Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Virology ; 595: 110066, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38574415

ABSTRACT

Avian influenza virus (AIV) is a constant threat to animal health with recent global outbreaks resulting in the death of hundreds of millions of birds with spillover into mammals. Myxovirus-resistance (Mx) proteins are key mediators of the antiviral response that block virus replication. Mouse (Mu) Mx (Mx1) is a strong antiviral protein that interacts with the viral nucleoprotein to inhibit polymerase function. The ability of avian Mx1 to inhibit AIV is unclear. In these studies, Mu Mx1 was stably introduced into chicken DF1 cells to enhance the immune response against AIV. Following infection, titers of AIV were significantly decreased in cells expressing Mu Mx1. In addition, considerably less cytopathic effect (CPE) and matrix protein staining was observed in gene-edited cells expressing Mu Mx1, suggesting Mu Mx1 is broadly effective against multiple AIV subtypes. This work provides foundational studies for use of gene-editing to enhance innate disease resistance against AIV.


Subject(s)
Chickens , Immunity, Innate , Influenza in Birds , Myxovirus Resistance Proteins , Virus Replication , Animals , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Cell Line , Influenza in Birds/virology , Influenza in Birds/immunology , Influenza in Birds/genetics , Mice , Mutagenesis, Insertional , Influenza A virus/immunology , Influenza A virus/genetics
2.
NPJ Vaccines ; 9(1): 45, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38409236

ABSTRACT

Influenza B virus (FLUBV) poses a significant infectious threat, with frequent vaccine mismatch limiting its effectiveness. Our previous work investigated the safety and efficacy of modified live attenuated FLUBV vaccines with rearranged genomes (FluB-RAM and FluB-RANS) or a temperature-sensitive PB1 segment with a C-terminal HA tag (FluB-att). In this study, we compared the immune responses of female and male DBA/2J mice vaccinated with these vaccines, including versions containing a chimeric HA segment with an N-terminal IgA-inducing peptide (IGIP). Importantly, both recombinant viruses with and without IGIP remained genetically stable during egg passage. We found that introducing IGIP strengthened vaccine attenuation, particularly for FluB-RAM/IGIP. Prime-boost vaccination completely protected mice against lethal challenge with a homologous FLUBV strain. Notably, recombinant viruses induced robust neutralizing antibody responses (hemagglutination inhibition titers ≥40) alongside antibodies against NA and NP. Interestingly, female mice displayed a consistent trend of enhanced humoral and cross-reactive IgG and IgA responses against HA, NA, and NP compared to male counterparts, regardless of the vaccine used. However, the presence of IGIP generally led to lower anti-HA responses but higher anti-NA and anti-NP responses, particularly of the IgA isotype. These trends were further reflected in mucosal and serological responses two weeks after challenge, with clear distinctions based on sex, vaccine backbone, and IGIP inclusion. These findings hold significant promise for advancing the development of universal influenza vaccines.

3.
J Virol ; 96(22): e0148022, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36317880

ABSTRACT

Influenza A viruses (FLUAV) cause respiratory diseases in many host species, including humans and pigs. The spillover of FLUAV between swine and humans has been a concern for both public health and the swine industry. With the emergence of the triple reassortant internal gene (TRIG) constellation, establishment of human-origin FLUAVs in pigs has become more common, leading to increased viral diversity. However, little is known about the adaptation processes that are needed for a human-origin FLUAV to transmit and become established in pigs. We generated a reassortant FLUAV (VIC11pTRIG) containing surface gene segments from a human FLUAV strain and internal gene segments from the 2009 pandemic and TRIG FLUAV lineages and demonstrated that it can replicate and transmit in pigs. Sequencing and variant analysis identified three mutants that emerged during replication in pigs, which were mapped near the receptor binding site of the hemagglutinin (HA). The variants replicated more efficiently in differentiated swine tracheal cells compared to the virus containing the wildtype human-origin HA, and one of them was present in all contact pigs. These results show that variants are selected quickly after replication of human-origin HA in pigs, leading to improved fitness in the swine host, likely contributing to transmission. IMPORTANCE Influenza A viruses cause respiratory disease in several species, including humans and pigs. The bidirectional transmission of FLUAV between humans and pigs plays a significant role in the generation of novel viral strains, greatly impacting viral epidemiology. However, little is known about the evolutionary processes that allow human FLUAV to become established in pigs. In this study, we generated reassortant viruses containing human seasonal HA and neuraminidase (NA) on different constellations of internal genes and tested their ability to replicate and transmit in pigs. We demonstrated that a virus containing a common internal gene constellation currently found in U.S. swine was able to transmit efficiently via the respiratory route. We identified a specific amino acid substitution that was fixed in the respiratory contact pigs that was associated with improved replication in primary swine tracheal epithelial cells, suggesting it was crucial for the transmissibility of the human virus in pigs.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus , Influenza A virus , Influenza, Human , Orthomyxoviridae Infections , Swine Diseases , Animals , Humans , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A virus/genetics , Influenza, Human/transmission , Mutation , Orthomyxoviridae Infections/transmission , Reassortant Viruses/genetics , Swine , Swine Diseases/virology
4.
Vaccines (Basel) ; 9(8)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34452022

ABSTRACT

Influenza B virus (IBV) is considered a major respiratory pathogen responsible for seasonal respiratory disease in humans, particularly severe in children and the elderly. Seasonal influenza vaccination is considered the most efficient strategy to prevent and control IBV infections. Live attenuated influenza virus vaccines (LAIVs) are thought to induce both humoral and cellular immune responses by mimicking a natural infection, but their effectiveness has recently come into question. Thus, the opportunity exists to find alternative approaches to improve overall influenza vaccine effectiveness. Two alternative IBV backbones were developed with rearranged genomes, rearranged M (FluB-RAM) and a rearranged NS (FluB-RANS). Both rearranged viruses showed temperature sensitivity in vitro compared with the WT type B/Bris strain, were genetically stable over multiple passages in embryonated chicken eggs and were attenuated in vivo in mice. In a prime-boost regime in naïve mice, both rearranged viruses induced antibodies against HA with hemagglutination inhibition titers considered of protective value. In addition, antibodies against NA and NP were readily detected with potential protective value. Upon lethal IBV challenge, mice previously vaccinated with either FluB-RAM or FluB-RANS were completely protected against clinical disease and mortality. In conclusion, genome re-arrangement renders efficacious LAIV candidates to protect mice against IBV.

5.
Vaccines (Basel) ; 9(7)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34358217

ABSTRACT

Influenza B virus (IBV) is a major respiratory pathogen of humans, particularly in the elderly and children, and vaccines are the most effective way to control it. In previous work, incorporation of two mutations (E580G, S660A) along with the addition of an HA epitope tag in the PB1 segment of B/Brisbane/60/2008 (B/Bris) resulted in an attenuated strain that was safe and effective as a live attenuated vaccine. A third attempted mutation (K391E) in PB1 was not always stable. Interestingly, viruses that maintained the K391E mutation were associated with the mutation E48K. To explore the contribution of the E48K mutation to stability of the K391E mutation, a vaccine candidate was generated by inserting both mutations, along with attenuating mutations E580G and S660A, in PB1 of B/Bris (B/Bris PB1att 4M). Serial passages of the B/Bris PB1att 4M vaccine candidate in eggs and MDCK indicated high stability. In silico structural analysis revealed a potential interaction between amino acids at positions 48 and 391. In mice, B/Bris PB1att 4M was safe and provided complete protection against homologous challenge. These results confirm the compensatory effect of mutation E48K to stabilize the K391E mutation, resulting in a safer, yet still protective, IBV LAIV vaccine.

6.
Virus Genes ; 54(3): 397-405, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29582231

ABSTRACT

Wild birds are natural hosts and reservoirs for influenza A viruses. However, many species, such as many waterfowl, are asymptomatic when infected and so facilitate the generation of viral genetic diversity. Mutations of key genes affect the replicability, pathogenicity, transmissibility, and antiviral resistance of influenza A viruses. In this study, we isolated avian influenza (AI) viruses from wild bird fecal samples and analyzed changes in amino acids over time and geographic region to monitor the biological change of the AI virus. Between 2014 and 2016, we collected 38,921 fresh fecal samples from major wild bird habitats located throughout Korea and isolated 123 AI viruses. We subsequently selected 22 amino acid sites to analyze for changes. These sites included ten sites associated with replication, ten sites associated with pathogenicity, three sites associated with transmission, and seven sites associated with antiviral resistance. We found substitution rates of 71.7% at the C38Y amino acid site within the polymerase basic protein 1 (PB1) gene, 66.7% at the D222G site within the hemagglutinin (HA) 1 gene, and 75.6% at the A184 site within the nucleoprotein (NP) gene. Alterations of the PB1, HA1, and NP genes are closely associated with increased pathogenicity in chickens and mammals. The remaining sites of interest exhibited few modifications. In this study, we confirmed that AI viruses circulating among wild birds in Korea consistently exhibit modifications at amino acid sites linked with replication and pathogenicity.


Subject(s)
Amino Acid Substitution , Birds/virology , Influenza A virus/genetics , Animals , Animals, Wild/virology , Feces/virology , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Mutation , RNA, Viral , Real-Time Polymerase Chain Reaction , Sequence Analysis, Protein , Sequence Analysis, RNA , Virus Replication/genetics
7.
Avian Dis ; 60(1 Suppl): 172-7, 2016 05.
Article in English | MEDLINE | ID: mdl-27309052

ABSTRACT

Since the first outbreak of low pathogenic avian influenza (LPAI) in 1996, outbreaks of LPAI have become more common in Korea, leading to the development of a nationwide mass vaccination program in 2007. In the case of highly pathogenic avian influenza (HPAI), four outbreaks took place in 2003-04, 2006-07, 2008, and 2010-11; a fifth outbreak began in 2014 and was ongoing at the time of this writing. The length of the four previous outbreaks varied, ranging from 42 days (2008) to 139 days (2010-11). The number of cases reported by farmers that were subsequently confirmed as HPAI also varied, from seven cases in 2006-07 to 53 in 2010-11. The number of farms affected by the outbreaks varied, from a low of 286 (2006-07) with depopulation of 6,473,000 birds, to a high of 1500 farms (2008) with depopulation of 10,200,000 birds. Government compensation for bird depopulation ranged from $253 million to $683 million in the five outbreaks. Despite the damage caused by the five HPAI outbreaks, efficient control strategies have yet to be established. Meanwhile, the situation in the field worsens. Analysis of the five HPAI outbreaks revealed horizontal farm-to-farm transmission as the main factor effecting major economic losses. However, horizontal transmission could not be efficiently prevented because of insufficient transparency within the poultry industry, especially within the duck industry, which is reluctant to report suspicious cases early. Moreover, the experiences and expertise garnered in previous outbreaks has yet to be effectively applied to the management of new outbreaks. Considering the magnitude of the economic damage caused by avian influenza and the increasing likelihood of its endemicity, careful and quantitative analysis of outbreaks and the establishment of control policies are urgently needed.


Subject(s)
Influenza A virus/isolation & purification , Influenza in Birds/epidemiology , Poultry Diseases/epidemiology , Animals , Birds , Disease Outbreaks/history , History, 21st Century , Influenza A virus/classification , Influenza A virus/genetics , Influenza in Birds/history , Influenza in Birds/virology , Poultry Diseases/history , Poultry Diseases/virology , Republic of Korea/epidemiology
8.
J Vet Sci ; 17(3): 323-30, 2016 Sep 30.
Article in English | MEDLINE | ID: mdl-26726018

ABSTRACT

Infectious coryza (IC) is an infectious disease caused by Avibacterium (Av.) paragallinarum. IC is known to cause economic losses in the poultry industry via decreased egg production in layers. Between 2012 and 2013, Av. paragallinarum was isolated from seven chicken farms by Chungbuk National University. We identified Av. paragallinarum, the causative pathogen of IC by polymerase chain reaction (PCR) and serovar serotype A, by multiplex PCR. Antibiotic sensitivity tests indicated that a few field-isolated strains showed susceptibility to erythromycin, gentamicin, lincomycin, neomycin, oxytetracycline, spectinomycin, and tylosin. A serological survey was conducted to evaluate the number of flocks that were positive for Av. paragallinarum by utilizing a HI test to determine the existence of serovar A. Serological surveys revealed high positivity rates of 86.4% in 2009, 78.9% in 2010, 70.0% in 2011, and 69.6% in 2012. We also challenged specific pathogen-free chickens with isolated domestic strains, ADL121286 and ADL121500, according to the measured efficacy of the commercial IC vaccine, PoulShot Coryza. We confirmed the effectiveness of the vaccine based on relief of clinical signs and a decreased re-isolation rate of ADL121500 strain. Our results indicate IC is currently prevalent in Korea, and that the commercial vaccine is effective at protecting against field strains.


Subject(s)
Chickens , Haemophilus Infections/veterinary , Haemophilus paragallinarum/physiology , Poultry Diseases/epidemiology , Viral Vaccines/pharmacology , Animals , Haemophilus Infections/epidemiology , Haemophilus Infections/prevention & control , Haemophilus Infections/virology , Haemophilus paragallinarum/genetics , Haemophilus paragallinarum/immunology , Polymerase Chain Reaction/veterinary , Poultry Diseases/prevention & control , Poultry Diseases/virology , Republic of Korea/epidemiology , Sequence Analysis, DNA/veterinary , Specific Pathogen-Free Organisms
9.
Avian Dis ; 59(1): 175-82, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26292555

ABSTRACT

Outbreaks of highly pathogenic avian influenza (HPAI) virus, subtype H5N8, were observed in two different flocks of local broiler breeder farms and a commercial layer farm in South Korea. Clinically, the cases were characterized by a gradual increase in mortality, slow transmission, and unrecognizable clinical signs of HPAI. Gross observations in both cases included hemorrhagic or necrotic lesions in internal organs, such as serosal and mucosal membranes, spleen, and pancreas. Both cases exhibited similar histopathologic lesions, including multifocal malacia in the brain and multifocal or diffuse necrosis in the spleen and pancreas. Immunohistochemical results indicated that neurons and glial cells in the brain, myocytes in the heart, acinar cells in the pancreas, and mononuclear phagocytic cells in several visceral organs were immunopositive for avian influenza viral antigen. To experimentally reproduce the low pathogenicity and the mortality observed in these two cases, 18 specific-pathogen-free chickens and 18 commercial layers were divided into an H5N8 virus-inoculated group and a contact-exposed group. The mortality of the chickens in the inoculation group was 50%-100%, whereas the mean time to death was delayed or death did not occur in the contact-exposed group. The distributions of the viral antigens and histopathologic lesions in the experimental study were similar to those observed in the field cases. These findings suggest that the H5N8 virus induces a different pattern of pathobiology, including slow transmission and low mortality, compared with that of other HPAI viruses. This is the first pathologic description of natural cases of H5N8 in South Korea, and it may be helpful in understanding the pathobiology of novel H5N8 HPAI viruses.


Subject(s)
Influenza A virus/pathogenicity , Influenza in Birds/virology , Animals , Chickens , Female , Influenza in Birds/epidemiology , Influenza in Birds/pathology , Republic of Korea/epidemiology , Virulence
10.
Avian Pathol ; 44(1): 28-34, 2015.
Article in English | MEDLINE | ID: mdl-25510852

ABSTRACT

Chicken parvovirus (ChPV) is one of the causative agents of viral enteritis. Recently, the genome of the ABU-P1 strain of ChPV was fully sequenced and determined to have a distinct genomic composition compared with that of vertebrate parvoviruses. However, no comparative sequence analysis of coding regions of ChPVs was possible because of the lack of other sequence information. In this study, we obtained the nucleotide sequences of all genomic coding regions of three ChPVs by polymerase chain reaction using 13 primer sets, and deduced the amino acid sequences from the nucleotide sequences. The non-structural protein 1 (NS1) gene of the three ChPVs showed 95.0 to 95.5% nucleotide sequence identity and 96.5 to 98.1% amino acid sequence identity to those of NS1 from the ABU-P1 strain, respectively, and even higher nucleotide and amino acid similarities to one another. The viral proteins (VP) gene was more divergent between the three ChPV Korean strains and ABU-P1, with 88.1 to 88.3% nucleotide identity and 93.0% amino acid identity. Analysis of the putative tertiary structure of the ChPV VP2 protein showed that variable regions with less than 80% nucleotide similarity between the three Korean strains and ABU-P1 occurred in large loops of the VP2 protein believed to be involved in antigenicity, pathogenicity, and tissue tropism in other parvoviruses. Based on our analysis of full-length coding sequences, we discovered greater variation in ChPV strains than reported previously, especially in partial regions of the VP2 protein.


Subject(s)
Chickens/virology , Genetic Variation , Parvovirus/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/genetics , Molecular Sequence Data , Polymerase Chain Reaction/veterinary , Sequence Analysis, DNA/veterinary , Sequence Homology , Species Specificity
11.
J Vet Sci ; 15(4): 579-82, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24962418

ABSTRACT

Eggs exhibiting eggshell apex abnormalities (EAA) were evaluated for changes in shell characteristics such as strength, thickness, and ultrastructure. Mycoplasma synoviae (MS) infection was confirmed by serological assay along with isolation of MS from the trachea and oviduct. Changes in eggshell quality were shown to be statistically significant (p < 0.01). We also identified ultrastructural changes in the mammillary knob layer by Scanning Electron Microscopy. While eggs may seem to be structurally sound, ultrastructural evaluation showed that affected eggs do not regain their former quality. In our knowledge, this is the first report describing the occurrence of EAA in Korea.


Subject(s)
Egg Shell/ultrastructure , Mycoplasma Infections/veterinary , Mycoplasma synoviae/physiology , Poultry Diseases/microbiology , Animals , Chickens , Egg Shell/microbiology , Microscopy, Electron, Scanning/veterinary , Mycoplasma Infections/microbiology , Republic of Korea
SELECTION OF CITATIONS
SEARCH DETAIL
...