Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 190
1.
Poult Sci ; 103(2): 103271, 2024 Feb.
Article En | MEDLINE | ID: mdl-38064882

Multiple outbreaks of avian infectious laryngotracheitis (ILT) in chickens, both domestically and internationally, have been directly correlate to widespread vaccine use in affected countries and regions. Phylogenetic and recombination event analyses have demonstrated that avian infectious laryngotracheitis virus (ILTV) field strains are progressively evolving toward the chicken embryo-origin (CEO) vaccine strain. Even with standardized biosecurity measures and effective prevention and control strategies implemented on large-scale farms, continuous ILT outbreaks result in significant economic losses to the poultry industry worldwide. These outbreaks undoubtedly hinder efforts to control and eradicate ILTV in the future. In this study, an ILTV isolate was successfully obtained by laboratory PCR detection and virus isolation from chickens that exhibited dyspnea and depression on a broiler farm in Hubei Province, China. The isolated strain exhibited robust propagation on chorioallantoic membranes of embryonated eggs, but failed to establish effective infection in chicken hepatocellular carcinoma (LMH) cells. Phylogenetic analysis revealed a unique T441P point mutation in the gJ protein of the isolate. Animal experiments confirmed the virulence of this strain, as it induced mortality in 6-wk-old chickens. This study expands current understanding of the epidemiology, genetic variations, and pathogenicity of ILTV isolates circulating domestically, contributing to the elucidate of ILTV molecular basis of pathogenicity and development of vaccine.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Chick Embryo , Animals , Chickens , Herpesvirus 1, Gallid/genetics , Virulence , Phylogeny , Ovum , Herpesviridae Infections/epidemiology , Herpesviridae Infections/veterinary , Poultry Diseases/prevention & control
2.
J Gen Virol ; 104(4)2023 04.
Article En | MEDLINE | ID: mdl-37010948

Infectious laryngotracheitis virus (ILTV; an alphaherpesvirus) is a respiratory pathogen of chickens and causes significant economic losses in the poultry industry globally, in addition to severe animal health and welfare concerns. To date, studying the role of ILTV genes in viral infection, replication or pathogenesis has largely been limited to genes that can be deleted from the ILTV genome and the resultant deletion mutants characterized in vitro or in vivo. However, this approach is not suitable for the study of essential genes. This study trialled two different codon deoptimization techniques that aimed to separately disrupt and downregulate the expression of two ILTV genes, ICP8 and UL12, which are essential or very important in viral replication. The target genes were partially recoded using codon usage deoptimization (CUD) and codon pair bias deoptimization (CPBD) approaches and characterized in vitro. Viruses deoptimized via CPBD showed decreased protein expression as assessed by Western blotting and/or fluorescence microscopy to measure the intensity of the fluorescent marker fused to the target protein. Viruses deoptimized by CUD showed less consistent results, with some mutants that could not be generated or isolated. The results indicate that CPBD is an attractive and viable tool for the study of essential or critically important genes in ILTV. This is the first study, to our knowledge, that utilizes CPBD and CUD techniques for the study of ILTV genes.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Animals , Chickens , Codon Usage , Genes, Essential , Herpesvirus 1, Gallid/genetics , Codon/genetics
3.
J Virol ; 97(4): e0140622, 2023 04 27.
Article En | MEDLINE | ID: mdl-37022163

The genomes of numerous herpesviruses have been cloned as infectious bacterial artificial chromosomes. However, attempts to clone the complete genome of infectious laryngotracheitis virus (ILTV), formally known as Gallid alphaherpesvirus-1, have been met with limited success. In this study, we report the development of a cosmid/yeast centromeric plasmid (YCp) genetic system to reconstitute ILTV. Overlapping cosmid clones were generated that encompassed 90% of the 151-Kb ILTV genome. Viable virus was produced by cotransfecting leghorn male hepatoma (LMH) cells with these cosmids and a YCp recombinant containing the missing genomic sequences - spanning the TRS/UL junction. An expression cassette for green fluorescent protein (GFP) was inserted within the redundant inverted packaging site (ipac2), and the cosmid/YCp-based system was used to generate recombinant replication-competent ILTV. Viable virus was also reconstituted with a YCp clone containing a BamHI linker within the deleted ipac2 site, further demonstrating the nonessential nature of this site. Recombinants deleted in the ipac2 site formed plaques undistinguished from those viruses containing intact ipac2. The 3 reconstituted viruses replicated in chicken kidney cells with growth kinetics and titers similar to the USDA ILTV reference strain. Specific pathogen-free chickens inoculated with the reconstituted ILTV recombinants succumbed to levels of clinical disease similar to that observed in birds inoculated with wildtype viruses, demonstrating the reconstituted viruses were virulent. IMPORTANCE Infectious laryngotracheitis virus (ILTV) is an important pathogen of chicken with morbidity of 100% and mortality rates as high as 70%. Factoring in decreased production, mortality, vaccination, and medication, a single outbreak can cost producers over a million dollars. Current attenuated and vectored vaccines lack safety and efficacy, leaving a need for better vaccines. In addition, the lack of an infectious clone has also impeded understanding viral gene function. Since infectious bacterial artificial chromosome (BAC) clones of ILTV with intact replication origins are not feasible, we reconstituted ILTV from a collection of yeast centromeric plasmids and bacterial cosmids, and identified a nonessential insertion site within a redundant packaging site. These constructs and the methodology necessary to manipulate them will facilitate the development of improved live virus vaccines by modifying genes encoding virulence factors and establishing ILTV-based viral vectors for expressing immunogens of other avian pathogens.


