayo de PCR múltiple capaz de diferenciar rápidamente dos especies principales de Avipoxvirus (APV) (viruela del pollo), el Fowlpox virus (FWPV) y el Pigeonpox virus (PGPV), (viruela de la gallina), que causan enfermedades en especies de aves. A pesar de la importancia de un ensayo de diferenciación rápida, no existe ningún ensayo que pueda diferenciar las especies de APV sin secuenciación. Para lograr esto, se seleccionaron fragmentos blanco de ADN específicos de especie del gene fpv122 de FWPV y el gene HM89_gp120 de Pigeonpox virus, que son únicos para cada genoma. Nueve muestras recolectadas de pollos, palomas y un pavo que no fueron vacunados con lesiones típicas de la viruela se identificaron genéticamente como FWPV y PGPV. Los iniciadores diseñados y los fragmentos de ADN blanco se validaron mediante análisis in silico mediante la herramienta de búsqueda de alineación local básica de nucleótidos (BLAST). El ensayo de PCR múltiple consistió en iniciadores específicos de especie y cebadores PanAPV previamente descritos (específicos de género) y fue capaz de diferenciar entre Fowlpox virus y Pigeonpox virus, de acuerdo con los resultados filogenéticos. Este estudio representa la primera diferenciación exitosa de los genomas de Fowlpox virus y Pigeonpox virus utilizando una prueba de PCR múltiple convencional. Este ensayo tiene el potencial de facilitar el diagnóstico rápido y el control de las infecciones por Avipoxvirus.


Subject(s)
Avipoxvirus , Chickens , Columbidae , Fowlpox virus , Multiplex Polymerase Chain Reaction , Poultry Diseases , Poxviridae Infections , Animals , Multiplex Polymerase Chain Reaction/veterinary , Multiplex Polymerase Chain Reaction/methods , Fowlpox virus/genetics , Fowlpox virus/isolation & purification , Poxviridae Infections/veterinary , Poxviridae Infections/virology , Poxviridae Infections/diagnosis , Poultry Diseases/virology , Poultry Diseases/diagnosis , Avipoxvirus/genetics , Avipoxvirus/isolation & purification , Avipoxvirus/classification , Turkeys , Fowlpox/virology , Fowlpox/diagnosis , Species Specificity , Phylogeny , Bird Diseases/virology , Bird Diseases/diagnosis
3.
Poult Sci ; 102(8): 102769, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37321035

ABSTRACT

A total of 45 samples of vaccinated and non-vaccinated layer chickens were collected from farms in the Egyptian governorates of Sharqia, Ismailia, Menofia, Gharbia, Kafr El Sheikh, Qalyubia, and Dakahlia in the year 2022. They exhibited nodular lesions on their combs, mouth corners, and eyelids, suggesting they were infected with pox disease, which was associated with a 3 to 5% mortality rate. The samples were grown on the chorioallantoic-membrane of embryonated chicken eggs to ensure their viability. In both vaccinated and non-vaccinated farms, 35 of 45 virus isolates were confirmed positive via polymerase chain reaction (PCR) of fpv167 (P4b), based on the amplicon length of the fpv167 gene locus. The 6 strains from various Egyptian governorates were chosen for sequencing and genetic characterization. Phylogenetic investigation of the fpv167 (P4b) gene of sequenced strains clustered within sub clade A1 showed 100% correlation between FWPVD, TKPV13401 and fowlpox-AN2, fowlpox-AN3, and fowlpox-AN6, but only a 98.6% correlation between fowlpox-AN1, fowlpox-AN4, and fowlpox-AN5. Comparing the fowlpox-AN1, fowlpox-AN4, and fowlpox-AN5 strains with commercial vaccine strains (HP1-444-(FP9), vaccine-VSVRI), they had 98.6% identity, while other strains had 100% identity. The results of this study's mutation research showed that fowlpox-AN1, fowlpox-AN4, and fowlpox-AN5 had acquired novel mutations; fowlpox-AN1 had R201G and T204A; fowlpox-AN4 and fowlpox-AN5 had L141F and H157P. Further research is required to determine the effectiveness of the current vaccine in order to develop a new vaccine.


Subject(s)
Fowlpox virus , Fowlpox , Poultry Diseases , Animals , Fowlpox virus/genetics , Chickens , Egypt , Phylogeny , Genomics
4.
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
5.
Vet Ital ; 59(4)2023 Dec 31.
Article in English | MEDLINE | ID: mdl-38756026

ABSTRACT

Fowlpox virus (FPV) infects 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 a 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-day-old specific pathogen-free embryonated chicken eggs, produces characteristic pock lesions in serial passages. However, 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, although the CAM is an ill-suited medium for any retroviral (like REV) growth and replication.


Subject(s)
Reverse Transcriptase Polymerase Chain Reaction , Animals , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Diarrhea/veterinary , Diarrhea/virology , India , Fowlpox virus/genetics , Fowlpox/virology , Sheep , Goat Diseases/virology , Turkeys/virology , Goats , Chickens/virology , Sheep Diseases/virology , Poultry Diseases/virology
6.
Microbiol Spectr ; 10(5): e0141522, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36073826

ABSTRACT

To determine the genomic variations of fowlpox virus (FPV)-the largest, very ancient, and still harmful avian virus-the complete genomes of 21 FPVs were analyzed. The genomes showed low genetic diversity relative to their overall size. Our studies revealed that FPVs could phylogenetically be divided into two clades, based on their regional distribution, and comparative analysis showed that 40 putative proteins of FPV were associated with geographic differences in viruses, viral pathogenicity, or the onset of diphtheritic lesions. The strain, classified into a subgroup different from others in the genomic analysis, showed relatively low pathogenicity in chickens, and the onset of diphtheritic lesions was observed to be caused only by the specific strain. Despite genetic differences, some commercial vaccines are protective against virulent strains, and intact reticuloendotheliosis virus inserted into field FPV strains was activated but there was no enhancement of the pathogenicity of FPV. These findings will expand our knowledge of the viral proteome and help us understand the pathogenicity of FPV. IMPORTANCE This study aims at determining molecular candidates using comparative genomics to differentiate between the diphtheritic and cutaneous forms of FPV infection, in addition to their association with the pathogenicity of the virus. Full-genomic analyses of multiple fowlpox strains, including field viruses, isolated between 1960s and 2019, and vaccine strains showed the genetic diversity due to regional differences. Comparative genomic analysis offered the clues related to viral virulence. We believe that our study makes a significant contribution to the literature because we are the first to perform such an elaborate study that compares 21 FPVs to study and highlight their diversity, despite the high level of homology between them. Our results shall help provide insights for tackling FPV that has been taking a toll on the poultry for years now.


Subject(s)
Fowlpox virus , Vaccines , Animals , Fowlpox virus/genetics , Virulence/genetics , Proteome/genetics , Chickens , Genetic Variation
7.
Viruses ; 14(4)2022 04 13.
Article in English | MEDLINE | ID: mdl-35458529

ABSTRACT

Reticuloendotheliosis virus (REV) is a retroviral pathogen capable of infecting several avian hosts and is associated with immunosuppression, anemia, proventriculitis, neoplasia, and runting-stunting syndrome. Its genome contains the three major genes, gag, pol, and env, and two flanking long terminal repeat (LTR) regions. Complete genome sequences of REV are limited in terms of geographical origin. The aim of this study was to characterize the complete genome of REV detected in Brazilian chickens with multiple viral coinfections and analyze the polymorphisms in the deduced amino acids sequences corresponding to its encoded proteins. We tested the presence and completeness of REV as well as other viral pathogens in samples from Brazilian poultry farms by qPCR. The complete genomes of two REV strains were sequenced by overlapping fragments through the dideoxy method. Phylogenetic analysis, pairwise identity matrix, polymorphism identification and protein modeling were performed along the entire genome. We detected REV in 65% (26/40) of the tested samples. Concomitant viral infections were detected in 82.5% (33/40) of the samples and in 90% (9/10) of the farms. Multiple infections included up to seven viruses. Phylogenetic analysis classified both Brazilian strains into REV subtype 3, and the pairwise comparison indicated that strains from the USA and fowlpox virus (FWPV)-related strains were the most identical. The subdomain p18 in gag, the reverse transcriptase/ribonuclease H in pol, and the surface (SU) in the env protein were the most polymorphic in genomic comparisons. The relevant motifs for each protein were highly conserved, with fewer polymorphisms in the fusion peptide, immunosuppression domain, and disulfide bonds on the surface (SU) and transmembrane (TM) of env. This is the first study to include complete genomes of REV in Brazil and South America detected in farms with multiple viral coinfections. Our findings suggest an involvement of REV as an immunosuppressor and active agent in the emergence and progression of multiple infectious diseases. We also found a possible etiological relationship between Brazilian strains and the USA and FWPV recombinant strains. This information highlights the need for epidemiological vigilance regarding REV in association with another pathogens.


