Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.039
Filter
1.
BMC Vet Res ; 20(1): 216, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773480

ABSTRACT

BACKGROUND: In this study, we investigated the prevalence of respiratory viruses in four Hybrid Converter Turkey (Meleagris gallopavo) farms in Egypt. The infected birds displayed severe respiratory signs, accompanied by high mortality rates, suggesting viral infections. Five representative samples from each farm were pooled and tested for H5 & H9 subtypes of avian influenza viruses (AIVs), Avian Orthoavulavirus-1 (AOAV-1), and turkey rhinotracheitis (TRT) using real-time RT-PCR and conventional RT-PCR. Representative tissue samples from positive cases were subjected to histopathology and immunohistochemistry (IHC). RESULTS: The PCR techniques confirmed the presence of AOAV-1 and H5 AIV genes, while none of the tested samples were positive for H9 or TRT. Microscopic examination of tissue samples revealed congestion and hemorrhage in the lungs, liver, and intestines with leukocytic infiltration. IHC revealed viral antigens in the lungs, liver, and intestines. Phylogenetic analysis revealed that H5 HA belonged to 2.3.4.4b H5 sublineage and AOAV-1 belonged to VII 1.1 genotype. CONCLUSIONS: The study highlights the need for proper monitoring of hybrid converter breeds for viral diseases, and the importance of vaccination programs to prevent unnecessary losses. To our knowledge, this is the first study that reports the isolation of AOAV-1 and H5Nx viruses from Hybrid Converter Turkeys in Egypt.


Subject(s)
Influenza in Birds , Phylogeny , Poultry Diseases , Animals , Poultry Diseases/virology , Poultry Diseases/epidemiology , Poultry Diseases/pathology , Influenza in Birds/virology , Influenza in Birds/pathology , Influenza in Birds/epidemiology , Egypt/epidemiology , Turkeys/virology , Influenza A virus/isolation & purification , Influenza A virus/genetics , Influenza A virus/classification
2.
BMC Vet Res ; 20(1): 203, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755641

ABSTRACT

BACKGROUND: Avian influenza virus (AIV) not only causes huge economic losses to the poultry industry, but also threatens human health. Reverse transcription recombinase-aided amplification (RT-RAA) is a novel isothermal nucleic acid amplification technology. This study aimed to improve the detection efficiency of H5, H7, and H9 subtypes of AIV and detect the disease in time. This study established RT-RAA-LFD and real-time fluorescence RT-RAA (RF-RT-RAA) detection methods, which combined RT-RAA with lateral flow dipstick (LFD) and exo probe respectively, while primers and probes were designed based on the reaction principle of RT-RAA. RESULTS: The results showed that RT-RAA-LFD could specifically amplify H5, H7, and H9 subtypes of AIV at 37 °C, 18 min, 39 °C, 20 min, and 38 °C, 18 min, respectively. The sensitivity of all three subtypes for RT-RAA-LFD was 102 copies/µL, which was 10 ∼100 times higher than that of reverse transcription polymerase chain reaction (RT-PCR) agarose electrophoresis method. RF-RT-RAA could specifically amplify H5, H7, and H9 subtypes of AIV at 40 °C, 20 min, 38 °C, 16 min, and 39 °C, 17 min, respectively. The sensitivity of all three subtypes for RF-RT-RAA was 101 copies/µL, which was consistent with the results of real-time fluorescence quantification RT-PCR, and 100 ∼1000 times higher than that of RT-PCR-agarose electrophoresis method. The total coincidence rate of the two methods and RT-PCR-agarose electrophoresis in the detection of clinical samples was higher than 95%. CONCLUSIONS: RT-RAA-LFD and RF-RT-RAA were successfully established in this experiment, with quick response, simple operation, strong specificity, high sensitivity, good repeatability, and stability. They are suitable for the early and rapid diagnosis of Avian influenza and they have positive significance for the prevention, control of the disease, and public health safety.


Subject(s)
Chickens , Influenza A virus , Influenza in Birds , Nucleic Acid Amplification Techniques , Recombinases , Reverse Transcription , Animals , Influenza in Birds/virology , Influenza in Birds/diagnosis , Nucleic Acid Amplification Techniques/veterinary , Nucleic Acid Amplification Techniques/methods , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Recombinases/metabolism , Sensitivity and Specificity , Poultry Diseases/virology , Poultry Diseases/diagnosis
3.
J Infect ; 88(6): 106164, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692359

ABSTRACT

OBJECTIVES: We evaluated Nanopore sequencing for influenza surveillance. METHODS: Influenza A and B PCR-positive samples from hospital patients in Oxfordshire, UK, and a UK-wide population survey from winter 2022-23 underwent Nanopore sequencing following targeted rt-PCR amplification. RESULTS: From 941 infections, successful sequencing was achieved in 292/388 (75 %) available Oxfordshire samples: 231 (79 %) A/H3N2, 53 (18 %) A/H1N1, and 8 (3 %) B/Victoria and in 53/113 (47 %) UK-wide samples. Sequencing was more successful at lower Ct values. Most same-sample replicate sequences had identical haemagglutinin segments (124/141, 88 %); 36/39 (92 %) Illumina vs. Nanopore comparisons were identical, and 3 (8 %) differed by 1 variant. Comparison of Oxfordshire and UK-wide sequences showed frequent inter-regional transmission. Infections were closely-related to 2022-23 vaccine strains. Only one sample had a neuraminidase inhibitor resistance mutation. 849/941 (90 %) Oxfordshire infections were community-acquired. 63/88 (72 %) potentially healthcare-associated cases shared a hospital ward with ≥ 1 known infectious case. 33 epidemiologically-plausible transmission links had sequencing data for both source and recipient: 8 were within ≤ 5 SNPs, of these, 5 (63 %) involved potential sources that were also hospital-acquired. CONCLUSIONS: Nanopore influenza sequencing was reproducible and antiviral resistance rare. Inter-regional transmission was common; most infections were genomically similar. Hospital-acquired infections are likely an important source of nosocomial transmission and should be prioritised for infection prevention and control.


Subject(s)
Influenza B virus , Influenza, Human , Nanopore Sequencing , Humans , Influenza, Human/epidemiology , Influenza, Human/virology , United Kingdom/epidemiology , Nanopore Sequencing/methods , Influenza B virus/genetics , Influenza B virus/isolation & purification , Influenza B virus/classification , Female , Male , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Adult , Middle Aged , Adolescent , Aged , Young Adult , Child , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/classification
4.
Emerg Infect Dis ; 30(6): 1223-1227, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703023

ABSTRACT

Highly pathogenic avian influenza H5N6 and H5N1 viruses of clade 2.3.4.4b were simultaneously introduced into South Korea at the end of 2023. An outbreak at a broiler duck farm consisted of concurrent infection by both viruses. Sharing genetic information and international surveillance of such viruses in wild birds and poultry is critical.


Subject(s)
Disease Outbreaks , Influenza A Virus, H5N1 Subtype , Influenza in Birds , Phylogeny , Influenza in Birds/virology , Influenza in Birds/epidemiology , Republic of Korea/epidemiology , Animals , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Ducks/virology , Influenza A virus/genetics , Influenza A virus/classification , Coinfection/virology , Coinfection/epidemiology , History, 21st Century , Poultry Diseases/virology , Poultry Diseases/epidemiology
5.
Emerg Infect Dis ; 30(6): 1285-1288, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703022

ABSTRACT

We isolated novel reassortant avian influenza A(H5N6) viruses containing genes from clade 2.3.4.4b H5N1 virus and low pathogenicity avian influenza viruses in carcasses of whooper swans and bean geese in South Korea during December 2023. Neuraminidase gene was from a clade 2.3.4.4b H5N6 virus infecting poultry and humans in China.


Subject(s)
Animals, Wild , Birds , Influenza A virus , Influenza in Birds , Phylogeny , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Republic of Korea/epidemiology , Animals, Wild/virology , Influenza A virus/genetics , Influenza A virus/classification , Birds/virology , Reassortant Viruses/genetics , History, 21st Century , Humans , Neuraminidase/genetics
6.
Viruses ; 16(5)2024 04 27.
Article in English | MEDLINE | ID: mdl-38793574

ABSTRACT

Influenza viruses are constantly evolving and are therefore monitored worldwide in the hope to reduce the burden of disease by annual updates to vaccine recommendations. We conducted genomic sequencing of 110 influenza A and 30 influenza B viruses from specimens collected between October 2023 and February 2024 in Arizona, USA. We identified mutations in the hemagglutinin (HA) antigenic sites as well as the neuraminidase (NA) gene in our samples. We also found no unique HA and NA mutations in vaccinated yet influenza-infected individuals. Real-time genomic sequencing surveillance is important to ensure influenza vaccine effectiveness.


Subject(s)
Genome, Viral , Influenza A virus , Influenza B virus , Influenza, Human , Mutation , Neuraminidase , Arizona/epidemiology , Humans , Influenza, Human/epidemiology , Influenza, Human/virology , Neuraminidase/genetics , Influenza B virus/genetics , Influenza A virus/genetics , Influenza A virus/classification , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Genomics/methods , Phylogeny , Adult , Epidemiological Monitoring , Child , Adolescent , Middle Aged , Male , Female , Child, Preschool , Aged , Influenza Vaccines/immunology , Influenza Vaccines/genetics , Young Adult , Whole Genome Sequencing
7.
Viruses ; 16(5)2024 04 28.
Article in English | MEDLINE | ID: mdl-38793579

ABSTRACT

Acute respiratory infections are a major global burden in resource-limited countries, including countries in Africa. Although COVID-19 has been well studied since the pandemic emerged in Gabon, Central Africa, less attention has been paid to other respiratory viral diseases, and very little data are available. Herein, we provide the first data on the genetic diversity and detection of 18 major respiratory viruses in Gabon during the COVID-19 pandemic. Of 582 nasopharyngeal swab specimens collected from March 2020 to July 2021, which were SARS-CoV-2 negative, 156 were positive (26%) for the following viruses: enterovirus (20.3%), human rhinovirus (HRV) (4.6%), human coronavirus OC43 (1.2%), human adenovirus (0.9%), human metapneumovirus (hMPV) (0.5%), influenza A virus (IAV) (0.3%), and human parainfluenza viruses (0.5%). To determine the genetic diversity and transmission route of the viruses, phylogenetic analyses were performed using genome sequences of the detected viruses. The IAV strain detected in this study was genetically similar to strains isolated in the USA, whereas the hMPV strain belonging to the A2b subtype formed a cluster with Kenyan strains. This study provides the first complete genomic sequences of HRV, IAV, and hMPV detected in Gabon, and provides insight into the circulation of respiratory viruses in the country.


Subject(s)
COVID-19 , Genetic Variation , Phylogeny , Respiratory Tract Infections , Humans , Gabon/epidemiology , COVID-19/epidemiology , COVID-19/virology , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , SARS-CoV-2/genetics , SARS-CoV-2/classification , SARS-CoV-2/isolation & purification , Male , Adult , Female , Child , Middle Aged , Adolescent , Child, Preschool , Young Adult , Rhinovirus/genetics , Rhinovirus/isolation & purification , Rhinovirus/classification , Viruses/genetics , Viruses/classification , Viruses/isolation & purification , Metapneumovirus/genetics , Metapneumovirus/isolation & purification , Metapneumovirus/classification , Genome, Viral , Nasopharynx/virology , Infant , Aged , Pandemics , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza A virus/classification
8.
Viruses ; 16(5)2024 05 10.
Article in English | MEDLINE | ID: mdl-38793634

ABSTRACT

Avian influenza viruses (AIVs) of the H5 subtype rank among the most serious pathogens, leading to significant economic losses in the global poultry industry and posing risks to human health. Therefore, rapid and accurate virus detection is crucial for the prevention and control of H5 AIVs. In this study, we established a novel detection method for H5 viruses by utilizing the precision of CRISPR/Cas12a and the efficiency of RT-RPA technologies. This assay facilitates the direct visualization of detection results through blue light and lateral flow strips, accurately identifying H5 viruses with high specificity and without cross-reactivity against other AIV subtypes, NDV, IBV, and IBDV. With detection thresholds of 1.9 copies/µL (blue light) and 1.9 × 103 copies/µL (lateral flow strips), our method not only competes with but also slightly surpasses RT-qPCR, demonstrating an 80.70% positive detection rate across 81 clinical samples. The RT-RPA/CRISPR-based detection method is characterized by high sensitivity, specificity, and independence from specialized equipment. The immediate field applicability of the RT-RPA/CRISPR approach underscores its importance as an effective tool for the early detection and management of outbreaks caused by the H5 subtype of AIVs.


Subject(s)
CRISPR-Cas Systems , Influenza in Birds , Sensitivity and Specificity , Animals , Influenza in Birds/virology , Influenza in Birds/diagnosis , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H5N1 Subtype/classification , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza A virus/classification , Poultry/virology , Poultry Diseases/virology , Poultry Diseases/diagnosis , Chickens/virology , Birds/virology
9.
J Gen Virol ; 105(5)2024 May.
Article in English | MEDLINE | ID: mdl-38695722

ABSTRACT

High-pathogenicity avian influenza viruses (HPAIVs) of the goose/Guangdong lineage are enzootically circulating in wild bird populations worldwide. This increases the risk of entry into poultry production and spill-over to mammalian species, including humans. Better understanding of the ecological and epizootiological networks of these viruses is essential to optimize mitigation measures. Based on full genome sequences of 26 HPAIV samples from Iceland, which were collected between spring and autumn 2022, as well as 1 sample from the 2023 summer period, we show that 3 different genotypes of HPAIV H5N1 clade 2.3.4.4b were circulating within the wild bird population in Iceland in 2022. Furthermore, in 2023 we observed a novel introduction of HPAIV H5N5 of the same clade to Iceland. The data support the role of Iceland as an utmost northwestern distribution area in Europe that might act also as a potential bridging point for intercontinental spread of HPAIV across the North Atlantic.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza in Birds , Phylogeny , Iceland/epidemiology , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Influenza in Birds/transmission , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Genotype , Animals, Wild/virology , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Genome, Viral , Birds/virology
10.
Comp Immunol Microbiol Infect Dis ; 109: 102182, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640701

ABSTRACT

In the 2021/22 winter, one H5N1 and nine H5N8 high pathogenicity avian influenza viruses (HPAIVs) of clade 2.3.3.4b were isolated from the water in crane roosts on the Izumi plain, Japan. Additionally, we isolated low pathogenicity avian influenza viruses (LPAIVs) of five subtypes: H1N1, H4N2, H4N6, H7N7, and H10N4. H5N8 HPAIVs belonging to the G2a group were isolated throughout winter, whereas H5N1 HPAIV belonging to the G2b group were isolated only in early winter. These findings suggest co-circulation of both G2a and G2b HPAIVs in early winter. Although two H7N7 LPAIVs were isolated from cranes' roost water collected on the same day, the gene constellations of the two isolates were clearly different, indicating the contemporary invasion of at least two different genotypes of H7N7 LPAIVs in the Izumi plain. This study underscores the importance of monitoring both HPAIVs and LPAIVs to understand avian influenza virus ecology in migratory waterfowl populations.


Subject(s)
Birds , Genotype , Influenza in Birds , Phylogeny , Seasons , Japan , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Birds/virology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/classification , Water Microbiology , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza A virus/classification , Influenza A Virus, H5N8 Subtype/genetics , Influenza A Virus, H5N8 Subtype/pathogenicity , Influenza A Virus, H5N8 Subtype/isolation & purification , Influenza A Virus, H5N8 Subtype/classification , Influenza A Virus, H7N7 Subtype/genetics , Influenza A Virus, H7N7 Subtype/pathogenicity , Influenza A Virus, H7N7 Subtype/isolation & purification
11.
Viruses ; 16(4)2024 04 07.
Article in English | MEDLINE | ID: mdl-38675910

ABSTRACT

Influenza A viruses (IAVs) pose a serious threat to global health. On the one hand, these viruses cause seasonal flu outbreaks in humans. On the other hand, they are a zoonotic infection that has the potential to cause a pandemic. The most important natural reservoir of IAVs are waterfowl. In this study, we investigated the occurrence of IAV in birds in the Republic of Buryatia (region in Russia). In 2020, a total of 3018 fecal samples were collected from wild migratory birds near Lake Baikal. Of these samples, 11 were found to be positive for the H13N8 subtype and whole-genome sequencing was performed on them. All samples contained the same virus with the designation A/Unknown/Buryatia/Arangatui-1/2020. To our knowledge, virus A/Unknown/Buryatia/Arangatui-1/2020 is the first representative of the H13N8 subtype collected on the territory of Russia, the sequence of which is available in the GenBank database. An analysis of reassortments based on the genome sequences of other known viruses has shown that A/Unknown/Buryatia/Arangatui-1/2020 arose as a result of reassortment. In addition, a reassortment most likely occurred several decades ago between the ancestors of the viruses recently collected in China, the Netherlands, the United States and Chile. The presence of such reassortment emphasizes the ongoing evolution of the H13N8 viruses distributed in Europe, North and East Asia, North and South America and Australia. This study underscores the importance of the continued surveillance and research of less-studied influenza subtypes.


Subject(s)
Birds , Genome, Viral , Influenza A virus , Influenza in Birds , Phylogeny , Reassortant Viruses , Whole Genome Sequencing , Animals , Reassortant Viruses/genetics , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Influenza in Birds/virology , Influenza in Birds/epidemiology , Russia/epidemiology , Birds/virology , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Feces/virology , Animals, Wild/virology
12.
Viruses ; 16(4)2024 04 12.
Article in English | MEDLINE | ID: mdl-38675939

ABSTRACT

The flyways of many different wild waterfowl pass through the Caspian Sea region. The western coast of the middle Caspian Sea is an area with many wetlands, where wintering grounds with large concentrations of birds are located. It is known that wild waterfowl are a natural reservoir of the influenza A virus. In the mid-2000s, in the north of this region, the mass deaths of swans, gulls, and pelicans from high pathogenicity avian influenza virus (HPAIV) were noted. At present, there is still little known about the presence of avian influenza virus (AIVs) and different avian paramyxoviruses (APMVs) in the region's waterfowl bird populations. Here, we report the results of monitoring these viruses in the wild waterfowl of the western coast of the middle Caspian Sea from 2017 to 2020. Samples from 1438 individuals of 26 bird species of 7 orders were collected, from which 21 strains of AIV were isolated, amounting to a 1.46% isolation rate of the total number of samples analyzed (none of these birds exhibited external signs of disease). The following subtypes were determined and whole-genome nucleotide sequences of the isolated strains were obtained: H1N1 (n = 2), H3N8 (n = 8), H4N6 (n = 2), H7N3 (n = 2), H8N4 (n = 1), H10N5 (n = 1), and H12N5 (n = 1). No high pathogenicity influenza virus H5 subtype was detected. Phylogenetic analysis of AIV genomes did not reveal any specific pattern for viruses in the Caspian Sea region, showing that all segments belong to the Eurasian clades of classic avian-like influenza viruses. We also did not find the amino acid substitutions in the polymerase complex (PA, PB1, and PB2) that are critical for the increase in virulence or adaptation to mammals. In total, 23 hemagglutinating viruses not related to influenza A virus were also isolated, of which 15 belonged to avian paramyxoviruses. We were able to sequence 12 avian paramyxoviruses of three species, as follows: Newcastle disease virus (n = 4); Avian paramyxovirus 4 (n = 5); and Avian paramyxovirus 6 (n = 3). In the Russian Federation, the Newcastle disease virus of the VII.1.1 sub-genotype was first isolated from a wild bird (common pheasant) in the Caspian Sea region. The five avian paramyxovirus 4 isolates obtained belonged to the common clade in Genotype I, whereas phylogenetic analysis of three isolates of Avian paramyxovirus 6 showed that two isolates, isolated in 2017, belonged to Genotype I and that an isolate identified in 2020 belonged to Genotype II. The continued regular monitoring of AIVs and APMVs, the obtaining of data on the biological properties of isolated strains, and the accumulation of information on virus host species will allow for the adequate planning of epidemiological measures, suggest the most likely routes of spread of the virus, and assist in the prediction of the introduction of the viruses in the western coastal region of the middle Caspian Sea.


Subject(s)
Animals, Wild , Avulavirus , Birds , Influenza A virus , Influenza in Birds , Phylogeny , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Birds/virology , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Animals, Wild/virology , Avulavirus/genetics , Avulavirus/classification , Avulavirus/isolation & purification , Avulavirus/pathogenicity , Genome, Viral , Avulavirus Infections/veterinary , Avulavirus Infections/virology , Avulavirus Infections/epidemiology
13.
Viruses ; 16(4)2024 04 18.
Article in English | MEDLINE | ID: mdl-38675967

ABSTRACT

Inactivated influenza A virus (IAV) vaccines help reduce clinical disease in suckling piglets, although endemic infections still exist. The objective of this study was to evaluate the detection of IAV in suckling and nursery piglets from IAV-vaccinated sows from farms with endemic IAV infections. Eight nasal swab collections were obtained from 135 two-week-old suckling piglets from four farms every other week from March to September 2013. Oral fluid samples were collected from the same group of nursery piglets. IAV RNA was detected in 1.64% and 31.01% of individual nasal swabs and oral fluids, respectively. H1N2 was detected most often, with sporadic detection of H1N1 and H3N2. Whole-genome sequences of IAV isolated from suckling piglets revealed an H1 hemagglutinin (HA) from the 1B.2.2.2 clade and N2 neuraminidase (NA) from the 2002A clade. The internal gene constellation of the endemic H1N2 was TTTTPT with a pandemic lineage matrix. The HA gene had 97.59% and 97.52% nucleotide and amino acid identities, respectively, to the H1 1B.2.2.2 used in the farm-specific vaccine. A similar H1 1B.2.2.2 was detected in the downstream nursery. These data demonstrate the low frequency of IAV detection in suckling piglets and downstream nurseries from farms with endemic infections in spite of using farm-specific IAV vaccines in sows.


Subject(s)
Farms , Influenza A virus , Influenza Vaccines , Orthomyxoviridae Infections , Phylogeny , Swine Diseases , Animals , Swine , Swine Diseases/virology , Swine Diseases/epidemiology , Swine Diseases/prevention & control , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/epidemiology , Influenza A virus/genetics , Influenza A virus/immunology , Influenza A virus/isolation & purification , Influenza A virus/classification , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Animals, Suckling , Vaccination/veterinary , Endemic Diseases/veterinary , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/isolation & purification , RNA, Viral/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/isolation & purification , Influenza A Virus, H1N2 Subtype/immunology , Genome, Viral
14.
Nature ; 628(8009): 835-843, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38600381

ABSTRACT

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Subject(s)
Lung Injury , Necroptosis , Orthomyxoviridae Infections , Protein Kinase Inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Female , Humans , Male , Mice , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Alveolar Epithelial Cells/metabolism , Influenza A virus/classification , Influenza A virus/drug effects , Influenza A virus/immunology , Influenza A virus/pathogenicity , Lung Injury/complications , Lung Injury/pathology , Lung Injury/prevention & control , Lung Injury/virology , Mice, Inbred C57BL , Necroptosis/drug effects , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/prevention & control , Respiratory Distress Syndrome/virology
15.
J Virol Methods ; 327: 114942, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670532

ABSTRACT

H5, H7 and H9 are the major subtypes of avian influenza virus (AIV) that cause economic losses in the poultry industry and sporadic zoonotic infection. Early detection of AIV is essential for preventing disease spread. Therefore, molecular diagnosis and subtyping of AIV via real-time RT-PCR (rRT-PCR) is preferred over other classical diagnostic methods, such as egg inoculation, RT-PCR and HI test, due to its high sensitivity, specificity and convenience. The singleplex rRT-PCRs for the Matrix, H5 and H7 gene used for the national surveillance program in Korea have been developed in 2017; however, these methods were not designed for multiplexing, and does not reflect the sequences of currently circulating strains completely. In this study, the multiplex H5/7/9 rRT-PCR assay was developed with sets of primers and probe updated or newly designed to simultaneously detect the H5, H7 and H9 genes. Multiplex H5/7/9 rRT-PCR showed 100% specificity without cross-reactivity with other subtypes of AIVs and avian disease-causing viruses or bacteria, and the limit of detection was 1-10 EID50/0.1 ml (50% egg infectious dose). Artificial mixed infections with the three different subtypes could be detected accurately with high analytical sensitivity even under highly biased relative molecular ratios by balancing the reactivities of each subtype by modifying the concentration of the primers and probes. The multiplex H5/7/9 rRT-PCR assay developed in this study could be a useful tool for large-scale surveillance programs for viral detection as well as subtyping due to its high specificity, sensitivity and robustness in discriminating viruses in mixed infections, and this approach would greatly decrease the time, cost, effort and chance of cross-contamination compared to the conventional method of testing three subtypes by different singleplex rRT-PCR methods in parallel or in series.


Subject(s)
Chickens , Influenza A virus , Influenza in Birds , Multiplex Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Influenza in Birds/virology , Influenza in Birds/diagnosis , Animals , Multiplex Polymerase Chain Reaction/methods , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Real-Time Polymerase Chain Reaction/methods , Chickens/virology , Republic of Korea , Poultry Diseases/virology , Poultry Diseases/diagnosis , DNA Primers/genetics , Poultry/virology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Birds/virology
16.
Emerg Microbes Infect ; 13(1): 2341142, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38581279

ABSTRACT

H6N6 avian influenza viruses (AIVs) have been widely detected in wild birds, poultry, and even mammals. Recently, H6N6 viruses were reported to be involved in the generation of H5 and H7 subtype viruses. To investigate the emergence, evolutionary pattern, and potential for an epidemic of H6N6 viruses, the complete genomes of 198 H6N6 viruses were analyzed, including 168 H6N6 viruses deposited in the NCBI and GISAID databases from inception to January 2019 and 30 isolates collected from China between November 2014 and January 2019. Using phylogenetic analysis, the 198 strains of H6N6 viruses were identified as 98 genotypes. Molecular clock analysis indicated that the evolution of H6N6 viruses in China was constant and not interrupted by selective pressure. Notably, the laboratory isolates reassorted with six subtype viruses: H6N2, H5N6, H7N9, H5N2, H4N2, and H6N8, resulting in nine novel H6N6 reassortment events. These results suggested that H6N6 viruses can act as an intermediary in the evolution of H5N6, H6N6, and H7N9 viruses. Animal experiments demonstrated that the 10 representative H6N6 viruses showed low pathogenicity in chickens and were capable of infecting mice without prior adaptation. Our findings suggest that H6N6 viruses play an important role in the evolution of AIVs, and it is necessary to continuously monitor and evaluate the potential epidemic of the H6N6 subtype viruses.


Subject(s)
Chickens , Evolution, Molecular , Genome, Viral , Influenza A virus , Influenza in Birds , Phylogeny , Reassortant Viruses , Animals , China/epidemiology , Reassortant Viruses/genetics , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Influenza in Birds/virology , Influenza in Birds/epidemiology , Mice , Chickens/virology , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Genotype , Humans
17.
J Virol ; 98(4): e0194123, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38470143

ABSTRACT

Influenza A viruses (IAVs) can overcome species barriers by adaptation of the receptor-binding site of the hemagglutinin (HA). To initiate infection, HAs bind to glycan receptors with terminal sialic acids, which are either N-acetylneuraminic acid (NeuAc) or N-glycolylneuraminic acid (NeuGc); the latter is mainly found in horses and pigs but not in birds and humans. We investigated the influence of previously identified equine NeuGc-adapting mutations (S128T, I130V, A135E, T189A, and K193R) in avian H7 IAVs in vitro and in vivo. We observed that these mutations negatively affected viral replication in chicken cells but not in duck cells and positively affected replication in horse cells. In vivo, the mutations reduced virus virulence and mortality in chickens. Ducks excreted high viral loads longer than chickens, although they appeared clinically healthy. To elucidate why these viruses infected chickens and ducks despite the absence of NeuGc, we re-evaluated the receptor binding of H7 HAs using glycan microarray and flow cytometry studies. This re-evaluation demonstrated that mutated avian H7 HAs also bound to α2,3-linked NeuAc and sialyl-LewisX, which have an additional fucose moiety in their terminal epitope, explaining why infection of ducks and chickens was possible. Interestingly, the α2,3-linked NeuAc and sialyl-LewisX epitopes were only bound when presented on tri-antennary N-glycans, emphasizing the importance of investigating the fine receptor specificities of IAVs. In conclusion, the binding of NeuGc-adapted H7 IAV to tri-antennary N-glycans enables viral replication and shedding by chickens and ducks, potentially facilitating interspecies transmission of equine-adapted H7 IAVs.IMPORTANCEInfluenza A viruses (IAVs) cause millions of deaths and illnesses in birds and mammals each year. The viral surface protein hemagglutinin initiates infection by binding to host cell terminal sialic acids. Hemagglutinin adaptations affect the binding affinity to these sialic acids and the potential host species targeted. While avian and human IAVs tend to bind to N-acetylneuraminic acid (sialic acid), equine H7 viruses prefer binding to N-glycolylneuraminic acid (NeuGc). To better understand the function of NeuGc-specific adaptations in hemagglutinin and to elucidate interspecies transmission potential NeuGc-adapted viruses, we evaluated the effects of NeuGc-specific mutations in avian H7 viruses in chickens and ducks, important economic hosts and reservoir birds, respectively. We also examined the impact on viral replication and found a binding affinity to tri-antennary N-glycans containing different terminal epitopes. These findings are significant as they contribute to the understanding of the role of receptor binding in avian influenza infection.


Subject(s)
Chickens , Ducks , Horses , Influenza A virus , Influenza in Birds , Neuraminic Acids , Animals , Humans , Chickens/genetics , Chickens/metabolism , Chickens/virology , Ducks/genetics , Ducks/metabolism , Ducks/virology , Epitopes/chemistry , Epitopes/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Horses/genetics , Horses/metabolism , Horses/virology , Influenza A virus/chemistry , Influenza A virus/classification , Influenza A virus/metabolism , Influenza in Birds/genetics , Influenza in Birds/transmission , Influenza in Birds/virology , Mutation , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Neuraminic Acids/chemistry , Neuraminic Acids/metabolism , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Swine/virology , Viral Zoonoses/metabolism , Viral Zoonoses/transmission , Viral Zoonoses/virology
18.
J Virol ; 98(3): e0170323, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38353535

ABSTRACT

The increased detection of H3 C-IVA (1990.4.a) clade influenza A viruses (IAVs) in US swine in 2019 was associated with a reassortment event to acquire an H1N1pdm09 lineage nucleoprotein (pdmNP) gene, replacing a TRIG lineage NP (trigNP). We hypothesized that acquiring the pdmNP conferred a selective advantage over prior circulating H3 viruses with a trigNP. To investigate the role of NP reassortment in transmission, we identified two contemporary 1990.4.a representative strains (NC/19 and MN/18) with different evolutionary origins of the NP gene. A reverse genetics system was used to generate wild-type (wt) strains and swap the pdm and TRIG lineage NP genes, generating four viruses: wtNC/19-pdmNP, NC/19-trigNP, wtMN/18-trigNP, and MN/18-pdmNP. The pathogenicity and transmission of the four viruses were compared in pigs. All four viruses infected 10 primary pigs and transmitted to five indirect contact pigs per group. Pigs infected via contact with MN/18-pdmNP shed virus 2 days earlier than pigs infected with wtMN/18-trigNP. The inverse did not occur for wtNC/19-pdmNP and NC/19-trigNP. This suggests that pdmNP reassortment resulted in a combination of genes that improved transmission efficiency when paired with the 1990.4.a hemagglutinin (HA). This is likely a multigenic trait, as replacing the trigNP gene did not diminish the transmission of a wild-type IAV in swine. This study demonstrates how reassortment and evolutionary change of internal genes can result in more transmissible viruses that influence HA clade detection frequency. Thus, rapidly identifying novel reassortants paired with dominant hemagglutinin/neuraminidase may improve the prediction of strains to include in vaccines.IMPORTANCEInfluenza A viruses (IAVs) are composed of eight non-continuous gene segments that can reassort during coinfection of a host, creating new combinations. Some gene combinations may convey a selective advantage and be paired together preferentially. A reassortment event was detected in swine in the United States that involved the exchange of two lineages of nucleoprotein (NP) genes (trigNP to pdmNP) that became a predominant genotype detected in surveillance. Using a transmission study, we demonstrated that exchanging the trigNP for a pdmNP caused the virus to shed from the nose at higher levels and transmit to other pigs more rapidly. Replacing a pdmNP with a trigNP did not hinder transmission, suggesting that transmission efficiency depends on interactions between multiple genes. This demonstrates how reassortment alters IAV transmission and that reassortment events can provide an explanation for why genetically related viruses with different internal gene combinations experience rapid fluxes in detection frequency.


Subject(s)
Influenza A virus , Nucleocapsid Proteins , Orthomyxoviridae Infections , Swine Diseases , Animals , Hemagglutinins , Influenza A virus/classification , Influenza A virus/genetics , Orthomyxoviridae Infections/veterinary , Orthomyxoviridae Infections/virology , Reassortant Viruses/genetics , Swine , United States , Nucleocapsid Proteins/metabolism
19.
J Virol ; 98(2): e0149423, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38294251

ABSTRACT

Influenza B viruses (IBV) cocirculate with influenza A viruses (IAV) and cause periodic epidemics of disease, yet antibody and cellular responses following IBV infection are less well understood. Using the ferret model for antisera generation for influenza surveillance purposes, IAV resulted in robust antibody responses following infection, whereas IBV required an additional booster dose, over 85% of the time, to generate equivalent antibody titers. In this study, we utilized primary differentiated ferret nasal epithelial cells (FNECs) which were inoculated with IAV and IBV to study differences in innate immune responses which may result in differences in adaptive immune responses in the host. FNECs were inoculated with IAV (H1N1pdm09 and H3N2 subtypes) or IBV (B/Victoria and B/Yamagata lineages) and assessed for 72 h. Cells were analyzed for gene expression by quantitative real-time PCR, and apical and basolateral supernatants were assessed for virus kinetics and interferon (IFN), respectively. Similar virus kinetics were observed with IAV and IBV in FNECs. A comparison of gene expression and protein secretion profiles demonstrated that IBV-inoculated FNEC expressed delayed type-I/II IFN responses and reduced type-III IFN secretion compared to IAV-inoculated cells. Concurrently, gene expression of Thymic Stromal Lymphopoietin (TSLP), a type-III IFN-induced gene that enhances adaptive immune responses, was significantly downregulated in IBV-inoculated FNECs. Significant differences in other proinflammatory and adaptive genes were suppressed and delayed following IBV inoculation. Following IBV infection, ex vivo cell cultures derived from the ferret upper respiratory tract exhibited reduced and delayed innate responses which may contribute to reduced antibody responses in vivo.IMPORTANCEInfluenza B viruses (IBV) represent nearly one-quarter of all human influenza cases and are responsible for significant clinical and socioeconomic impacts but do not pose the same pandemic risks as influenza A viruses (IAV) and have thus received much less attention. IBV accounts for greater severity and deaths in children, and vaccine efficacy remains low. The ferret can be readily infected with human clinical isolates and demonstrates a similar course of disease and immune responses. IBV, however, generates lower antibodies in ferrets than IAV following the challenge. To determine whether differences in initial innate responses following infection may affect the development of robust adaptive immune responses, ferret respiratory tract cells were isolated, infected with IAV/IBV, and compared. Understanding the differences in the initial innate immune responses to IAV and IBV may be important in the development of more effective vaccines and interventions to generate more robust protective immune responses.


Subject(s)
Adaptive Immunity , Epithelial Cells , Ferrets , Immunity, Innate , Influenza A virus , Influenza B virus , Interferons , Nasal Mucosa , Animals , Child , Humans , Antibodies, Viral/analysis , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/immunology , Epithelial Cells/virology , Ferrets/immunology , Ferrets/virology , Influenza A virus/classification , Influenza A virus/growth & development , Influenza A virus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/classification , Influenza B virus/growth & development , Influenza B virus/immunology , Influenza Vaccines , Influenza, Human/virology , Interferons/immunology , Nasal Mucosa/cytology , Nasal Mucosa/immunology , Nasal Mucosa/virology , Thymic Stromal Lymphopoietin/genetics , Thymic Stromal Lymphopoietin/immunology , Cells, Cultured
20.
J Virol ; 97(11): e0137023, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37877722

ABSTRACT

The H6 subtype of avian influenza virus (AIV) is a pervasive subtype that is ubiquitously found in both wild bird and poultry populations across the globe. Recent investigations have unveiled its capacity to infect mammals, thereby expanding its host range beyond that of other subtypes and potentially facilitating its global transmission. This heightened breadth also endows H6 AIVs with the potential to serve as a genetic reservoir for the emergence of highly pathogenic avian influenza strains through genetic reassortment and adaptive mutations. Furthermore, alterations in key amino acid loci within the H6 AIV genome foster the evolution of viral infection mechanisms, which may enable the virus to surmount interspecies barriers and infect mammals, including humans, thus posing a potential threat to human well-being. In this review, we summarize the origins, dissemination patterns, geographical distribution, cross-species transmission dynamics, and genetic attributes of H6 influenza viruses. This study holds implications for the timely detection and surveillance of H6 AIVs.


Subject(s)
Birds , Host Specificity , Influenza A virus , Influenza in Birds , Mammals , Viral Zoonoses , Animals , Humans , Birds/virology , Influenza A virus/classification , Influenza A virus/genetics , Influenza A virus/isolation & purification , Influenza in Birds/transmission , Influenza in Birds/virology , Mammals/virology , Poultry/virology , Viral Zoonoses/transmission , Viral Zoonoses/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...