Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.201
Filtrar
1.
Euro Surveill ; 29(42)2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39421953

RESUMEN

The emergence of clade 2.3.4.4b H5N1 highly pathogenic avian influenza (HPAI) viruses in 2021 has led to unprecedented epidemics in poultry, changing epidemiological patterns of year-round infections in resident wild avifauna and more frequent spill-over events to mammals. Given this situation, it is important that we recognise that traditional HPAI management strategies are no longer sufficient, and policy changes are required. Poultry vaccination has emerged as a crucial intervention in the current control of HPAI, as evidenced by France's nationwide campaign targeting domestic ducks. However, due to the logistical challenges and potential trade implications of vaccination, broader structural reforms appear also necessary. These include a shift from farm-level to territorial-level biosecurity approaches, putting into practice the concept of 'regional biosecurity'. Given the role duck farm density has played in successive HPAI epidemics in France, there is a need to think about the spatial distribution of poultry farms as a structural component of regional biosecurity and to consider the reduction of farm concentration as a measure to prevent viral spread. The integration of regional biosecurity and poultry vaccination into prevention strategies should impact the way poultry are produced and traded in the future.


Asunto(s)
Gripe Aviar , Aves de Corral , Vacunación , Animales , Gripe Aviar/prevención & control , Gripe Aviar/epidemiología , Aves de Corral/virología , Vacunación/veterinaria , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Patos/virología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Brotes de Enfermedades/prevención & control , Francia/epidemiología , Humanos , Crianza de Animales Domésticos , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/virología
2.
Influenza Other Respir Viruses ; 18(10): e70010, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39420513

RESUMEN

The current highly pathogenic avian influenza H5N1 panzootic is having substantial impacts on wild birds and marine mammals. Following major and widespread outbreaks in South America, an incursion to Antarctica occurred late in the austral summer of 2023/2024 and was confined to the region of the Antarctic Peninsula. To infer potential underlying processes, we compiled H5N1 surveillance data from Antarctica and sub-Antarctic Islands prior to the first confirmed cases.


Asunto(s)
Aves , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Regiones Antárticas , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Animales , Gripe Aviar/virología , Gripe Aviar/epidemiología , Aves/virología , Brotes de Enfermedades , Humanos , Gripe Humana/virología , Gripe Humana/epidemiología
3.
Viruses ; 16(9)2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39339917

RESUMEN

Since early 2024, a multistate outbreak of highly pathogenic avian influenza H5N1 has been affecting dairy cattle in the USA. The influenza viral RNA concentrations in milk make it an ideal matrix for surveillance purposes. However, viral RNA detection in multi-component fluids such as milk can be complex, and optimization of influenza detection methods is thus required. Raw bulk tank milk and mastitis milk samples were artificially contaminated with an avian influenza strain and subjected to five extraction methods. HCoV-229E and synthetic RNA were included as exogenous internal process controls. Given the high viral load usually observed in individual raw milk samples, four out of five tested methods would enable influenza detection in milk with normal texture, over a time window of at least 2 weeks post-onset of clinical signs. Nevertheless, sample dilution 1:3 in molecular transport medium prior to RNA extraction provided the best results for dilution of inhibitory substances and a good recovery rate of influenza RNA, that reached 12.5 ± 1.2% and 10.4 ± 3.8% in two independent experiments in bulk milk and 11.2 ± 3.6% and 10.0 ± 2.9% on two cohorts of mastitis milk samples. We have also shown compatibility of an influenza RT-qPCR system with synthetic RNA detection for simultaneous validation of the RNA extraction and RT-qPCR processes.


Asunto(s)
Leche , ARN Viral , Animales , Leche/virología , ARN Viral/aislamiento & purificación , ARN Viral/genética , ARN Viral/análisis , Bovinos , Femenino , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar/virología , Gripe Aviar/diagnóstico , Carga Viral , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/genética
4.
MMWR Morb Mortal Wkly Rep ; 73(37): 804-809, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39298357

RESUMEN

As part of the response to the highly pathogenic avian influenza A(H5N1) virus outbreak in U.S. cattle and poultry and the associated human cases, CDC and partners are monitoring influenza A virus levels and detection of the H5 subtype in wastewater. Among 48 states and the District of Columbia that performed influenza A testing of wastewater during May 12-July 13, 2024, a weekly average of 309 sites in 38 states had sufficient data for analysis, and 11 sites in four states reported high levels of influenza A virus. H5 subtype testing was conducted at 203 sites in 41 states, with H5 detections at 24 sites in nine states. For each detection or high level, CDC and state and local health departments evaluated data from other influenza surveillance systems and partnered with wastewater utilities and agriculture departments to investigate potential sources. Among the four states with high influenza A virus levels detected in wastewater, three states had corresponding evidence of human influenza activity from other influenza surveillance systems. Among the 24 sites with H5 detections, 15 identified animal sources within the sewershed or adjacent county, including eight milk-processing inputs. Data from these early investigations can help health officials optimize the use of wastewater surveillance during the upcoming respiratory illness season.


Asunto(s)
Brotes de Enfermedades , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Aves de Corral , Aguas Residuales , Animales , Humanos , Aguas Residuales/virología , Bovinos , Estados Unidos/epidemiología , Gripe Humana/epidemiología , Gripe Humana/virología , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/epidemiología , Gripe Aviar/virología , Virus de la Influenza A/aislamiento & purificación , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/virología , Monitoreo Epidemiológico Basado en Aguas Residuales , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/virología
5.
Nat Commun ; 15(1): 7433, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39227574

RESUMEN

Until recent events, the Antarctic was the only major geographical region in which high pathogenicity avian influenza virus (HPAIV) had never previously been detected. Here we report on the detection of clade 2.3.4.4b H5N1 HPAIV in the Antarctic and sub-Antarctic regions of South Georgia and the Falkland Islands, respectively. We initially detected H5N1 HPAIV in samples collected from brown skuas at Bird Island, South Georgia on 8th October 2023. Since this detection, mortalities were observed in several avian and mammalian species at multiple sites across South Georgia. Subsequent testing confirmed H5N1 HPAIV across several sampling locations in multiple avian species and two seal species. Simultaneously, we also confirmed H5N1 HPAIV in southern fulmar and black-browed albatross in the Falkland Islands. Genetic assessment of the virus indicates spread from South America, likely through movement of migratory birds. Critically, genetic assessment of sequences from mammalian species demonstrates no increased risk to human populations above that observed in other instances of mammalian infections globally. Here we describe the detection, species impact and genetic composition of the virus and propose both introductory routes and potential long-term impact on avian and mammalian species across the Antarctic region. We also speculate on the threat to specific populations following recent reports in the area.


Asunto(s)
Aves , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Filogenia , Animales , Regiones Antárticas , Gripe Aviar/virología , Gripe Aviar/epidemiología , Gripe Aviar/transmisión , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Aves/virología , Phocidae/virología , Mamíferos/virología
7.
Emerg Microbes Infect ; 13(1): 2406291, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39287422

RESUMEN

The ongoing panzootic of highly pathogenic H5 clade 2.3.4.4b avian influenza (HPAI) spread to North America in late 2021, with detections of HPAI viruses in Alaska beginning in April 2022. HPAI viruses have since spread across the state, affecting many species of wild birds as well as domestic poultry and wild mammals. To better understand the dissemination of HPAI viruses spatiotemporally and among hosts in Alaska and adjacent regions, we compared the genomes of 177 confirmed HPAI viruses detected in Alaska during April-December 2022. Results suggest multiple viral introductions into Alaska between November 2021 and August or September 2022, as well as dissemination to areas within and outside of the state. Viral genotypes differed in their spatiotemporal spread, likely influenced by timing of introductions relative to population immunity. We found evidence for dissemination of HPAI viruses between wild bird species, wild birds and domestic poultry, as well as wild birds and wild mammals. Continued monitoring for and genomic characterization of HPAI viruses in Alaska can improve our understanding of the evolution and dispersal of these economically costly and ecologically relevant pathogens.


Asunto(s)
Animales Salvajes , Aves , Genoma Viral , Genotipo , Gripe Aviar , Filogenia , Animales , Gripe Aviar/virología , Gripe Aviar/epidemiología , Alaska/epidemiología , Aves/virología , Animales Salvajes/virología , Aves de Corral/virología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Genómica , Análisis Espacio-Temporal , Mamíferos/virología , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/patogenicidad
8.
ACS Appl Mater Interfaces ; 16(38): 50360-50368, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39285756