Cosmids , Herpesvirus 1, Gallid , Mutagenesis , Plasmids , Animals , Male , Chickens , Cosmids/genetics , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/genetics , Herpesvirus 1, Gallid/pathogenicity , Plasmids/genetics , Poultry Diseases/virology , Saccharomyces cerevisiae/genetics , Cell Line , Genome, Viral/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
4.
Viruses ; 15(2)2023 02 11.
Article En | MEDLINE | ID: mdl-36851714

Infectious laryngotracheitis (ILT) and Newcastle disease (ND) are two important avian diseases that have caused huge economic losses to the poultry industry worldwide. Newcastle disease virus (NDV) has been used as a vector in the development of vaccines and gene delivery. In the present study, we generated a thermostable recombinant NDV (rNDV) expressing the glycoprotein gB (gB) of infectious laryngotracheitis virus (ITLV) based on the full-length cDNA clone of the thermostable TS09-C strain. This thermostable rNDV, named rTS-gB, displayed similar thermostability, growth kinetics, and pathogenicity compared with the parental TS09-C virus. The immunization data showed that rTS-gB induced effective ILTV- and NDV-specific antibody responses and conferred immunization protection against ILTV challenge in chickens. The efficacy of rTS-gB in alleviating clinical signs was similar to that of the commercial attenuated ILTV K317 strain. Furthermore, rTS-gB could significantly reduce viral shedding in cloacal and tracheal samples. Our study suggested that the rNDV strain rTS-gB is a thermostable, safe, and highly efficient vaccine candidate against ILT and ND.


Bird Diseases , Herpesvirus 1, Gallid , Newcastle Disease , Animals , Newcastle disease virus/genetics , Chickens , Newcastle Disease/prevention & control , Antibodies, Viral , Herpesvirus 1, Gallid/genetics
5.
Braz J Microbiol ; 53(4): 2223-2232, 2022 Dec.
Article En | MEDLINE | ID: mdl-36269555

Avian infectious laryngotracheitis (ILT) is a respiratory disease that causes severe economic losses in the poultry industry, mainly due to high morbidity and mortality and reduced egg production. Molecular characterization was performed on samples collected from flocks in the Brazilian States of São Paulo, Pernambuco, and Minas Gerais during 2015 and 2016 that presented clinical signs of respiratory disease. End-point PCR was used for viral detection, and DNA sequencing was used for differentiation of vaccine and field strains. Molecular analysis based on the infected cell protein (ICP4) gene separated four of the nine samples together with previous Brazilian isolates (São Paulo and Minas Gerais), one sample was grouped on the same branch as Minas Gerais strains (along with another related sample), one sample was separately branched but still related to the tissue culture origin (TCO) vaccine strain, and two samples were grouped on the same branch as the TCO vaccine strain. Molecular analysis of the thymidine kinase (TK) gene showed the existence of strains of both high and low virulence. The characterization of two fragments of the ICP4 gene and a fragment of the TK gene in this study suggested that the virus circulating in Guatapará, as well as those in Barretos and Itanhandu, that is causing respiratory problems in birds is a highly virulent field strain. The clinical signs point to a TCO vaccine strain that most likely underwent some reversal event and is a latent reactivated infection.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Animals , Chickens , Brazil/epidemiology , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics
6.
Virus Genes ; 58(6): 540-549, 2022 Dec.
Article En | MEDLINE | ID: mdl-36127475

In alphaherpesviruses, glycoproteins E and I (gE and gI, respectively) form a heterodimer that facilitates cell-to-cell spread of virus. Using traditional homologous recombination techniques, as well as CRISPR/Cas9-assisted homologous recombination, we separately deleted gE and gI coding sequences from an Australian field strain (CSW-1) and a vaccine strain (A20) of infectious laryngotracheitis virus (ILTV) and replaced each coding sequence with sequence encoding green fluorescent protein (GFP). Virus mutants in which gE and gI gene sequences had been replaced with GFP were identified by fluorescence microscopy but were unable to be propagated separately from the wildtype virus in either primary chicken cells or the LMH continuous chicken cell line. These findings build on findings from a previous study of CSW-1 ILTV in which a double deletion mutant of gE and gI could not be propagated separately from wildtype virus and produced an in vivo phenotype of single-infected cells with no cell-to-cell spread observed. Taken together these studies suggest that both the gE and gI genes have a significant role in cell-to-cell spread in both CSW-1 and A20 strains of ILTV. The CRISPR/Cas9-assisted deletion of genes from the ILTV genome described in this study adds this virus to a growing list of viruses to which this approach has been used to study viral gene function.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Animals , CRISPR-Cas Systems , Australia , Herpesvirus 1, Gallid/genetics , Chickens , Glycoproteins/genetics , Green Fluorescent Proteins/genetics , Homologous Recombination
7.
Avian Dis ; 66(3): 1-9, 2022 10.
Article En | MEDLINE | ID: mdl-36106910

Population-level sampling based on qPCR detection of infectious laryngotracheitis virus (ILTV) in poultry dust can be used to assess ILT vaccination outcomes following mass administration in drinking water. We report on the field application of this approach to assess the success of vaccine administration and its use in ILT outbreak control in meat chickens. In Study 1, dust samples were collected from 26 meat chicken flocks at 0, 4, 7, 14, and 21 days post drinking water vaccination (DPV) given between 7 to 13 days of age with the Serva or A20 live attenuated ILT vaccines. Unexpectedly, ILTV DNA was detected in dust samples collected prior to vaccination in 22/26 flocks. Typing revealed that the detected ILTV was different from the vaccine virus. To determine whether the detected ILTV DNA was from active infection or carryover of a noninfectious virus, Study 2 was implemented in 14 additional flocks with dust samples collected at 0, 7, 14, and 21 DPV and tracheal swabs collected from 15 birds/flock at 0 and 21 DPV. The results indicated that there was active infection with ILTV in those flocks before vaccination. This approach contributed to a statewide control program resulting in the eradication of ILT from South Australia as confirmed by negative ILTV test results for dust samples from 50 flocks and the absence of clinical ILT. These findings show that ILTV infection prior to vaccination is common in outbreak situations and that dust samples must be collected at 0 and 7 DPV for meaningful interpretation of vaccination outcomes and ILTV status. Comparatively low-cost dust testing during an outbreak, coupled with typing information, greatly assisted with decision making and control strategies during a major outbreak, including confirmation of the absence of infection in the final stages.


Aplicación de campo del monitoreo por qPCR del virus de la laringotraqueítis infecciosa en el polvo de casetas avícolas y su función en el control de un brote importante El muestreo a nivel de población basado en la detección por qPCR del virus de la laringotraqueítis infecciosa (ILTV) en el polvo de instalaciones avícolas se puede utilizar para evaluar los resultados de la vacunación contra esta enfermedad después de la administración masiva en el agua de bebida. Se reporta la aplicación de campo de este enfoque para evaluar el éxito de la administración de vacunas y su uso en el control de brotes por laringotraqueítis infecciosa en pollos de engorde. En el Estudio 1, se recolectaron muestras de polvo de 26 parvadas de pollos de engorda a los 0, 4, 7, 14 y 21 días después de la vacunación en el agua de bebida (DPV) a los 7 a 13 días de edad con las vacunas de laringotraqueítis vivas atenuadas Serva o A20. Inesperadamente, se detectó ADN del virus de laringotraqueítis en muestras de polvo recolectadas antes de la vacunación en 22/26 parvadas. La tipificación reveló que el virus detectado era diferente del virus de la vacuna. Para determinar si el ADN del virus de laringotraqueítis detectado procedía de una infección activa o del remanente de un virus no infeccioso, se implementó el Estudio 2 en 14 parvadas adicionales con muestras de polvo recolectadas a los 0, 7, 14 y 21 días después de la vacunación y de hisopos traqueales recolectados de 15 aves/parvada a los cero y 21 días después de la vacunación. Los resultados indicaron que había infección activa con el virus de laringotraqueítis en esas parvadas antes de la vacunación. Este enfoque contribuyó a un programa de control estatal que resultó en la erradicación de laringotraqueítis del sur de Australia, como lo confirmaron los resultados negativos de las pruebas del mismo virus para muestras de polvo de 50 parvadas y la ausencia de laringotraqueítis infecciosa clínica. Estos hallazgos muestran que la infección por el virus de la laringotraqueítis antes de la vacunación es común en situaciones de brotes y que las muestras de polvo deben recolectarse a los cero y 7 días después de la vacunación para una interpretación significativa de los resultados de la vacunación y el estado de esta enfermedad. Las pruebas de polvo comparativamente de bajo costo durante un brote, junto con la información de tipificación, ayudaron mucho con la toma de decisiones y con las estrategias de control durante un brote importante, incluida la confirmación de la ausencia de infección en las etapas finales.


Drinking Water , Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Animals , Chickens , Disease Outbreaks/prevention & control , Disease Outbreaks/veterinary , Dust , Herpesviridae Infections/diagnosis , Herpesviridae Infections/epidemiology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Poultry Diseases/diagnosis , Poultry Diseases/epidemiology , Poultry Diseases/prevention & control , Vaccines, Attenuated
8.
BMC Vet Res ; 18(1): 358, 2022 Sep 26.
Article En | MEDLINE | ID: mdl-36163027

Infectious laryngotracheitis (ILT) is an economically crucial respiratory disease of poultry that affects the industry worldwide. Vaccination is the principal tool in the control of the disease outbreak. In an earlier study, we comprehensively characterized the circulating strains in Egypt and identified both CEO-like and recombinant strains are dominant. Herein, we investigated the pathogenicity of two virulent strains representing the CEO-like (Sharkia_2018) and recombinant strain (Qalubia_2018). Additionally, we evaluated the efficacy of different commercial vaccines (HVT-LT, CEO, and TCO) against the two isolates in terms of the histopathological lesion scores and the viral (gC) gene load. A total of 270 White Leghorn-specific pathogen-free male chicks were divided into nine groups of 30 birds, each housed in separate isolators. Birds were distributed as follows; one group was non-vaccinated, non-challenged, and served as a negative control. Two groups were non-vaccinated and infected with the two isolates of interest and served as a positive control to test the pathogenicity. Six groups were vaccinated and challenged; two groups were vaccinated with vector vaccine at one day old. The other four groups were vaccinated with either the CEO- or TCO- vaccine (two groups each) at four weeks of age. Three weeks after vaccination, birds were infected with the virulent ILTV isolates. The larynx, trachea, and harderian gland samples were taken at 1, 3, and 7 days post-infection for histopathological lesion score and molecular detection. Notably, The recombinant strain was more virulent and pathogenic than CEO-like ILTV strains. Moreover, the TCO vaccine was less immunogenic than the vector and CEO vaccines.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Animals , Chickens , Egypt/epidemiology , Herpesviridae Infections/prevention & control , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Male , Vaccine Efficacy , Vaccines, Attenuated , Virulence
9.
Infect Genet Evol ; 104: 105350, 2022 10.
Article En | MEDLINE | ID: mdl-35977653

Infectious laryngotracheitis (ILT), caused by infectious laryngotracheitis virus (ILTV), occurs sporadically in poultry flocks in Canada. Live attenuated chicken embryo origin (CEO) vaccines are being used routinely to prevent and control ILTV infections. However, ILT outbreaks still occur since vaccine strains could revert to virulence in the field. In this study, 7 Canadian ILTV isolates linked to ILT outbreaks across different time in Eastern Canada (Ontario; ON and Quebec; QC) were whole genome sequenced. Phylogenetic analysis confirmed the close relationship between the ON isolates and the CEO vaccines, whereas the QC isolates clustered with strains previously known as CEO revertant and wild-type ILTVs. Recombination network analysis of ILTV sequences revealed clear evidence of historical recombination between ILTV strains circulating in Canada and other geographical regions. The comparison of ON CEO clustered and QC CEO revertant clustered isolates with the LT Blen® CEO vaccine reference sequence showed amino acid differences in 5 and 12 open reading frames (ORFs), respectively. Similar analysis revealed amino acid differences in 32 ORFs in QC wild-type isolates. Compared to all CEO vaccine strains in the public domain, the QC wild-type isolates showed 15 unique mutational sites leading to amino acid changes in 13 ORFs. Our outcomes add to the knowledge of the molecular mechanisms behind ILTV genetic variance and provide genetic markers between wild-type and vaccine strains.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Amino Acids/genetics , Animals , Chick Embryo , Chickens , Genetic Markers , Herpesviridae Infections/epidemiology , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Ontario , Phylogeny , Sequence Analysis, DNA , Vaccines, Attenuated/genetics , Viral Vaccines/genetics
10.
Arch Virol ; 167(9): 1819-1829, 2022 Sep.
Article En | MEDLINE | ID: mdl-35716265

Infectious laryngotracheitis (ILT) is an acute respiratory disease in chickens that is a serious threat to poultry-producing countries worldwide. In the present study, we isolated and characterized infectious laryngotracheitis (ILTV) virus isolates by sequencing and restriction fragment length polymorphism analysis of PCR-amplified products (PCR-RFLP). A total of 26 ILTV outbreaks were investigated that occurred between 2019 and 2020 in flocks that had not been vaccinated against ILTV. ILTV was isolated by cultivating tracheal samples in embryonated chicken eggs, which showed multiple opaque pock lesions and thickening of the chorioallantoic membrane after 120 hours of infection. The ILTV isolates were identified and characterized by PCR and sequencing a portion of the ICP4 and TK genes. Phylogenetic analysis based on the ICP4 region showed that the sequences clustered with chicken-embryo-origin vaccine-like strains. Sequence analysis of the ICP4 region differentiated chicken-embryo-origin (CEO), tissue-culture-origin (TCO), and field ILTV strains, with significant differences in nucleotide and amino acid sequences. Furthermore, PCR-RFLP analysis of the TK gene showed that the patterns were identical to those obtained with low-virulence and vaccine strains. In conclusion, sequencing of a portion of the ICP4 region of ILTV allowed differentiation of ILTV field, CEO, and TCO vaccine strains. In this study, CEO-vaccine-like strains were found to be the cause of ILTV outbreaks between 2019 and 2020 in Tamil Nadu in southern India.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Animals , Chickens , Disease Outbreaks/veterinary , Female , Herpesviridae Infections/epidemiology , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , India/epidemiology , Phylogeny , Poultry Diseases/epidemiology , Viral Vaccines/genetics
11.
Vet Microbiol ; 269: 109435, 2022 Jun.
Article En | MEDLINE | ID: mdl-35462119

Therapeutics targeting virus-host interactions have been considered promising strategies for treating herpesvirus infection. Our previous study on avian infectious laryngotracheitis virus (ILTV), an avian herpesvirus economically important to the poultry industry worldwide, identified the small molecule Pifithrin-α (PFT-α) as a potential therapeutic agent. However, the underlying mechanisms of its antiviral function remain largely unknown. Using the ILTV-permissive chicken cell line LMH as the model, we found that PFT-α effectively suppressed the transcription and genome replication of ILTV and greatly reduced the level of infectious virions. Genome-wide transcriptome analysis revealed extensive repression of the metabolic processes of infected cells by PFT-α administration. Further metabolome assays of ILTV-infected cells using liquid chromatography coupled with mass spectrometry suggest host nucleotide metabolism and ATP synthesis as the key targets of PFT-α treatment during its repression of ILTV replication, which was experimentally supported by the reduced transcription of many key enzymes essential to nucleotide metabolism and ATP synthesis. The present study provides insights into the mechanisms by which PFT-α inhibits ILTV infection, which may increase the probability of successful clinical application of this molecule.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Adenosine Triphosphate , Animals , Benzothiazoles , Chickens , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Nucleotides , Toluene/analogs & derivatives
12.
Arch Virol ; 167(4): 1151-1155, 2022 Apr.
Article En | MEDLINE | ID: mdl-35244762

Infectious laryngotracheitis virus (ILTV) is the causative agent of an economically important disease of chickens causing upper respiratory tract infection. Strains of ILTV are commonly identified by polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) and/or PCR high resolution melt (PCR-HRM) curve analysis targeting several genes. However, these techniques examine only a limited number of mutations present inside the target regions and may generate unreliable results when the sample contains more than one strain. Here, we attempted to sequence the whole genome of ILTV with known identity (class 9) directly from tracheal scrapings to circumvent in vitro culturing, which can potentially introduce variations into the genome. Despite the large number of quality reads, mapping was compromised by poor overlapping and gaps, and assembly of the complete genome sequence was not possible. In a map-to-reference alignment, the regions with low coverage were deleted, those with high coverage were concatenated and a genome sequence of 139,465 bp was obtained, which covered 91% of the ILTV genome. Sixteen single-nucleotide polymorphisms (SNPs) were found between the ILTV isolate examined and ILTV class 9 (JN804827). Despite only 91% genome coverage, using sequence analysis and comparison with previously sequenced ILTVs, we were able to classify the isolate as class 9. Therefore, this technique has the potential to replace the current PCR-HRM technique, as it provides detailed information about the ILTV isolates.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Animals , Chickens , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA
13.
Avian Dis ; 65(3): 500-507, 2021 09.
Article En | MEDLINE | ID: mdl-34699149

To produce more-stable, live attenuated vaccines for infectious laryngotracheitis virus (ILTV), deletion of genes related to virulence has been extensively pursued. Although its function remains unknown, the open reading frame C (ORF C) is among the genes potentially associated with viral virulence that is nonessential for replication in vitro. Earlier results indicated that the ILT virus with deletion of the ORF C gene (BΔORFC) was suitable and safe for eye drop administration but was not sufficiently attenuated for in ovo administration. The objective of this study was to evaluate the safety and protection efficacy of a cell line-adapted, gene-deleted strain (BΔORFC) of ILTV when administered in ovo and/or spray (SP) by itself, or in combination with the recombinant HVT-LT (rHVT-LT) vaccine. Results indicated that vaccination with the BΔORFC strain, either by itself or in combination with an rHVT-LT vaccine, did not affect hatchability, and only marginal signs of respiratory distress were recorded for groups of chickens that received the BΔORFC strain via SP. The replication and seroconversion induced by the BΔORFC strain after in ovo and SP administration was very limited, whereas the replication of the rHVT-LT vaccine was delayed when combined with the BΔORFC strain in ovo. Compared to rHVT-LT or BΔORFC when administered alone, dual vaccination with rHVT-LT + BΔORFC was more effective in mitigating clinical signs of the disease and reducing challenge virus load in the trachea. To our knowledge, this study provides the first proof of concept that ILTV strains can be sufficiently attenuated for early vaccination in ovo or at hatch; also, this study documented the benefits of using a dual (recombinant and live attenuated) hatchery vaccination strategy for ILTV.


Una cepa del virus de la laringotraqueítis infecciosa adaptada a una línea celular (BΔORFC) para vacunación in ovo y en aerosol en incubadora aplicada por sí sola o en combinación con una vacuna recombinante para laringotraqueítis con el vector HVT. Para producir vacunas vivas atenuadas más estables contra el virus de la laringotraqueítis infecciosa (ILTV), se ha buscado ampliamente la eliminación de genes relacionados con la virulencia. Aunque su función sigue siendo desconocida, el marco de lectura continuo C (ORF C) se encuentra entre los genes potencialmente asociados con la virulencia viral que no es esencial para la replicación in vitro. Resultados anteriores indican que el virus de la laringotraqueítis infecciosa con deleción del gene ORF C (BΔORFC) era adecuado y seguro para la administración ocular, pero no estaba lo suficientemente atenuado para su administración in ovo. El objetivo de este estudio fue evaluar la seguridad y la eficacia de la protección de una cepa del virus de la laringotraqueítis infecciosa con deleción genética y adaptada a una línea celular (BΔORFC) cuando se administra in ovo y/o en aerosol por sí sola, o en combinación con una vacuna recombinante con el vector HVT (vacuna rHVT-LT). Los resultados indicaron que la vacunación con la cepa BΔORFC, ya sea sola o en combinación con la vacuna rHVT-LT, no afectó la incubabilidad, y solo se registraron signos marginales de dificultad respiratoria para los grupos de pollos que recibieron la cepa BΔORFC por aspersión. La replicación y seroconversión inducida por la cepa BΔORFC después de la administración in ovo y por aspersión fue muy limitada, mientras que la replicación de la vacuna rHVT-LT se retrasó cuando se combinó con la cepa BΔORFC in ovo. En comparación con las vacunas rHVT-LT o BΔORFC administradas por sí solas, la vacunación dual con rHVT-LT + BΔORFC fue más eficaz para mitigar los signos clínicos de la enfermedad y reducir la carga del virus de desafío en la tráquea. Hasta donde se conoce, este estudio proporciona la primera prueba del concepto de que las cepas del virus de la laringotraqueítis infecciosa pueden atenuarse lo suficiente para la vacunación temprana in ovo o en la incubadora. Además, este estudio documentó los beneficios de utilizar una estrategia de vacunación de incubadora dual (vacuna recombinante y viva atenuada) para la laringotraqueítis infecciosa.


Herpesviridae Infections , Herpesvirus 1, Gallid , Poultry Diseases , Viral Vaccines , Animals , Cell Line , Chickens , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Poultry Diseases/prevention & control , Vaccination/veterinary , Vaccines, Attenuated
14.
Viruses ; 13(8)2021 07 21.
Article En | MEDLINE | ID: mdl-34452285

Marek's disease (MD) in chickens is caused by Gallid alphaherpesvirus 2, better known as MD herpesvirus (MDV). Current vaccines do not block interindividual spread from chicken-to-chicken, therefore, understanding MDV interindividual spread provides important information for the development of potential therapies to protect against MD, while also providing a natural host to study herpesvirus dissemination. It has long been thought that glycoprotein C (gC) of alphaherpesviruses evolved with their host based on their ability to bind and inhibit complement in a species-selective manner. Here, we tested the functional importance of gC during interindividual spread and host specificity using the natural model system of MDV in chickens through classical compensation experiments. By exchanging MDV gC with another chicken alphaherpesvirus (Gallid alphaherpesvirus 1 or infectious laryngotracheitis virus; ILTV) gC, we determined that ILTV gC could not compensate for MDV gC during interindividual spread. In contrast, exchanging turkey herpesvirus (Meleagrid alphaherpesvirus 1 or HVT) gC could compensate for chicken MDV gC. Both ILTV and MDV are Gallid alphaherpesviruses; however, ILTV is a member of the Iltovirus genus, while MDV is classified as a Mardivirus along with HVT. These results suggest that gC is functionally conserved based on the virus genera (Mardivirus vs. Iltovirus) and not the host (Gallid vs. Meleagrid).


Antigens, Viral/metabolism , Chickens/virology , Herpesvirus 2, Gallid/physiology , Marek Disease/transmission , Marek Disease/virology , Viral Envelope Proteins/metabolism , Animals , Antigens, Viral/genetics , Cells, Cultured , Herpesvirus 1, Gallid/classification , Herpesvirus 1, Gallid/genetics , Herpesvirus 1, Meleagrid/classification , Herpesvirus 1, Meleagrid/genetics , Herpesvirus 2, Gallid/classification , Herpesvirus 2, Gallid/genetics , Recombinant Proteins/metabolism , Turkeys/virology , Viral Envelope Proteins/genetics , Virus Replication
15.
Rev. argent. microbiol ; 53(2): 41-50, June 2021. graf
Article En | LILACS | ID: biblio-1376406

Resumen En un estudio epidemiológico realizado previamente en Argentina, se analizó la secuencia de un fragmento del gen US5 del virus de la laringotraqueítis infecciosa (ILTV), lo que permitió diferenciar las cepas de campo de las vacunales. También esto permitió definir cinco haplotipos del ILTV, con variaciones específicas en las posiciones 461, 484, 832, 878 y 894 del gen US5. La caracterización de las cepas virales también puede lograrse mediante el análisis de la disociación de alta resolución o high-resolution melting analysis (HRMA), descripto como un método efectivo, rápido y sensible para detectar mutaciones en productos de PCR. En el presente estudio se desarrolló un protocolo de disociación de alta resolución con el objetivo de caracterizar cepas del ILTV circulantes en Argentina. Para ello,se confirmó la especificidad de esta herramienta en diferentes diluyentes del ADN de las muestras, sin observarse interferencias en presencia de ADN heterólogo u otros metabolitos celulares. Asimismo, la concentración de sales en el buffer de elución utilizado durante la extracción de ADN no alteró los perfiles de las curvas. Se obtuvieron perfiles bien definidos con concentraciones de ADN más elevadas (Ct = 26.0), mientras que concentraciones más bajas presentaron curvas heterogéneas (Ct = 32.5). El HRMA mostró una concordancia del 97.49% con la técnica de referencia, la secuenciación. El protocolo de disociación de alta resolución amplifica el ADN antes de su caracterización, por lo que esta técnica podría ser eventualmente utilizada para confirmar la presencia del ILTV y, al mismo tiempo, distinguir haplotipos, optimizando su valor como herramienta de diagnóstico. Esta característica implica una reducción significativa en el tiempo dedicado al procesamiento de muestras.


Polymerase Chain Reaction , Herpesvirus 1, Gallid , DNA, Viral/genetics , Herpesvirus 1, Gallid/genetics
16.
Viruses ; 13(4)2021 04 19.
Article En | MEDLINE | ID: mdl-33921858

Infectious laryngotracheitis (ILT) is an acute, highly contagious infectious disease of the upper respiratory tract in chickens and other poultry species that causes significant economic losses in countries worldwide. Between 2017 and 2019, seven outbreaks of mild to severe respiratory disorders with high suspicion of ILT occurred in commercial and backyard poultry flocks in Slovenia. In all submissions, infection with ILT virus (ILTV) was confirmed by PCR, which is the first report of ILT in Slovenia. Circulating ILT strains were characterized by the sequence and phylogenetic analysis of two fragments of the ICP4 gene. Four strains-three detected in non-vaccinated flocks and one in a flock vaccinated against ILT-were identical or very similar to the chicken embryo-origin live virus vaccines, and the other three were closely related to Russian, Chinese, Australian, and American field strains and to tissue culture origin vaccine strains. As in other diseases, coinfections with other respiratory pathogens in confirmed ILT cases may cause a more severe condition and prolong the course of the disease. In our study, coinfections with Mycoplasma synoviae (7/7 tested flocks), infectious bronchitis virus (5/5 tested flocks), Mycoplasma gallisepticum (4/7 tested flocks), Ornithobacterium rhinotracheale (3/4 tested flocks), and avian pox virus (1/2 tested flocks) were confirmed, indicating the importance of these pathogens in the occurrence of ILT infections.


Coinfection/veterinary , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Herpesvirus 1, Gallid/pathogenicity , Poultry Diseases/virology , Poultry/virology , Respiratory Tract Diseases/veterinary , Animals , Chickens/virology , Coinfection/microbiology , Coinfection/virology , Herpesviridae Infections/diagnosis , Herpesviridae Infections/epidemiology , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/classification , Herpesvirus 1, Gallid/isolation & purification , Phylogeny , Poultry Diseases/diagnosis , Poultry Diseases/epidemiology , Respiratory Tract Diseases/diagnosis , Respiratory Tract Diseases/epidemiology , Respiratory Tract Diseases/virology , Sequence Analysis, DNA , Slovenia/epidemiology
17.
Viruses ; 13(4)2021 03 24.
Article En | MEDLINE | ID: mdl-33805117

Infectious laryngotracheitis (ILT) is an infectious upper respiratory tract disease that impacts the poultry industry worldwide. ILT is caused by an alphaherpesvirus commonly referred to as infectious laryngotracheitis virus (ILTV). Vaccination with live attenuated vaccines is practiced regularly for the control of ILT. However, extensive and improper use of live attenuated vaccines is related to vaccine viruses reverting to virulence. An increase in mortality and pathogenicity has been attributed to these vaccine revertant viruses. Recent studies characterized Canadian ILTV strains originating from ILT outbreaks as related to live attenuated vaccine virus revertants. However, information is scarce on the pathogenicity and transmission potential of these Canadian isolates. Hence, in this study, the pathogenicity and transmission potential of two wildtype ILTVs and a chicken embryo origin (CEO) vaccine revertant ILTV of Canadian origin were evaluated. To this end, 3-week-old specific pathogen-free chickens were experimentally infected with each of the ILTV isolates and compared to uninfected controls. Additionally, naïve chickens were exposed to the experimentally infected chickens to mimic naturally occurring infection. Pathogenicity of each of these ILTV isolates was evaluated by the severity of clinical signs, weight loss, mortality, and lesions observed at the necropsy. The transmission potential was evaluated by quantification of ILTV genome loads in oropharyngeal and cloacal swabs and tissue samples of the experimentally infected and contact-exposed chickens, as well as in the capacity to produce ILT in contact-exposed chickens. We observed that the CEO vaccine revertant ILTV isolate induced severe disease in comparison to the two wildtype ILTV isolates used in this study. According to ILTV genome load data, CEO vaccine revertant ILTV isolate was successfully transmitted to naïve contact-exposed chickens in comparison to the tested wildtype ILTV isolates. Overall, the Canadian origin CEO vaccine revertant ILTV isolate possesses higher virulence, and dissemination potential, when compared to the wildtype ILTV isolates used in this study. These findings have serious implications in ILT control in chickens.


Herpesviridae Infections/transmission , Herpesviridae Infections/veterinary , Herpesvirus 1, Gallid/genetics , Herpesvirus 1, Gallid/pathogenicity , Poultry Diseases/transmission , Viral Vaccines/analysis , Animals , Canada , Cells, Cultured , Chick Embryo , Chickens/virology , Disease Outbreaks , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/isolation & purification , Liver/cytology , Poultry Diseases/virology , Specific Pathogen-Free Organisms , Vaccines, Attenuated/analysis , Virulence
18.
Poult Sci ; 100(4): 100986, 2021 Apr.
Article En | MEDLINE | ID: mdl-33647723

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.


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
19.
Poult Sci ; 100(3): 100895, 2021 Mar.
Article En | MEDLINE | ID: mdl-33518305

The purpose of this study was to explore a specific, simple, and sensitive method for diagnosis of avian infectious laryngotracheitis virus. Recombinase-aided amplification (RAA) and lateral flow dipstick (LFD) were combined for labeling the optimized RAA probe with 6-carboxyfluorescein (FAM) and the 5'-end of the downstream primer with biotin, respectively. By optimizing the reaction time, temperature, and primer concentration of RAA, a RAA-LFD assay, which could be used for detection of infectious laryngotracheitis, was established. After the specificity and sensitivity test, the target gene fragments could be amplified by RAA-LFD assay in 20 min under isothermal conditions (37°C), and the amplification products could be visually observed and determined by LFD within 3 min. There was no cross-reaction with nucleic acids of other avian pathogens, the lowest detectable limit of RAA-LFD was 102 copies/µL, and the sensitivity of this method was 100 times higher than that of conventional PCR with the lowest detectable limit of 104 copies/µL. The results showed that RAA-LFD assay was highly sensitive, easy to use, and more suitable for clinical detection.


Bird Diseases , Diagnostic Techniques and Procedures , Herpesviridae Infections , Herpesvirus 1, Gallid , Nucleic Acid Amplification Techniques , Animals , Bird Diseases/diagnosis , Bird Diseases/virology , Diagnostic Techniques and Procedures/standards , Diagnostic Techniques and Procedures/veterinary , Herpesviridae Infections/diagnosis , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Herpesvirus 1, Gallid/genetics , Nucleic Acid Amplification Techniques/standards , Nucleic Acid Amplification Techniques/veterinary , Recombinases/metabolism , Sensitivity and Specificity
20.
Rev Argent Microbiol ; 53(2): 89-97, 2021.
Article En | MEDLINE | ID: mdl-32921516

A previous sequence analysis of a US5 gene fragment of infectious laryngotracheitis virus (ILTV) performed in an Argentinian epidemiological study allowed to differentiate between wild and vaccine strains. This analysis also defined five ILTV haplotypes with specific variations at positions 461, 484, 832, 878 and 894 of the US5 gene. This characterization of viral strains may also be accomplished using the High-Resolution Melting Analysis (HRMA), which has been described as an effective, fast and sensitive method to detect mutations in PCR products. In the present study, an HRM protocol was developed with the aim of characterizing the circulating ILTV strains in Argentina. The specificity of this tool was confirmed in different DNA diluents, without interference from heterologous DNA or other cellular metabolites. Additionally, the salt concentration in the elution buffer used for DNA extraction did not alter the curve profiles. Higher concentrations of DNA (Ct≅26.0) displayed well-defined curve profiles, whereas lower concentrations (Ct≅32.5) exhibited more heterogeneous curves. The HRMA showed 97.49% concordance with the reference technique, i.e., sequencing. The HRM protocol has the capability to perform DNA amplification prior to its characterization. Thus, eventually this technique may be used simultaneously as a diagnostic tool. This advantage implies a significant reduction in the time and effort involved in sample processing.


Herpesvirus 1, Gallid , Polymerase Chain Reaction , DNA, Viral/genetics , Herpesvirus 1, Gallid/genetics
...