Subject(s)
Coinfection , Fowlpox virus , Poultry Diseases , Reticuloendotheliosis virus , Animals , Brazil/epidemiology , Chickens/genetics , Coinfection/genetics , Coinfection/veterinary , Fowlpox virus/genetics , Genome, Viral , Phylogeny , Reticuloendotheliosis virus/genetics
8.
Transbound Emerg Dis ; 69(5): 2913-2923, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34974640

ABSTRACT

Cutaneous fowlpox is a disease of chickens and turkeys caused by the fowlpox virus (FWPV), characterized by the development of proliferative lesions and scabs on unfeathered areas. FWPVs regularly carry an integrated, active copy of the reticuloendotheliosis virus (REV), and it has been hypothesized that such FWPVs are more problematic in the field. Extensive outbreaks are usually observed in tropical and sub-tropical climates, where biting insects are more difficult to control. Here, we report an epidemic of 65 cutaneous fowlpox cases in Austria in layer chickens (91% of the cases) and broiler breeders and turkeys, all of them unvaccinated against the disease, from October 2018 to February 2020. The field data revealed appearance in flocks of different sizes ranging from less than 5000 birds up to more than 20,000 animals, with the majority raised indoors in a barn system. The clinical presentation was characterized by typical epithelial lesions on the head of the affected birds, with an average decrease of 6% in egg production and an average weekly mortality of 1.2% being observed in the flocks. A real-time multiplex polymerase chain reaction (PCR) confirmed the presence of FWPV-REV DNA, not only in the lesions but also in the environmental dust from the poultry houses. The integration of the REV provirus into the FWPV genome was confirmed by PCR, and revealed different FWPV genome populations carrying either the REV long terminal repeats (LTRs) or the full-length REV genome, reiterating the instability of the inserted REV. Two selected samples were fully sequenced by next generation sequencing (NGS), and the whole genome phylogenetic analysis revealed a regional clustering of the FWPV genomes. The extensive nature of these outbreaks in host populations naïve for the virus is a remarkable feature of the present report, highlighting new challenges associated with FWPV infections that need to be considered.


Subject(s)
Fowlpox virus , Fowlpox , Poultry Diseases , Reticuloendotheliosis virus , Animals , Austria/epidemiology , Chickens , Dust , Fowlpox/epidemiology , Fowlpox virus/genetics , Phylogeny , Poultry Diseases/epidemiology , Reticuloendotheliosis virus/genetics , Turkeys
9.
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
10.
Poult Sci ; 100(4): 100986, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33647723

ABSTRACT

Infectious laryngotracheitis (ILT), fowlpox (FP), and reticuloendotheliosis are important poultry diseases caused by gallid herpesvirus 1 (ILTV), fowlpox virus (FWPV), and reticuloendotheliosis virus (REV), respectively. Coinfections with ILTV and FWPV occur naturally in chickens, and FP in its more virulent wet form is characterized by diphtheritic lesions and easily confused with ILT. Moreover, the insertion of only partial REV-LTR or a nearly full-length REV into the FWPV genome, located between the ORF 201 and ORF 203, has increased recently in wild-type field FWPV isolates. Therefore, it is critical to detect ILTV, FWPV, REV-integrated FWPV, and REV early and accurately. In this study, we successfully developed a multiplex PCR assay for the simultaneous detection of ILTV, FWPV, REV-integrated FWPV, and REV, and the detection limits was 1 × 54 copies/tube. When used to test clinical samples, the results of the multiplex PCR were in 100% agreement with singleplex PCRs and sequencing. This new multiplex PCR is a simple, rapid, sensitive, specific, and cost-effective method for detection of 4 viruses in clinical specimens.


Subject(s)
Coinfection , Fowlpox , Herpesviridae Infections , Multiplex Polymerase Chain Reaction , Poultry Diseases , Retroviridae Infections , Animals , Chickens , Coinfection/veterinary , Coinfection/virology , Fowlpox/complications , Fowlpox/diagnosis , Fowlpox virus/genetics , Herpesviridae Infections/complications , Herpesviridae Infections/diagnosis , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Limit of Detection , Multiplex Polymerase Chain Reaction/economics , Multiplex Polymerase Chain Reaction/standards , Multiplex Polymerase Chain Reaction/veterinary , Poultry Diseases/diagnosis , Poultry Diseases/virology , Reproducibility of Results , Reticuloendotheliosis virus/genetics , Retroviridae Infections/complications , Retroviridae Infections/diagnosis , Retroviridae Infections/veterinary
11.
Arch Virol ; 166(5): 1485-1488, 2021 May.
Article in English | MEDLINE | ID: mdl-33620554

ABSTRACT

Fowlpox virus (FWPV), which is the type member of the genus Avipoxvirus, subfamily Chordopoxvirinae, family Poxviridae, can lead to significant losses to the poultry industry. Although a large number of fowlpox virus genomes have been sequenced and characterised globally, there are no sequences available at the genomic level from Australian isolates. Here, we present the first complete genome sequence of a fowlpox virus vaccine strain (FWPV-S) containing an integrated near-full-length reticuloendotheliosis virus (REV) provirus. The genome of FWPV-S showed the highest sequence similarity to a fowlpox virus from the USA (97.74% identity). The FWPV-S genome contained 16 predicted unique genes, while a further two genes were fragmented compared to previously reported FWPV genome sequences. Subsequent phylogenetic analysis showed that FWPV-S was most closely related to other fowlpox viruses. This is the first reported genome sequence of FWPV from Australia.


Subject(s)
Fowlpox virus/genetics , Proviruses/genetics , Reticuloendotheliosis virus/genetics , Viral Vaccines/genetics , Animals , Australia , Base Sequence , Cells, Cultured , Chick Embryo , DNA, Viral/genetics , Fowlpox virus/classification , Fowlpox virus/isolation & purification , Genes, Viral , Genome, Viral/genetics , Open Reading Frames , Phylogeny , Viral Vaccines/classification , Viral Vaccines/isolation & purification , Virus Integration
12.
Arch Razi Inst ; 75(4): 501-508, 2021 01.
Article in English | MEDLINE | ID: mdl-33403845

ABSTRACT

Fowlpox is an economically significant viral disease in poultry, characterized by two forms of clinical signs, including cutaneous and diphtheritic lesions. This infection can have several adverse effects on flock performance, such as a reduction in egg production and growth and an increase in mortality. In winter 2018, an infection suspected to fowlpox was reported from a Hy-line W-36 laying farm in Isfahan province, Iran. The birds were 38 weeks of age and showed obvious diphtheritic signs in mucous membranes with increased mortality and reduced egg production. In total, 20 samples were collected from diphtheritic lesions (Trachea and Esophagus) of infected birds. The Polymerase Chain Reaction method was used to amplify a 578 bp fragment of the poxvirus 4b core protein gene. Phylogenetic relationships of avian poxviruses are usually analyzed using the 4b core protein-coding gene sequences with molecular weights of 75.2 kDa. The major elements had the fowlpox genome, and sequencing was performed for one isolate as representative. The nucleotide sequence result showed that this isolate (FP\UT-POX-2018) had a similarity rate of 99.53% with the previous Iranian fowlpox isolate (FP\GHPCRLAB.3) sequenced in the GenBank.Moreover, there was a 100% similarity among the current isolate nucleotide sequence, FP/NobilisVarioleW, and FP/FPV-VR250. The derived phylogenetic tree showed that these isolates were clustered in A1 subclades. Therefore, Iranian isolates of fowlpox virus have remained in the same subclade of phylogenetic classification (subclade A1), and they show high genomic similarity with previous isolates of Iran. Veterinarians and farmers must not underestimate fowlpox. However, they should consider the importance of vaccination against this disease like any other disease care.


Subject(s)
Chickens , Disease Outbreaks/veterinary , Fowlpox virus/isolation & purification , Fowlpox/epidemiology , Poultry Diseases/epidemiology , Animals , Female , Fowlpox/virology , Fowlpox virus/classification , Fowlpox virus/genetics , Iran/epidemiology , Phylogeny , Polymerase Chain Reaction/veterinary , Poultry Diseases/virology
13.
Avian Pathol ; 50(2): 109-111, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33464927

ABSTRACT

The use of novel vector vaccines (viral, bacterial and apicomplexan) can have a significant impact on the control of poultry disease. They offer a cost effective, convenient and effective means of mass vaccine delivery combined with the ability to switch on both antibody and cell-mediated immunity. In addition, recent viral vector constructs have enabled farmers to vaccinate against up to three important pathogens with a single in ovo administration. As the technology develops, it is likely that this means of vaccine administration will be utilized further and it will play a key role in the control of both existing and new emerging diseases of poultry in the future.


Subject(s)
Communicable Diseases, Emerging/prevention & control , Eimeria/immunology , Fowlpox virus/immunology , Poultry Diseases/prevention & control , Salmonella/immunology , Vaccines/administration & dosage , Animals , Communicable Diseases, Emerging/pathology , Fowlpox virus/genetics , Genetic Vectors , Immunity, Cellular , Immunity, Humoral , Poultry , Poultry Diseases/pathology , Vaccination/veterinary , Vaccines, Synthetic
14.
Dev Comp Immunol ; 114: 103850, 2021 01.
Article in English | MEDLINE | ID: mdl-32918930

ABSTRACT

The human IL-1 receptor family is comprised of 11 membrane bound or soluble receptors and the IL-18 binding protein (IL-18BP). These receptors are dispersed across seven genomic loci, with the majority at a single locus. Direct orthologues were identified in the chicken at conserved genomic loci; however, the IL-18BP remained absent from the first four builds of the chicken genome sequence. Subsequent assemblies identified the gene at a locus syntenic with mammals; however, these predicted sequences differed between genome builds and contained multiple errors. A partial IL-18BP-like sequence in the NCBI EST database was used to clone the full-length cDNA. A splice variant, which lacks the exon that encodes part of the signal peptide, was also cloned. Human IL-18BP is differentially spliced to produce a number of variants, which are all secreted. By contrast, the spliced chicken isoform was predicted to be intracellular, and we identified similar variants with the same exon missing in a limited number of divergent vertebrate species. Mammalian and viral IL-18BPs inhibit IL-18 activity by directly binding to this cytokine. Full-length and intracellular chicken IL-18BPs were equally effective at inhibiting IL-18-mediated IFN-γ release from an avian B-cell line. Analysis of the predicted chIL-18BP protein sequence revealed two crucial residues, which account for 50% of the binding affinity between human IL-18 and IL-18BP, are conserved in the chicken and a fowlpox-encoded homologue, fpv214. This suggests specific fowlpox viruses used in humans as a vaccine vector have the potential to dampen anti-viral host immune responses.


Subject(s)
Avian Proteins/genetics , B-Lymphocytes/immunology , Chickens/immunology , Fowlpox virus/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-18/metabolism , Protein Isoforms/genetics , Viral Proteins/metabolism , Animals , Avian Proteins/metabolism , Cell Line , Cloning, Molecular , Fowlpox virus/genetics , Genetic Loci/genetics , Genetic Vectors/genetics , Host-Pathogen Interactions , Immunomodulation , Intercellular Signaling Peptides and Proteins/metabolism , Interferon-gamma/metabolism , Lymphocyte Activation , Mammals , Protein Binding , Synteny , Viral Proteins/genetics
15.
Virus Genes ; 56(6): 734-748, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33009986

ABSTRACT

Fowlpox virus (FPV) is used as a vaccine vector to prevent diseases in poultry and mammals. The insertion site is considered as one of the main factors influencing foreign gene expression. Therefore, the identification of insertion sites that can stably and efficiently express foreign genes is crucial for the construction of recombinant vaccines. In this study, we found that the insertion of foreign genes into ORF054 and the ORF161/ORF162 intergenic region of the FPV genome did not affect replication, and that the foreign genes inserted into the intergenic region were more efficiently expressed than when they were inserted into a gene. Based on these results, the recombinant virus rFPVNX10-NDV F-E was constructed and immune protection against virulent FPV and Newcastle disease virus (NDV) was evaluated. Tests for anti-FPV antibodies in the vaccinated chickens were positive within 14 days post-vaccination. After challenge with FPV102, no clinical signs of FP were observed in vaccinated chickens, as compared to that in the control group (unvaccinated), which showed 100% morbidity. Low levels of NDV-specific neutralizing antibodies were detected in vaccinated chickens before challenge. After challenge with NDV ck/CH/LHLJ/01/06, all control chickens died within 4 days post-challenge, whereas 5/15 vaccinated chickens died between 4 and 12 days post-challenge. Vaccination provided an immune protection rate of 66.7%, whereas the control group showed 100% mortality. These results indicate that the ORF161/ORF162 intergenic region of FPVNX10 can be used as a recombination site for foreign gene expression in vivo and in vitro.


Subject(s)
Fowlpox virus/genetics , Fowlpox/prevention & control , Newcastle Disease/prevention & control , Poultry Diseases/prevention & control , Viral Fusion Proteins/genetics , Viral Vaccines/genetics , Animals , Cell Line , Chick Embryo , Chickens , DNA, Intergenic , Fibroblasts , Vaccination/veterinary , Vaccines, Synthetic/genetics
16.
Methods Mol Biol ; 2203: 147-165, 2020.
Article in English | MEDLINE | ID: mdl-32833211

ABSTRACT

We have developed a reverse genetics system for the avian coronavirus infectious bronchitis virus (IBV) in which a full-length cDNA corresponding to the IBV genome is inserted into the vaccinia virus genome under the control of a T7 promoter sequence. Vaccinia virus as a vector for the full-length IBV cDNA has the advantage that modifications can be introduced into the IBV cDNA using homologous recombination, a method frequently used to insert and delete sequences from the vaccinia virus genome. Here, we describe the use of transient dominant selection as a method for introducing modifications into the IBV cDNA that has been successfully used for the substitution of specific nucleotides, deletion of genomic regions, and the exchange of complete genes. Infectious recombinant IBVs are generated in situ following the transfection of vaccinia virus DNA, containing the modified IBV cDNA, into cells infected with a recombinant fowlpox virus expressing T7 DNA-dependent RNA polymerase.


Subject(s)
Infectious bronchitis virus/genetics , Transfection/methods , Vaccinia virus/genetics , Animals , Bacteriophages/genetics , Chlorocebus aethiops , DNA-Directed DNA Polymerase/metabolism , Fowlpox virus/genetics , Homologous Recombination , Microorganisms, Genetically-Modified , Vaccinia virus/isolation & purification , Vero Cells
17.
J Gen Virol ; 101(7): 772-777, 2020 07.
Article in English | MEDLINE | ID: mdl-32427095

ABSTRACT

In comparison to the extensive characterization of haemagglutinin antibodies of avian influenza virus (AIV), the role of neuraminidase (NA) as an immunogen is less well understood. This study describes the construction and cellular responses of recombinant fowlpox viruses (rFWPV) strain FP9, co-expressing NA N1 gene of AIV A/Chicken/Malaysia/5858/2004, and chicken IL-12 gene. Our data shows that the N1 and IL-12 proteins were successfully expressed from the recombinants with 48 kD and 70 kD molecular weights, respectively. Upon inoculation into specific-pathogen-free (SPF) chickens at 105 p.f.u. ml-1, levels of CD3+/CD4+ and CD3+/CD8+ populations were higher in the wild-type fowlpox virus FP9 strain, compared to those of rFWPV-N1 and rFWPV-N1-IL-12 at weeks 2 and 5 time points. Furthermore, rFWPV-N1-IL-12 showed a suppressive effect on chicken body weight within 4 weeks after inoculation. We suggest that co-expression of N1 with or without IL-12 offers undesirable quality as a potential AIV vaccine candidate.


Subject(s)
Fowlpox virus/genetics , Gene Expression , Genetic Vectors/genetics , Influenza A virus/genetics , Interleukin-12/genetics , Neuraminidase/genetics , Viral Proteins/genetics , Animals , Chickens , Fowlpox virus/immunology , Host-Pathogen Interactions/immunology , Immunity, Cellular , Influenza in Birds/virology , Recombination, Genetic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
18.
Front Immunol ; 11: 613079, 2020.
Article in English | MEDLINE | ID: mdl-33633733

ABSTRACT

The anti-viral immune response is dependent on the ability of infected cells to sense foreign nucleic acids. In multiple species, the pattern recognition receptor (PRR) cyclic GMP-AMP synthase (cGAS) senses viral DNA as an essential component of the innate response. cGAS initiates a range of signaling outputs that are dependent on generation of the second messenger cGAMP that binds to the adaptor protein stimulator of interferon genes (STING). Here we show that in chicken macrophages, the cGAS/STING pathway is essential not only for the production of type-I interferons in response to intracellular DNA stimulation, but also for regulation of macrophage effector functions including the expression of MHC-II and co-stimulatory molecules. In the context of fowlpox, an avian DNA virus infection, the cGAS/STING pathway was found to be responsible for type-I interferon production and MHC-II transcription. The sensing of fowlpox virus DNA is therefore essential for mounting an anti-viral response in chicken cells and for regulation of a specific set of macrophage effector functions.


Subject(s)
Chickens/metabolism , Chickens/virology , Fowlpox/metabolism , Macrophages/metabolism , Nucleotides, Cyclic/metabolism , Animals , Cell Line , DNA Viruses/genetics , DNA, Viral/genetics , Fowlpox virus/genetics , Histocompatibility Antigens Class II/metabolism , Interferon Type I/metabolism , Macrophages/virology , Membrane Proteins/metabolism , Signal Transduction/physiology
19.
Cancer Gene Ther ; 27(6): 438-447, 2020 06.
Article in English | MEDLINE | ID: mdl-31222182

ABSTRACT

Intravesical BCG is a highly effective treatment for high-grade nonmuscle invasive bladder cancer and carcinoma in situ (CIS); however, for patients who are either resistant or become unresponsive to BCG therapy there is a need for alternative treatment approaches. This study examined the safety and feasibility of intravesically administered recombinant fowlpox virus encoding GM-CSF (Arm A) or TRICOM (Arm B); and the local and systemic immunologic responses generated to the vector(s). Twenty bladder cancer patients scheduled for cystectomy as their standard of care received preoperatively four weekly doses of intravesical recombinant fowlpox. Treatment was well tolerated, however, three patients experienced transient elevations of liver transaminases, with one rising to the level of a DLT. Cystectomy derived tumor and normal bladder mucosa demonstrated mRNA for the virally encoded LacZ gene supporting effective infection/transfection. Detected serum antibody to the LacZ encoding ß-galactosidase indicated successful expression of vector-encoding gene products and the ability to immunize via the bladder site. H&E and IHC using a panel of immune cell specific antigens demonstrated immune cell infiltration of the bladder wall. These findings demonstrate good safety profile, successful infection/transfection, ability to generate systemic immune response, and local recruitment of immune cell populations with intravesical administration of fowlpox-based constructs encoding for GM-CSF(rF-GM-CSF) or TRICOM (rF-TRICOM), and support further evaluation of this treatment modality for bladder cancer.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder/physiopathology , Administration, Intravesical , Aged , Animals , Dose-Response Relationship, Drug , Fowlpox virus/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Middle Aged , Neoadjuvant Therapy , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Urinary Bladder Neoplasms/pathology
20.
J Gen Virol ; 100(5): 838-850, 2019 05.
Article in English | MEDLINE | ID: mdl-30907721

ABSTRACT

Avian pox is a highly contagious avian disease, yet relatively little is known about the epidemiology and transmission of Avipoxviruses. Using a molecular approach, we report evidence for a potential link between birds and field-caught mosquitoes in the transmission of Fowlpox virus (FWPV) in Singapore. Comparison of fpv167 (P4b), fpv126 (VLTF-1), fpv175-176 (A11R-A12L) and fpv140 (H3L) gene sequences revealed close relatedness between FWPV strains obtained from cutaneous lesions of a chicken and four pools of Culex pseudovishnui, Culex spp. (vishnui group) and Coquellitidea crassipes caught in the vicinity of the study site. Chicken-derived viruses characterized during two separate infections two years later were also identical to those detected in the first event, suggesting repeated transmission of closely related FWPV strains in the locality. Since the study location is home to resident and migratory birds, we postulated that wild birds could be the source of FWPV and that bird-biting mosquitoes could act as bridging mechanical vectors. Therefore, we determined whether the FWPV-positive mosquito pools (n=4) were positive for avian DNA using a polymerase chain reaction-sequencing assay. Our findings confirmed the presence of avian host DNA in all mosquito pools, suggesting a role for Cx. pseudovishnui, Culex spp. (vishnui group) and Cq. crassipes mosquitoes in FWPV transmission. Our study exemplifies the utilization of molecular tools to understand transmission networks of pathogens affecting avian populations, which has important implications for the design of effective control measures to minimize disease burden and economic loss.


Subject(s)
Bird Diseases/virology , Chickens/virology , Culicidae/virology , Fowlpox virus/genetics , Fowlpox/transmission , Fowlpox/virology , Mosquito Vectors/genetics , Animals , Animals, Wild , Phylogeny , Sequence Analysis, DNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL