Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.161
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39000173

RESUMEN

Tagetes erecta Linn. (TE) is traditionally used to treat cardiovascular, renal, and gastrointestinal diseases. In this study, we investigated the active compounds and targets of TE extract that may exert antiviral effects against influenza A. Active compounds and targets of TE extract were identified using the Traditional Chinese Medicine Systems Pharmacology database (TCSMP). The influenza A-related gene set was screened using GeneCards and the Kyoto Encyclopedia of Genes and Genomes (KEGG). A protein-protein interaction (PPI) network was built to establish the hub targets. Pathway and target studies were conducted using Gene Expression Omnibus (GEO). The interactions between active compounds and potential targets were assessed by molecular docking. An in vitro study was performed using antiviral and plaque reduction assays. From the compound and target search, we identified 6 active compounds and 95 potential targets. We retrieved 887 influenza-associated target genes and determined 14 intersecting core targets between TE and influenza. After constructing a compound-target network, we discovered lutein and beta-carotene to be the key compounds. Next, PPI network analysis identified the top three hub genes associated with influenza (IL-6, HIF1A, and IL-1ß). Similarly, GEO analysis revealed IL-6, TGFB1, and CXCL8 to be the top three target genes. In our docking study, we identified that lutein and IL-6 had the strongest bindings. Our in vitro experimental results revealed that the TE extract exhibited therapeutic rather than prophylactic effects on influenza disease. We identified lutein as a main active compound in TE extract, and IL-6 as an important target associated with influenza, by using data mining and bioinformatics. Our in vitro findings indicated that TE extract exerted protective properties against the influenza A virus. We speculated that lutein, as a key active component in TE extract, is largely responsible for its antiviral effects. Therefore, we suggest TE extract as an alternative in the treatment of influenza.


Asunto(s)
Antivirales , Biología Computacional , Simulación del Acoplamiento Molecular , Extractos Vegetales , Mapas de Interacción de Proteínas , Tagetes , Antivirales/farmacología , Antivirales/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , Biología Computacional/métodos , Mapas de Interacción de Proteínas/efectos de los fármacos , Humanos , Tagetes/química , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/genética , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Animales , Células de Riñón Canino Madin Darby , Perros , Medicina Tradicional China/métodos
2.
J Gen Virol ; 105(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38975739

RESUMEN

The 2020/2021 epidemic in Europe of highly pathogenic avian influenza virus (HPAIV) of subtype H5 surpassed all previously recorded European outbreaks in size, genotype constellations and reassortment frequency and continued into 2022 and 2023. The causative 2.3.4.4b viral lineage proved to be highly proficient with respect to reassortment with cocirculating low pathogenic avian influenza viruses and seems to establish an endemic status in northern Europe. A specific HPAIV reassortant of the subtype H5N3 was detected almost exclusively in red knots (Calidris canutus islandica) in December 2020. It caused systemic and rapidly fatal disease leading to a singular and self-limiting mass mortality affecting about 3500 birds in the German Wadden Sea, roughly 1 % of the entire flyway population of islandica red knots. Phylogenetic analyses revealed that the H5N3 reassortant very likely had formed in red knots and remained confined to this species. While mechanisms of virus circulation in potential reservoir species, dynamics of spill-over and reassortment events and the roles of environmental virus sources remain to be identified, the year-round infection pressure poses severe threats to endangered avian species and prompts adaptation of habitat and species conservation practices.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Filogenia , Virus Reordenados , Animales , Gripe Aviar/virología , Gripe Aviar/epidemiología , Europa (Continente)/epidemiología , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/patogenicidad , Virus Reordenados/genética , Brotes de Enfermedades/veterinaria , Charadriiformes/virología , Aves/virología
3.
Emerg Microbes Infect ; 13(1): 2368202, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38970562

RESUMEN

Influenza A viruses (IAV) impose significant respiratory disease burdens in both swine and humans worldwide, with frequent human-to-swine transmission driving viral evolution in pigs and highlighting the risk at the animal-human interface. Therefore, a comprehensive One Health approach (interconnection among human, animal, and environmental health) is needed for IAV prevention, control, and response. Animal influenza genomic surveillance remains limited in many Latin American countries, including Colombia. To address this gap, we genetically characterized 170 swine specimens from Colombia (2011-2017). Whole genome sequencing revealed a predominance of pandemic-like H1N1 lineage, with a minority belonging to H3N2 and H1N2 human seasonal-like lineage and H1N1 early classical swine lineages. Significantly, we have identified reassortant and recombinant viruses (H3N2, H1N1) not previously reported in Colombia. This suggests a broad genotypic viral diversity, likely resulting from reassortment between classical endemic viruses and new introductions established in Colombia's swine population (e.g. the 2009 H1N1 pandemic). Our study highlights the importance of a One Health approach in disease control, particularly in an ecosystem where humans are a main source of IAV to swine populations, and emphasizes the need for continued surveillance and enhanced biosecurity measures. The co-circulation of multiple subtypes in regions with high swine density facilitates viral exchange, underscoring the importance of monitoring viral evolution to inform vaccine selection and public health policies locally and globally.


Asunto(s)
Evolución Molecular , Variación Genética , Subtipo H1N1 del Virus de la Influenza A , Subtipo H3N2 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , Filogenia , Enfermedades de los Porcinos , Animales , Porcinos , Colombia/epidemiología , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/epidemiología , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/epidemiología , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/clasificación , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/clasificación , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Salud Única , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Secuenciación Completa del Genoma , Genoma Viral , Monitoreo Epidemiológico , Virus Reordenados/genética , Virus Reordenados/clasificación , Virus Reordenados/aislamiento & purificación , Subtipo H1N2 del Virus de la Influenza A/genética , Subtipo H1N2 del Virus de la Influenza A/aislamiento & purificación , Subtipo H1N2 del Virus de la Influenza A/clasificación , Gripe Humana/virología , Gripe Humana/epidemiología
4.
Front Cell Infect Microbiol ; 14: 1433661, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38979510

RESUMEN

In recent years, the avian influenza virus has emerged as a significant threat to both human and public health. This study focuses on a patient infected with the H10N3 subtype of avian influenza virus, admitted to the Third People's Hospital of Kunming City on March 6, 2024. Metagenomic RNA sequencing and polymerase chain reaction (PCR) analysis were conducted on the patient's sputum, confirming the H10N3 infection. The patient presented severe pneumonia symptoms such as fever, expectoration, chest tightness, shortness of breath, and cough. Phylogenetic analysis of the Haemagglutinin (HA) and neuraminidase (NA) genes of the virus showed that the virus was most closely related to a case of human infection with the H10N3 subtype of avian influenza virus found in Zhejiang Province, China. Analysis of amino acid mutation sites identified four mutations potentially hazardous to human health. Consequently, this underscores the importance of continuous and vigilant monitoring of the dynamics surrounding the H10N3 subtype of avian influenza virus, utilizing advanced genomic surveillance techniques.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza , Virus de la Influenza A , Gripe Humana , Neuraminidasa , Filogenia , Humanos , China/epidemiología , Gripe Humana/virología , Neuraminidasa/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Mutación , Análisis Mutacional de ADN , Animales , Gripe Aviar/virología , Proteínas Virales/genética , Esputo/virología , Aves/virología , Masculino , ARN Viral/genética
5.
Virulence ; 15(1): 2359470, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38918890

RESUMEN

Influenza A virus (IAV) is the leading cause of highly contagious respiratory infections, which poses a serious threat to public health. The non-structural protein 1 (NS1) is encoded by segment 8 of IAV genome and is expressed in high levels in host cells upon IAV infection. It is the determinant of virulence and has multiple functions by targeting type Ι interferon (IFN-I) and type III interferon (IFN-III) production, disrupting cell apoptosis and autophagy in IAV-infected cells, and regulating the host fitness of influenza viruses. This review will summarize the current research on the NS1 including the structure and related biological functions of the NS1 as well as the interaction between the NS1 and host cells. It is hoped that this will provide some scientific basis for the prevention and control of the influenza virus.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Proteínas no Estructurales Virales , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Gripe Humana/virología , Animales , Autofagia , Virulencia , Interacciones Huésped-Patógeno , Apoptosis , Interferones/metabolismo , Interferones/inmunología , Interferones/genética
6.
Anal Methods ; 16(27): 4582-4589, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38919038

RESUMEN

As of now, the global COVID-19 pandemic caused by SARS-CoV-2, which began in 2019, has been effectively controlled. However, the symptoms of influenza A virus infection were similar to those of SARS-CoV-2 infection, but they required different treatment approaches. To make the detection more accurate and the treatment more targeted. We developed a system that integrates RPA and CRISPR assays, allowing for the rapid, highly specific, and sensitive detection and differentiation of SARS-CoV-2, H1N1, and H3N2. Under isothermal amplification conditions, the RPA-CRISPR Cas12a detection system achieved a detection limit as low as 5 copies per µL, demonstrating excellent specificity. The measurement time was approximately 30 minutes. The RPA-CRISPR Cas12a detection system combined with the microfluidic chip we designed to simultaneously detect three viruses, providing a potential solution for efficient and reliable diagnosis.


Asunto(s)
COVID-19 , Gripe Humana , SARS-CoV-2 , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Humanos , Gripe Humana/diagnóstico , COVID-19/diagnóstico , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Subtipo H1N1 del Virus de la Influenza A/genética , Técnicas de Amplificación de Ácido Nucleico/métodos , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Subtipo H3N2 del Virus de la Influenza A/genética , Sistemas CRISPR-Cas/genética , Dispositivos Laboratorio en un Chip , Técnicas Analíticas Microfluídicas/métodos , Técnicas Analíticas Microfluídicas/instrumentación , Límite de Detección , Técnicas de Diagnóstico Molecular/métodos , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/genética , Sensibilidad y Especificidad
7.
Cell Host Microbe ; 32(7): 1089-1102.e10, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38889725

RESUMEN

Avian influenza A virus (IAV) surveillance in Northern California, USA, revealed unique IAV hemagglutinin (HA) genome sequences in cloacal swabs from lesser scaups. We found two closely related HA sequences in the same duck species in 2010 and 2013. Phylogenetic analyses suggest that both sequences belong to the recently discovered H19 subtype, which thus far has remained uncharacterized. We demonstrate that H19 does not bind the canonical IAV receptor sialic acid (Sia). Instead, H19 binds to the major histocompatibility complex class II (MHC class II), which facilitates viral entry. Unlike the broad MHC class II specificity of H17 and H18 from bat IAV, H19 exhibits a species-specific MHC class II usage that suggests a limited host range and zoonotic potential. Using cell lines overexpressing MHC class II, we rescued recombinant H19 IAV. We solved the H19 crystal structure and identified residues within the putative Sia receptor binding site (RBS) that impede Sia-dependent entry.


Asunto(s)
Patos , Glicoproteínas Hemaglutininas del Virus de la Influenza , Antígenos de Histocompatibilidad Clase II , Virus de la Influenza A , Filogenia , Receptores Virales , Animales , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Receptores Virales/metabolismo , Receptores Virales/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Patos/virología , Humanos , Internalización del Virus , Gripe Aviar/virología , Sitios de Unión , Unión Proteica , Cristalografía por Rayos X , Línea Celular , Ácido N-Acetilneuramínico/metabolismo , Especificidad del Huésped , Especificidad de la Especie
8.
Viruses ; 16(6)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38932115

RESUMEN

In this study, we investigated the concentration of airborne influenza virus in daycare centers and influencing factors, such as common cold prevalence, air pollutants, and meteorological factors. A total of 209 air samples were collected from daycare centers in Kaohsiung and the influenza virus was analyzed using real-time quantitative polymerase chain reaction. Air pollutants and metrological factors were measured using real-time monitoring equipment. Winter had the highest positive rates of airborne influenza virus and the highest prevalence of the common cold, followed by summer and autumn. The concentration of CO was significantly positively correlated with airborne influenza virus. Daycare center A, with natural ventilation and air condition systems, had a higher concentration of airborne influenza A virus, airborne fungi, and airborne bacteria, as well as a higher prevalence of the common cold, than daycare center B, with a mechanical ventilation system and air purifiers, while the concentrations of CO2, CO, and UFPs in daycare center A were lower than those in daycare center B. We successfully detected airborne influenza virus in daycare centers, demonstrating that aerosol sampling for influenza can provide novel epidemiological insights and inform the management of influenza in daycare centers.


Asunto(s)
Microbiología del Aire , Guarderías Infantiles , Gripe Humana , Estaciones del Año , Humanos , Gripe Humana/epidemiología , Gripe Humana/virología , Gripe Humana/transmisión , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/genética , Orthomyxoviridae/aislamiento & purificación , Orthomyxoviridae/genética , Contaminantes Atmosféricos/análisis , Resfriado Común/epidemiología , Resfriado Común/virología , Resfriado Común/transmisión , Preescolar , Prevalencia , Monitoreo del Ambiente
9.
Viruses ; 16(6)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38932165

RESUMEN

Recently, high-throughput sequencing of influenza A viruses has become a routine test. It should be noted that the extremely high diversity of the influenza A virus complicates the task of determining the sequences of all eight genome segments. For a fast and accurate analysis, it is necessary to select the most suitable reference for each segment. At the same time, there is no standardized method in the field of decoding sequencing results that allows the user to update the sequence databases to which the reads obtained by virus sequencing are compared. The IAVCP (influenza A virus consensus and phylogeny) was developed with the goal of automatically analyzing high-throughput sequencing data of influenza A viruses. Its goals include the extraction of a consensus genome directly from paired raw reads. In addition, the pipeline enables the identification of potential reassortment events in the evolutionary history of the virus of interest by analyzing the topological structure of phylogenetic trees that are automatically reconstructed.


Asunto(s)
Genoma Viral , Secuenciación de Nucleótidos de Alto Rendimiento , Virus de la Influenza A , Filogenia , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Humanos , Genómica/métodos , Gripe Humana/virología , Biología Computacional/métodos
10.
Viruses ; 16(6)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38932174

RESUMEN

Influenza A viruses continue to be a serious health risk to people and result in a large-scale socio-economic loss. Avian influenza viruses typically do not replicate efficiently in mammals, but through the accumulation of mutations or genetic reassortment, they can overcome interspecies barriers, adapt to new hosts, and spread among them. Zoonotic influenza A viruses sporadically infect humans and exhibit limited human-to-human transmission. However, further adaptation of these viruses to humans may result in airborne transmissible viruses with pandemic potential. Therefore, we are beginning to understand genetic changes and mechanisms that may influence interspecific adaptation, cross-species transmission, and the pandemic potential of influenza A viruses. We also discuss the genetic and phenotypic traits associated with the airborne transmission of influenza A viruses in order to provide theoretical guidance for the surveillance of new strains with pandemic potential and the prevention of pandemics.


Asunto(s)
Adaptación al Huésped , Virus de la Influenza A , Gripe Humana , Humanos , Gripe Humana/transmisión , Gripe Humana/virología , Gripe Humana/epidemiología , Animales , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Gripe Aviar/transmisión , Gripe Aviar/virología , Aves/virología , Pandemias
11.
Viruses ; 16(6)2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38932181

RESUMEN

High pathogenicity avian influenza viruses (HPAIVs) cause high morbidity and mortality in poultry species. HPAIV prevalence means high numbers of infected wild birds could lead to spill over events for farmed poultry. How these pathogens survive in the environment is important for disease maintenance and potential dissemination. We evaluated the temperature-associated survival kinetics for five clade 2.3.4.4 H5Nx HPAIVs (UK field strains between 2014 and 2021) incubated at up to three temperatures for up to ten weeks. The selected temperatures represented northern European winter (4 °C) and summer (20 °C); and a southern European summer temperature (30 °C). For each clade 2.3.4.4 HPAIV, the time in days to reduce the viral infectivity by 90% at temperature T was established (DT), showing that a lower incubation temperature prolonged virus survival (stability), where DT ranged from days to weeks. The fastest loss of viral infectivity was observed at 30 °C. Extrapolation of the graphical DT plots to the x-axis intercept provided the corresponding time to extinction for viral decay. Statistical tests of the difference between the DT values and extinction times of each clade 2.3.4.4 strain at each temperature indicated that the majority displayed different survival kinetics from the other strains at 4 °C and 20 °C.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Temperatura , Animales , Gripe Aviar/virología , Gripe Aviar/mortalidad , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/fisiología , Cinética , Aves de Corral/virología , Animales Salvajes/virología , Aves/virología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/mortalidad
12.
Viruses ; 16(6)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38932250

RESUMEN

This study aimed to determine the incidence and etiological, seasonal, and genetic characteristics of respiratory viral coinfections involving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Between October 2020 and January 2024, nasopharyngeal samples were collected from 2277 SARS-CoV-2-positive patients. Two multiplex approaches were used to detect and sequence SARS-CoV-2, influenza A/B viruses, and other seasonal respiratory viruses: multiplex real-time polymerase chain reaction (PCR) and multiplex next-generation sequencing. Coinfections of SARS-CoV-2 with other respiratory viruses were detected in 164 (7.2%) patients. The most common co-infecting virus was respiratory syncytial virus (RSV) (38 cases, 1.7%), followed by bocavirus (BoV) (1.2%) and rhinovirus (RV) (1.1%). Patients ≤ 16 years of age had the highest rate (15%) of mixed infections. Whole-genome sequencing produced 19 complete genomes of seasonal respiratory viral co-pathogens, which were subjected to phylogenetic and amino acid analyses. The detected influenza viruses were classified into the genetic groups 6B.1A.5a.2a and 6B.1A.5a.2a.1 for A(H1N1)pdm09, 3C.2a1b.2a.2a.1 and 3C.2a.2b for A(H3N2), and V1A.3a.2 for the B/Victoria lineage. The RSV-B sequences belonged to the genetic group GB5.0.5a, with HAdV-C belonging to type 1, BoV to genotype VP1, and PIV3 to lineage 1a(i). Multiple amino acid substitutions were identified, including at the antibody-binding sites. This study provides insights into respiratory viral coinfections involving SARS-CoV-2 and reinforces the importance of genetic characterization of co-pathogens in the development of therapeutic and preventive strategies.


Asunto(s)
COVID-19 , Coinfección , Filogenia , SARS-CoV-2 , Humanos , Coinfección/virología , Coinfección/epidemiología , SARS-CoV-2/genética , SARS-CoV-2/clasificación , SARS-CoV-2/aislamiento & purificación , COVID-19/virología , COVID-19/epidemiología , Persona de Mediana Edad , Adulto , Femenino , Masculino , Adolescente , Preescolar , Niño , Anciano , Adulto Joven , Lactante , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/epidemiología , Rhinovirus/genética , Rhinovirus/clasificación , Rhinovirus/aislamiento & purificación , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Virus Sincitial Respiratorio Humano/genética , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Virus Sincitial Respiratorio Humano/clasificación , Nasofaringe/virología , Secuenciación Completa del Genoma , China/epidemiología , Estaciones del Año , Anciano de 80 o más Años , Genoma Viral , Virus de la Influenza B/genética , Virus de la Influenza B/aislamiento & purificación , Virus de la Influenza B/clasificación
13.
J Mol Diagn ; 26(7): 599-612, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38901927

RESUMEN

The high disease burden of influenza virus poses a significant threat to human health. Optimized diagnostic technologies that combine speed, sensitivity, and specificity with minimal equipment requirements are urgently needed to detect the many circulating species, subtypes, and variants of influenza at the point of need. Here, we introduce such a method using Streamlined Highlighting of Infections to Navigate Epidemics (SHINE), a clustered regularly interspaced short palindromic repeats (CRISPR)-based RNA detection platform. Four SHINE assays were designed and validated for the detection and differentiation of clinically relevant influenza species (A and B) and subtypes (H1N1 and H3N2). When tested on clinical samples, these optimized assays achieved 100% concordance with quantitative RT-PCR. Duplex Cas12a/Cas13a SHINE assays were also developed to detect two targets simultaneously. This study demonstrates the utility of this duplex assay in discriminating two alleles of an oseltamivir resistance (H275Y) mutation as well as in simultaneously detecting influenza A and human RNAse P in patient samples. These assays have the potential to expand influenza detection outside of clinical laboratories for enhanced influenza diagnosis and surveillance.


Asunto(s)
Sistemas CRISPR-Cas , Gripe Humana , Humanos , Gripe Humana/diagnóstico , Gripe Humana/virología , Sistemas CRISPR-Cas/genética , Sensibilidad y Especificidad , ARN Viral/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Técnicas de Diagnóstico Molecular/métodos , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/aislamiento & purificación , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/clasificación
14.
Emerg Microbes Infect ; 13(1): 2364732, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38832658

RESUMEN

Recently, an outbreak of highly pathogenic avian influenza A (H5N1), which carries the clade 2.3.4.4b hemagglutinin (HA) gene and has been prevalent among North American bird populations since the winter of 2021, was reported in dairy cows in the United States. As of 24 May 2024, the virus has affected 63 dairy herds across nine states and has resulted in two human infections. The virus causes unusual symptoms in dairy cows, including an unexpected drop in milk production, and thick colostrum-like milk. Notably, The US Food and Drug Administration reported that around 20% of tested retail milk samples contained H5N1 viruses, with a higher percentage of positive results from regions with infected cattle herds. Data are scant regarding how effectively pasteurization inactivates the H5N1 virus in milk. Therefore, in this study, we evaluated the thermal stability of the H5 clade 2.3.4.4b viruses, along with one human H3N2 virus and other influenza subtype viruses, including H1, H3, H7, H9, and H10 subtype viruses. We also assessed the effectiveness of pasteurization in inactivating these viruses. We found that the avian H3 virus exhibits the highest thermal stability, whereas the H5N1 viruses that belong to clade 2.3.4.4b display moderate thermal stability. Importantly, our data provide direct evidence that the standard pasteurization methods used by dairy companies are effective in inactivating all tested subtypes of influenza viruses in raw milk. Our findings indicate that thermally pasteurized milk products do not pose a safety risk to consumers.


Asunto(s)
Leche , Pasteurización , Animales , Pasteurización/métodos , Leche/virología , Bovinos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/aislamiento & purificación , Humanos , Gripe Aviar/virología , Gripe Aviar/transmisión , Gripe Aviar/prevención & control , Gripe Aviar/epidemiología , Inactivación de Virus , Estados Unidos , Gripe Humana/virología , Gripe Humana/transmisión , Gripe Humana/prevención & control , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Femenino
15.
Respir Res ; 25(1): 234, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38840154

RESUMEN

BACKGROUND: The concurrent circulation of SARS-CoV-2 with other respiratory viruses is unstoppable and represents a new diagnostic reality for clinicians and clinical microbiology laboratories. Multiplexed molecular testing on automated platforms that focus on the simultaneous detection of multiple respiratory viruses in a single tube is a useful approach for current and future diagnosis of respiratory infections in the clinical setting. METHODS: Two time periods were included in the study: from February to April 2022, an early 2022 period, during the gradual lifting of COVID-19 prevention measures in the country, and from October 2022 to April 2023, the 2022/23 respiratory infections season. We analysed a total of 1,918 samples in the first period and 18,131 respiratory samples in the second period using a multiplex molecular assay for the simultaneous detection of Influenza A (Flu-A), Influenza B (Flu-B), Human Respiratory Syncytial Virus (HRSV) and SARS-CoV-2. RESULTS: The results from early 2022 showed a strong dominance of SARS-CoV-2 infections with 1,267/1,918 (66.1%) cases. Flu-A was detected in 30/1,918 (1.6%) samples, HRSV in 14/1,918 (0.7%) samples, and Flu-B in 2/1,918 (0.1%) samples. Flu-A/SARS-CoV-2 co-detections were observed in 11/1,267 (0.9%) samples, and HRSV/SARS-CoV-2 co-detection in 5/1,267 (0.4%) samples. During the 2022/23 winter respiratory season, SARS-CoV-2 was detected in 1,738/18,131 (9.6%), Flu-A in 628/18,131 (3.5%), Flu-B in 106/18,131 (0.6%), and HRSV in 505/18,131 (2.8%) samples. Interestingly, co-detections were present to a similar extent as in early 2022. CONCLUSION: The results show that the multiplex molecular approach is a valuable tool for the simultaneous laboratory diagnosis of SARS-CoV-2, Flu-A/B, and HRSV in hospitalized and outpatients. Infections with Flu-A/B, and HRSV occurred shortly after the COVID-19 control measures were lifted, so a strong reoccurrence of various respiratory infections and co-detections in the post COVID-19 period was to be expected.


Asunto(s)
COVID-19 , Virus de la Influenza A , Virus de la Influenza B , Gripe Humana , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , SARS-CoV-2 , Humanos , COVID-19/epidemiología , COVID-19/diagnóstico , Virus de la Influenza B/aislamiento & purificación , Virus de la Influenza B/genética , Gripe Humana/epidemiología , Gripe Humana/diagnóstico , Gripe Humana/virología , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Virus Sincitial Respiratorio Humano/aislamiento & purificación , Virus Sincitial Respiratorio Humano/genética , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/genética , Masculino , Femenino , Coinfección/epidemiología , Coinfección/diagnóstico , Persona de Mediana Edad , Adulto , Técnicas de Diagnóstico Molecular/métodos , Estaciones del Año , Anciano
16.
Rev Med Virol ; 34(4): e2559, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38886173

RESUMEN

The World Organization for Animal Health defines Avian Influenza Virus as a highly infectious disease caused by diverse subtypes that continue to evolve rapidly, impacting poultry species, pet birds, wild birds, non-human mammals, and occasionally humans. The effects of Avian influenza viruses have been recognised as a precursor for serious health concerns among affected birds, poultry, and human populations in the Middle East. Furthermore, low and high pathogenic avian influenza viruses lead to respiratory illness with varying severity, depending on the virus subtype (e.g., H5, H7, H9, etc.). Possible future outbreaks and endemics of newly emerging subtypes are expected to occur, as many studies have reported the emergence of novel mutations and viral subtypes. However, proper surveillance programs and biosecurity applications should be developed, and countries with incapacitated defences against such outbreaks should be encouraged to undergo complete reinstation and reinforcement in their health and research sectors. Public education regarding biosafety and virus prevention is necessary to ensure minimal spread of avian influenza endemic.


Asunto(s)
Aves , Virus de la Influenza A , Gripe Aviar , Gripe Humana , Animales , Gripe Aviar/epidemiología , Gripe Aviar/virología , Gripe Aviar/prevención & control , Gripe Aviar/transmisión , Humanos , Gripe Humana/prevención & control , Gripe Humana/epidemiología , Gripe Humana/virología , Región Mediterránea/epidemiología , Aves/virología , Virus de la Influenza A/genética , Virus de la Influenza A/fisiología , Virus de la Influenza A/patogenicidad , Brotes de Enfermedades/prevención & control , Brotes de Enfermedades/veterinaria
17.
PLoS One ; 19(6): e0303756, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38829903

RESUMEN

The rapid spread of highly pathogenic avian influenza (HPAI) A (H5N1) viruses in Southeast Asia in 2004 prompted the New Zealand Ministry for Primary Industries to expand its avian influenza surveillance in wild birds. A total of 18,693 birds were sampled between 2004 and 2020, including migratory shorebirds (in 2004-2009), other coastal species (in 2009-2010), and resident waterfowl (in 2004-2020). No avian influenza viruses (AIVs) were isolated from cloacal or oropharyngeal samples from migratory shorebirds or resident coastal species. Two samples from red knots (Calidris canutus) tested positive by influenza A RT-qPCR, but virus could not be isolated and no further characterization could be undertaken. In contrast, 6179 samples from 15,740 mallards (Anas platyrhynchos) tested positive by influenza A RT-qPCR. Of these, 344 were positive for H5 and 51 for H7. All H5 and H7 viruses detected were of low pathogenicity confirmed by a lack of multiple basic amino acids at the hemagglutinin (HA) cleavage site. Twenty H5 viruses (six different neuraminidase [NA] subtypes) and 10 H7 viruses (two different NA subtypes) were propagated and characterized genetically. From H5- or H7-negative samples that tested positive by influenza A RT-qPCR, 326 AIVs were isolated, representing 41 HA/NA combinations. The most frequently isolated subtypes were H4N6, H3N8, H3N2, and H10N3. Multivariable logistic regression analysis of the relations between the location and year of sampling, and presence of AIV in individual waterfowl showed that the AIV risk at a given location varied from year to year. The H5 and H7 isolates both formed monophyletic HA groups. The H5 viruses were most closely related to North American lineages, whereas the H7 viruses formed a sister cluster relationship with wild bird viruses of the Eurasian and Australian lineages. Bayesian analysis indicates that the H5 and H7 viruses have circulated in resident mallards in New Zealand for some time. Correspondingly, we found limited evidence of influenza viruses in the major migratory bird populations visiting New Zealand. Findings suggest a low probability of introduction of HPAI viruses via long-distance bird migration and a unique epidemiology of AIV in New Zealand.


Asunto(s)
Animales Salvajes , Aves , Gripe Aviar , Filogenia , Animales , Nueva Zelanda/epidemiología , Gripe Aviar/virología , Gripe Aviar/epidemiología , Animales Salvajes/virología , Aves/virología , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/clasificación , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Genoma Viral , Patos/virología
19.
Emerg Infect Dis ; 30(6): 1285-1288, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38703022

RESUMEN

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.


Asunto(s)
Animales Salvajes , Aves , Virus de la Influenza A , Gripe Aviar , Filogenia , Animales , Gripe Aviar/virología , Gripe Aviar/epidemiología , República de Corea/epidemiología , Animales Salvajes/virología , Virus de la Influenza A/genética , Virus de la Influenza A/clasificación , Aves/virología , Virus Reordenados/genética , Historia del Siglo XXI , Humanos , Neuraminidasa/genética
20.
Protein Expr Purif ; 221: 106506, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38772430

RESUMEN

Influenza poses a substantial health risk, with infants and the elderly being particularly susceptible to its grave impacts. The primary challenge lies in its rapid genetic evolution, leading to the emergence of new Influenza A strains annually. These changes involve punctual mutations predominantly affecting the two main glycoproteins: Hemagglutinin (HA) and Neuraminidase (NA). Our existing vaccines target these proteins, providing short-term protection, but fall short when unexpected pandemics strike. Delving deeper into Influenza's genetic makeup, we spotlight the nucleoprotein (NP) - a key player in the transcription, replication, and packaging of RNA. An intriguing characteristic of the NP is that it is highly conserved across all Influenza A variants, potentially paving the way for a more versatile and broadly protective vaccine. We designed and synthesized a novel NP-Hoc fusion protein combining Influenza A nucleoprotein and T4 phage Hoc, cloned using Gibson assembly in E. coli, and purified via ion affinity chromatography. Simultaneously, we explore the T4 coat protein Hoc, typically regarded as inconsequential in controlled viral replication. Yet, it possesses a unique ability: it can link with another protein, showcasing it on the T4 phage coat. Fusing these concepts, our study designs, expresses, and purifies a novel fusion protein named NP-Hoc. We propose this protein as the basis for a new generation of vaccines, engineered to guard broadly against Influenza A. The excitement lies not just in the immediate application, but the promise this holds for future pandemic resilience, with NP-Hoc marking a significant leap in adaptive, broad-spectrum influenza prevention.


Asunto(s)
Bacteriófago T4 , Escherichia coli , Proteínas Recombinantes de Fusión , Bacteriófago T4/genética , Bacteriófago T4/química , Bacteriófago T4/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/metabolismo , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/biosíntesis , Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/química , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...