RESUMEN

A one-step logical analysis of multiple targets remains challenging. Herein, we report a one-pot and intelligent DNA logical analysis platform for the diagnosis of avian influenza virus (AIV) biomarkers based on chemiluminescence resonance energy transfer (CRET) and logic operations. On the surface of Lum/PEI/CaCO3 microparticles, the excited state of luminol underwent CRET with fluorescein isothiocyanate or rhodamine B isothiocyanate, producing three well-separated light emissions at 425, 530, and 590 nm, respectively. Taking advantage of the close distance between fluorophores aligned by the catalytic hairpin assembly reaction, the CRET efficiency was greatly enhanced (53.1%). H1N1, H7N9, and H5N1 were detected with limits of detection values as low as 15, 34, and 58 pM, respectively. Three-input logic circuits were simultaneously conducted on the surface of Lum/PEI/CaCO3 microparticles, enabling the rapid and accurate discrimination of multiple AIV biomarkers in one solution. In terms of peak positions and the normalized value of the total peak intensity, three biomarkers can be simultaneously discriminated without any other complex operations. In summary, the CRET-based multiple analytical assay was developed as an intelligent biosensor for identifying AIV biomarkers, having promising application prospects in the field of multiple analysis and precise disease diagnosis.


Asunto(s)
Biomarcadores , Técnicas Biosensibles , Subtipo H1N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Biomarcadores/análisis , Técnicas Biosensibles/métodos , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Animales , Gripe Aviar/virología , Gripe Aviar/diagnóstico , Aves/virología , Subtipo H7N9 del Virus de la Influenza A/aislamiento & purificación , ADN Viral/análisis , Mediciones Luminiscentes/métodos
10.
J Infect Dis ; 230(3): 533-542, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39283944

RESUMEN

Since the resurgence of highly pathogenic avian influenza (HPAI) A(H5N1) virus, clade 2.3.4.4b, during 2021, these viruses have spread widely among birds worldwide, causing poultry outbreaks and infections of a wide range of terrestrial and marine mammal species. During 2024, HPAI A(H5N1) virus, clade 2.3.4.4b, was detected in dairy cattle for the first time and caused an ongoing multistate outbreak, with high levels of virus documented in raw cow milk. Human infections with clade 2.3.4.4b viruses from exposures to infected poultry or dairy cattle have resulted in a wide spectrum of illness severity, from conjunctivitis or mild respiratory illness to severe and fatal pneumonia in different countries. Vigilance, and stronger global virologic surveillance among birds, poultry, terrestrial and marine mammals, and humans, with virus characterization and rapid data sharing, is needed to inform the threat of clade 2.3.4.4b viruses, as they continue to evolve, to public health.


Asunto(s)
Aves , Brotes de Enfermedades , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Animales , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N1 del Virus de la Influenza A/clasificación , Gripe Aviar/virología , Gripe Aviar/epidemiología , Humanos , Gripe Humana/virología , Gripe Humana/epidemiología , Aves/virología , Aves de Corral/virología , Bovinos , Filogenia
11.
MMWR Morb Mortal Wkly Rep ; 73(34): 734-739, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39207932

RESUMEN

Persons who work in close contact with dairy cattle and poultry that are infected with highly pathogenic avian influenza (HPAI) A(H5N1) virus are at increased risk for infection. In July 2024, the Colorado Department of Public Health & Environment responded to two poultry facilities with HPAI A(H5N1) virus detections in poultry. Across the two facilities, 663 workers assisting with poultry depopulation (i.e., euthanasia) received screening for illness; 109 (16.4%) reported symptoms and consented to testing. Among those who received testing, nine (8.3%) received a positive influenza A(H5) virus test result, and 19 (17.4%) received a positive SARS-CoV-2 test result. All nine workers who received positive influenza A(H5) test results had conjunctivitis, experienced mild illness, and received oseltamivir. This poultry exposure-associated cluster of human cases of influenza A(H5) is the first reported in the United States. The identification of these cases highlights the ongoing risk to persons who work in close contact with infected animals. Early response to each facility using multidisciplinary, multilingual teams facilitated case-finding, worker screening, and treatment. As the prevalence of HPAI A(H5N1) virus clade 2.3.4.4b genotype B3.13 increases, U.S. public health agencies should prepare to rapidly investigate and respond to illness in agricultural workers, including workers with limited access to health care.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Exposición Profesional , Aves de Corral , Animales , Humanos , Colorado/epidemiología , Gripe Humana/epidemiología , Adulto , Exposición Profesional/efectos adversos , Masculino , Persona de Mediana Edad , Femenino , Gripe Aviar/epidemiología , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Adulto Joven
12.
Microb Pathog ; 195: 106895, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39208965

RESUMEN

Deadly outbreaks among poultry, wild birds, and carnivorous mammals by the highly pathogenic H5N1 virus of the clade 2.3.4.4b have been reported in South America. The increasing virus incidence in various mammal species poses a severe zoonotic and pandemic threat. In Uruguay, the clade 2.3.4.4b viruses were first detected in February 2023, affecting wild birds and backyard poultry. Three months after the first reported case in Uruguay, the disease affected a population of 23 coatis (Nasua) in an ecological park. Most animals became infected, likely directly or indirectly from wild birds in the park, and experienced sudden death. Five animals from the colony survived, and four of them developed antibodies. The genomes of the H5N1 strains infecting coatis belonged to the B3.2 genotype of the clade 2.3.4.4b. Genomes from coatis were closely associated with those infecting backyard poultry, but transmission likely occurred through wild birds. Notable, two genomes have a 627K substitution in the RNA polymerase PB2 subunit, a hallmark amino acid linked to mammalian adaptation. Our findings support the ability of the avian influenza virus of the 2.3.4.4b clade to infect and transmit among terrestrial mammals with high pathogenicity and undergo rapid adaptive changes. It also highlights the coatis' ability to develop immunity and naturally clear the infection.


Asunto(s)
Animales Salvajes , Genoma Viral , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Mutación , Filogenia , Procyonidae , Animales , Procyonidae/virología , Gripe Aviar/virología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Genoma Viral/genética , Uruguay , Animales Salvajes/virología , Aves/virología , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/veterinaria , Aves de Corral/virología , Genotipo , Mamíferos/virología , América del Sur , Brotes de Enfermedades/veterinaria
13.
Emerg Microbes Infect ; 13(1): 2396874, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39193629

RESUMEN

We detected highly pathogenic avian influenza A(H5N1) virus in wild rats collected from a rural area in Giza, Egypt, near poultry farms, markets, and backyard flocks. Sequence and phylogenetic analyses indicated that the virus from the rats belonged to clade 2.3.4.4b, which has been the predominant virus genotype circulating in Egypt and worldwide since 2021-2022. Active surveillance of avian influenza viruses in wild and domestic mammals is recommended to prevent further spread to mammals and humans.


Asunto(s)
Animales Salvajes , Subtipo H5N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Filogenia , Animales , Egipto/epidemiología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Ratas/virología , Animales Salvajes/virología , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/epidemiología , Genotipo , Gripe Aviar/virología , Gripe Aviar/epidemiología
15.
J Clin Virol ; 174: 105723, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39213758

RESUMEN

On March 25, 2024 an outbreak of highly pathogenic avian influenza (HPAI) A H5N1 was identified in dairy cows across multiple farms in the United States. Zoonotic cases originating in individuals with close contact to infected herds and poultry flocks have been subsequently identified. Spillover events such as this raise the specter of recent pandemics including COVID-19 and Mpox and may lead clinical laboratories to assess their capacity for diagnosis of HPAI H5N1. In this review, we detail the origins of the H5N1 clade 2.3.4.4b outbreak as well as the existing capacity to identify HPAI H5N1 as influenza A virus by commercially available assays. Furthermore, we highlight the absence of commercially available influenza A H5 subtyping assays and limitations associated with the current 510(k)-cleared assay. This outbreak also serves as an early opportunity to assess the new and unknown regulatory challenges faced by laboratory-developed tests in light of the FDA's final rule on in vitro diagnostic devices. National agencies along with public health and clinical laboratories all serve an essential role in the response to HPAI H5N1. To most effectively utilize each group's strength requires open communication and willingness to embrace novel approaches.


Asunto(s)
Brotes de Enfermedades , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Animales , Estados Unidos/epidemiología , Humanos , Gripe Humana/epidemiología , Gripe Humana/virología , Gripe Humana/diagnóstico , Gripe Aviar/virología , Gripe Aviar/epidemiología , Aves de Corral/virología , Laboratorios
16.
Emerg Microbes Infect ; 13(1): 2399268, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39207215

RESUMEN

High pathogenicity avian influenza (HPAI) virus H5N1 first emerged in Bangladesh in 2007. Despite the use of vaccines in chickens since 2012 to control HPAI, HPAI H5Nx viruses have continued to infect poultry, and wild birds, resulting in notable mass mortalities in house crows (Corvus splendens). The first HPAI H5Nx viruses in Bangladesh belonged to clade 2.2.2, followed by clade 2.3.4.2 and 2.3.2.1 viruses in 2011. After the implementation of chicken vaccination in 2012, these viruses were mostly replaced by clade 2.3.2.1a viruses and more recently clade 2.3.4.4b and h viruses. In this study, we reconstruct the phylogenetic history of HPAI H5Nx viruses in Bangladesh to evaluate the role of major host species in the maintenance and evolution of HPAI H5Nx virus in Bangladesh and reveal the role of heavily impacted crows in virus epidemiology. Epizootic waves caused by HPAI H5N1 and H5N6 viruses amongst house crows occurred annually in winter. Bayesian phylodynamic analysis of clade 2.3.2.1a revealed frequent bidirectional viral transitions between domestic ducks, chickens, and house crows that was markedly skewed towards ducks; domestic ducks might be the source, or reservoir, of HPAI H5Nx in Bangladesh, as the number of viral transitions from ducks to chickens and house crows was by far more numerous than the other transitions. Our results suggest viral circulation in domestic birds despite vaccination, with crow epizootics acting as a sentinel. The vaccination strategy needs to be updated to use more effective vaccinations, assess vaccine efficacy, and extension of vaccination to domestic ducks, the key reservoir.


Asunto(s)
Pollos , Reservorios de Enfermedades , Patos , Gripe Aviar , Filogenia , Animales , Gripe Aviar/virología , Gripe Aviar/epidemiología , Gripe Aviar/prevención & control , Patos/virología , Bangladesh/epidemiología , Reservorios de Enfermedades/virología , Pollos/virología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Cuervos/virología , Animales Salvajes/virología , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/clasificación , Virus de la Influenza A/inmunología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/prevención & control
17.
Talanta ; 280: 126704, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39151319

RESUMEN

The outbreak of highly pathogenic influenza virus subtypes, such as H7 and H5, presents a significant global health challenge, necessitating the development of rapid and sensitive diagnostic methods. In this study, we have developed a novel dual-component biosensor assembly, each component of which incorporates an antibody fused with a nano-luciferase subunit. Our results demonstrate the effectiveness of this biosensor in enabling the rapid and sensitive detection of influenza H7 and other subtypes. Additionally, we successfully applied the biosensor in paper-based assay and lateral flow assay formats, expanding its versatility and potential for field-deployable applications. Notably, we achieved effective detection of the H7N9 virus using this biosensor. Furthermore, we designed and optimized a dedicated biosensor to the sensitive detection of the influenza H5 subtype. Collectively, our findings underscore the significant potential of this dual-component biosensor assembly as a valuable and versatile tool for accurate and timely diagnosis of influenza virus infections, promising to advance the field of influenza diagnostics and contribute to outbreak management and surveillance efforts.


Asunto(s)
Técnicas Biosensibles , Técnicas Biosensibles/métodos , Humanos , Gripe Humana/diagnóstico , Glicoproteínas Hemaglutininas del Virus de la Influenza/análisis , Subtipo H7N9 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Luciferasas/química , Luciferasas/metabolismo , Luciferasas/genética
19.
Emerg Microbes Infect ; 13(1): 2392667, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39143912

RESUMEN

Surveillance data from wildlife and poultry was used to describe the spread of highly pathogenic avian influenza (HPAI) H5N1 clade 2.3.4.4b in British Columbia (B.C.) and the Yukon, Canada from September 2022 - June 2023 compared to the first "wave" of the outbreak in this region, which occurred April - August 2022, after the initial viral introduction. Although the number of HPAI-positive poultry farms and wildlife samples was greater in "Wave 2", cases were more tightly clustered in southwestern B.C. and the most commonly affected species differed, likely due to an influx of overwintering waterfowl in the area. Eight HPAI genetic clusters, representing seven genotypes and two inter-continental viral incursions, were detected, with significant variation in the relative abundance of each cluster between the waves. Phylogenetic data suggests multiple spillover events from wild birds to poultry and mammals but could not rule out transmission among farms and among mammals.


Asunto(s)
Animales Salvajes , Aves , Brotes de Enfermedades , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Filogenia , Aves de Corral , Animales , Colombia Británica/epidemiología , Gripe Aviar/virología , Gripe Aviar/epidemiología , Aves de Corral/virología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/clasificación , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Animales Salvajes/virología , Brotes de Enfermedades/veterinaria , Aves/virología , Genotipo , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA