Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 525
Filtrar
1.
Avian Dis ; 68(3): 225-230, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39400217

RESUMEN

Avian influenza virus (AIV) causes frequent outbreaks in poultry with high morbidity and mortality. The virus can survive on different fomites, resulting in indirect transmission to susceptible hosts. We investigated the inactivation by ozonated water (O3W) of three different subtypes of AIV (H4N8, H4N6, and H9N9) on seven different fomites. All subtypes were sensitive on all fomites, but there was a slight variation in the sensitivity of different subtypes. For example, AIV H9N9 showed more than 99% reduction on denim fabric, polypropylene, and Styrofoam after 3 min of exposure. More than 97% of H4N8 was eliminated from cardboard, denim fabric, and stainless steel after 3 min of exposure. Subtype H4N6 was the least sensitive; highest inactivation (98%) was seen on cardboard and polypropylene after 3 min of exposure. In conclusion, O3W can inactivate a large percentage of AIV applied to fomites within 3 min in all tested subtypes. Interestingly, an increase in contact time to 10 min did not result in an increase in the virus inactivation rate, probably because of the low half-life of ozone. Further studies are needed to determine how the residual virus can be inactivated so that it does not pose a problem to naïve birds.


Inactivación comparativa de tres subtipos diferentes del virus de la influenza aviar mediante agua ozonizada. El virus de la influenza aviar causa frecuentes brotes en aves la avicultura con alta morbilidad y mortalidad. El virus puede sobrevivir en diferentes fómites, lo que resulta en transmisión indirecta a huéspedes susceptibles. Se investigó la inactivación mediante agua ozonizada de tres subtipos diferentes del virus de la influenza aviar (H4N8, H4N6 y H9N9) en siete fómites diferentes. Todos los subtipos fueron sensibles al agua ozonizada (O3W) en todos los fómites, pero hubo una ligera variación en la sensibilidad de los diferentes subtipos. Por ejemplo, el virus subtipo H9N9 mostró una reducción de más del 99 % en tela de mezclilla, polipropileno y espuma de poliestireno después de tres minutos de exposición. Más del 97% del subtipo H4N8 se eliminó del cartón, la mezclilla y el acero inoxidable después de tres minutos de exposición. El subtipo H4N6 fue el menos sensible; La inactivación más alta (98%) se observó en cartón y polipropileno después de tres minutos de exposición. En conclusión, el agua ozonizada puede inactivar un gran porcentaje del virus de influenza aviar aplicado a fómites en tres minutos en todos los subtipos estudiados. Curiosamente, un aumento en el tiempo de contacto a 10 minutos no resultó en un aumento en la tasa de inactivación del virus, probablemente debido a la baja vida media del ozono. Se necesitan más estudios para determinar cómo se puede inactivar el virus residual para que no represente un problema para las aves susceptibles.


Asunto(s)
Virus de la Influenza A , Ozono , Agua , Ozono/farmacología , Animales , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/fisiología , Agua/química , Gripe Aviar/virología , Inactivación de Virus/efectos de los fármacos , Fómites/virología
2.
BMC Infect Dis ; 24(1): 918, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39232670

RESUMEN

BACKGROUND: On March 16th 2024, the first case of Human infection with avian influenza H10N3 since the end of the global COVID-19 Pandemic was reported in Kunming, China. To enhance comprehension of the source of infection and risk factors of the H10N3 virus infection, this case report summarizes the clinical features, epidemiological investigation, and laboratory test results. Provides recommendations for the prevention and control of Human infection with avian influenza H10N3. CASE PRESENTATION: A 51-year-old male with a history of COVID-19 infection and a smoking habit of 30 years, worked in livestock breeding and was exposed to sick and dead poultry before falling ill with fever and chills on 28th February 2024. A week later, he was diagnosed with severe pneumonia, influenza, and respiratory failure by the Third People's Hospital of Kunming(KM-TPH). He was discharged on 17th April and none of his 6 close contacts showed any symptoms of illness. Environmental samples taken from the epidemic spot revealed that peacock feces tested positive for avian influenza sub-type H9 and waterfowl specimens showed positive results for avian influenza sub-type H5. Gene sequencing conducted on positive specimens from the patient's respiratory tract by the Chinese Centre for Disease Control and Prevention (CCDC) showed a high degree of similarity (98.6-99.5%) with the strain responsible for the second global case of human infected with H10N3 (reported from Zhejiang, China 2022). CONCLUSIONS: According to the available epidemiological information, there is limited evidence to suggest that H10N3 viruses are excessively lethal. However, adaptive site mutations have been observed in the H10N3 isoform of mammals. While it is unlikely that the H10N3 virus will spread among humans, the possibility of additional cases cannot be entirely ruled out. Symptoms of human infection with H10N3 avian influenza are similar to those of common respiratory infections, which may result in them being overlooked during initial clinical consultations. Therefore, it is essential to improve surveillance of the H10 sub-type of avian influenza and to increase the awareness of hospital-related workers of cases of pneumonia of unknown origin.


Asunto(s)
COVID-19 , Gripe Aviar , Gripe Humana , Humanos , Masculino , Persona de Mediana Edad , Gripe Humana/virología , Animales , Gripe Aviar/virología , COVID-19/epidemiología , China/epidemiología , Aves de Corral/virología , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , SARS-CoV-2/genética , Filogenia
3.
Front Immunol ; 15: 1374838, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39281683

RESUMEN

Dendritic cells (DCs) are professional antigen-presenting cells, which are key components of the immune system and involved in early immune responses. DCs are specialized in capturing, processing, and presenting antigens to facilitate immune interactions. Chickens infected with avian influenza virus (AIV) demonstrate a wide range of clinical symptoms, based on pathogenicity of the virus. Low pathogenic avian influenza (LPAI) viruses typically induce mild clinical signs, whereas high pathogenic avian influenza (HPAI) induce more severe disease, which can lead to death. For this study, chicken bone marrow-derived DC (ckBM-DC)s were produced and infected with high and low pathogenic avian influenza viruses of H5N2 or H7N3 subtypes to characterize innate immune responses, study effect on cell morphologies, and evaluate virus replication. A strong proinflammatory response was observed at 8 hours post infection, via upregulation of chicken interleukin-1ß and stimulation of the interferon response pathway. Microscopically, the DCs underwent morphological changes from classic elongated dendrites to a more general rounded shape that eventually led to cell death with the presence of scattered cellular debris. Differences in onset of morphologic changes were observed between H5 and H7 subtypes. Increases in viral titers demonstrated that both HPAI and LPAI are capable of infecting and replicating in DCs. The increase in activation of infected DCs may be indicative of a dysregulated immune response typically seen with HPAI infections.


Asunto(s)
Pollos , Citocinas , Células Dendríticas , Gripe Aviar , Animales , Células Dendríticas/inmunología , Células Dendríticas/virología , Pollos/virología , Gripe Aviar/inmunología , Gripe Aviar/virología , Gripe Aviar/patología , Citocinas/metabolismo , Citocinas/inmunología , Virus de la Influenza A/inmunología , Replicación Viral , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/virología
4.
Poult Sci ; 103(10): 104135, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39106695

RESUMEN

During poultry immunization, antibiotics are typically added to inactivated oil-adjuvant avian influenza (AI) vaccines. Here, we evaluated the effects of adding ceftiofur, a third-generation cephalosporin, to an AI vaccine on vaccine stability and structure and on chick growth, immune efficacy, blood concentrations, biochemical and immunological indices, and gut microbiota. The results demonstrated that neither aqueous ceftiofur sodium nor ceftiofur hydrochloride oil emulsion formed a stable mixture with the vaccine. Adding ceftiofur formulations, particularly ceftiofur hydrochloride, at >4% significantly destabilized the vaccine's water-in-oil structures. Adding ceftiofur also increased vaccine malabsorption at the injection site; specifically, adding ceftiofur hydrochloride reduced H5N8 and H7N9 antibody titers after the first immunization (P < 0.05) and H7N9 antibody titers after the second immunization (P < 0.01). Serum drug concentrations did not differ significantly between the groups with ceftiofur sodium and hydrochloride addition. Ceftiofur addition increased postvaccination chick weight loss; compared with the vaccine alone, ceftiofur sodium-vaccine mixture increased chick weight significantly (P < 0.05). Ceftiofur addition also increased stress indices and reduced antioxidant capacity significantly (P < 0.05 or P < 0.01). Vaccination-related immune stress reduced gut microbiota diversity in chicks; ceftiofur addition reversed this change. AI vaccine immunization significantly reduced the relative abundance of Lactobacillus and Muribaculaceae but significantly increased that of Bacteroides and Eubacterium coprostanoligenes group. Ceftiofur addition restored the gut microbiota structure; in particular, ceftiofur hydrochloride addition significantly increased the abundance of the harmful gut microbes Escherichia-Shigella and Enterococcus, whereas ceftiofur sodium addition significantly reduced it. The changes in gut microbiota led to alterations in metabolic pathways related to membrane transport, amino acids, and carbohydrates. In conclusion, adding ceftiofur to the AI vaccine had positive effects on chick growth and gut microbiota modulation; however, different antibiotic concentrations and formulations may disrupt vaccine structure, possibly affecting vaccine safety and immunization efficacy. Thus, the addition of antibiotics to oil-adjuvant vaccines is associated with a risk of immunization failure and should be applied to poultry with caution.


Asunto(s)
Antibacterianos , Cefalosporinas , Pollos , Vacunas contra la Influenza , Gripe Aviar , Animales , Cefalosporinas/administración & dosificación , Cefalosporinas/farmacología , Gripe Aviar/prevención & control , Antibacterianos/farmacología , Antibacterianos/administración & dosificación , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunas de Productos Inactivados/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Adyuvantes Inmunológicos/farmacología , Adyuvantes Inmunológicos/administración & dosificación
5.
Poult Sci ; 103(10): 104125, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39137496

RESUMEN

After viral infection, the virus relies on the host cell's complex metabolic and biosynthetic machinery for replication. However, the impact of avian influenza virus (AIV) on metabolites and gene expression in poultry cells remains unclear. To investigate this, we infected chicken embryo fibroblasts DF1 cells with H9N2 AIV at an MOI of 3. Our aim was to explore how H9N2 AIV alters DF1 cells metabolic pathways to facilitate its replication. We employed metabolomics and transcriptomics techniques to analyze changes in metabolite content and gene expression. Metabolomics analysis revealed a significant increase in glutathione-related metabolites, including reduced glutathione (GSH), oxidized glutathione (GSSG) and total glutathione (T-GSH) upon H9N2 AIV infection in DF1 cells. Elisa results confirmed elevated levels of GSH, GSSG, and T-GSH consistent with metabolomics findings, noting a pronounced increase in GSSG compared to GSH. Transcriptomics showed significant alterations in genes involved in glutathione synthesis and metabolism post-H9N2 infection. However, adding the glutathione synthesis inhibitor BSO exogenously significantly promoted H9N2 replication in DF1 cells. This was accompanied by increased mRNA levels of pro-inflammatory cytokines (IL-1ß, IFN-γ) and decreased mRNA levels of anti-inflammatory cytokines (TGF-ß, IL-13). BSO also reduced catalase (CAT) gene expression and inhibited its activity, leading to higher reactive oxygen species (ROS) and malondialdehyde (MDA) level in DF1 cells. qPCR results indicated decreased mRNA levels of Nrf2, NQO1, and HO-1 with BSO, ultimately increasing oxidative stress in DF1 cells. Therefore, the above results indicated that H9N2 AIV infection in DF1 cells activated the glutathione metabolic pathway to enhance the cell's self-defense mechanism against H9N2 replication. However, when GSH synthesis is inhibited within the cells, it leads to an elevated oxidative stress level, thereby promoting H9N2 replication within the cells through Nrf2/HO-1 pathway. This study provides a theoretical basis for future rational utilization of the glutathione metabolic pathway to prevent viral replication.


Asunto(s)
Pollos , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Animales , Subtipo H9N2 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Embrión de Pollo , Línea Celular , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/genética , Replicación Viral/efectos de los fármacos , Glutatión/metabolismo , Fibroblastos/virología , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos
6.
J Food Prot ; 87(8): 100325, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38964610

RESUMEN

With the emergence of clade 2.3.4.4b H5N1 highly pathogenic avian influenza virus (AIV) infection of dairy cattle and its subsequent detection in raw milk, coupled with recent AIV infections affecting dairy farm workers, experiments were conducted to affirm the safety of cooked ground beef related to AIV because such meat is often derived from cull dairy cows. Specifically, retail ground beef (percent lean:fat = ca. 80:20) was inoculated with a low pathogenic AIV (LPAIV) isolate to an initial level of 5.6 log10 50% egg infectious doses (EID50)  per 300 g patty. The inoculated meat was pressed into patties (ca. 2.54 cm thick, ca. 300 g each) and then held at 4 °C for up to 60 min. In each of the two trials, two patties for each of the following three treatments were cooked on a commercial open-flame gas grill to internal instantaneous temperatures of 48.9 °C (120°F), 62.8 °C (145°F), or 71.1 °C (160°F), but without any dwell time. Cooking inoculated ground beef patties to 48.9 °C (ave. cooking time of ca. 15 min) resulted in a mean reduction of ≥2.5 ± 0.9 log10 EID50 per 300 g of ground beef as assessed via quantification of virus in embryonating chicken eggs (ECEs). Likewise, cooking patties on a gas grill to 62.8 °C (ave. cooking time of ca. 21 min) or to the USDA FSIS recommended minimum internal temperature for ground beef of 71.1 °C (ave. cooking time of ca. 24 min) resulted in a reduction to nondetectable levels from initial levels of ≥5.6 log10 EID50 per 300 g. These data establish that levels of infectious AIV are substantially reduced within inoculated ground beef patties (20% fat) using recommended cooking procedures.


Asunto(s)
Culinaria , Animales , Bovinos , Humanos , Gripe Aviar , Carne Roja , Subtipo H5N1 del Virus de la Influenza A , Carne , Aves
7.
Poult Sci ; 103(8): 103885, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38851182

RESUMEN

Avian influenza, particularly the H9N2 subtype, presents significant challenges to poultry health, underscoring the need for effective antiviral interventions. This study explores the antiviral capabilities of Belamcanda extract, a traditional Chinese medicinal herb, against H9N2 Avian influenza virus (AIV) in specific pathogen-free (SPF) chicks. Through a comprehensive approach, we evaluated the impact of the extract on cytokine modulation and crucial immunological signaling pathways, essential for understanding the host-virus interaction. Our findings demonstrate that Belamcanda extract significantly modulates the expression of key inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interleukin-1 (IL-1), interleukin-2 (IL-2), and interleukin-6 (IL-6), which are pivotal to the host's response to H9N2 AIV infection. Western blot analysis further revealed that the extract markedly reduces the expression of critical immune signaling molecules such as toll-like receptor 3 (TLR3), TIR-domain-containing adapter-inducing interferon-ß (TRIF), and nuclear factor kappa B (NF-κB). These insights into the mechanisms by which Belamcanda extract influences host immune responses and hinders viral replication highlight its potential as an innovative antiviral agent for poultry health management. The study advances our comprehension of natural compounds' antiviral mechanisms and lays the groundwork for developing strategies to manage viral infections in poultry. The demonstrated ability of Belamcanda extract to modulate immune responses and inhibit viral replication establishes it as a promising candidate for future antiviral therapy development, especially in light of the need for effective treatments against evolving influenza virus strains and the critical demand for enhanced poultry health management strategies.


Asunto(s)
Antivirales , Pollos , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Enfermedades de las Aves de Corral , Animales , Subtipo H9N2 del Virus de la Influenza A/fisiología , Subtipo H9N2 del Virus de la Influenza A/efectos de los fármacos , Gripe Aviar/virología , Gripe Aviar/tratamiento farmacológico , Antivirales/farmacología , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/tratamiento farmacológico , Enfermedades de las Aves de Corral/inmunología , Organismos Libres de Patógenos Específicos , Inflamación/tratamiento farmacológico , Inflamación/veterinaria , Inflamación/virología , Citocinas/metabolismo , Citocinas/genética , Extractos Vegetales/farmacología
8.
J Immunol ; 213(2): 187-203, 2024 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-38829131

RESUMEN

The RING finger (RNF) family, a group of E3 ubiquitin ligases, plays multiple essential roles in the regulation of innate immunity and resistance to viral infection in mammals. However, it is still unclear whether RNF proteins affect the production of IFN-I and the replication of avian influenza virus (AIV) in ducks. In this article, we found that duck RNF216 (duRNF216) inhibited the duRIG-I signaling pathway. Conversely, duRNF216 deficiency enhanced innate immune responses in duck embryonic fibroblasts. duRNF216 did not interacted with duRIG-I, duMDA5, duMAVS, duSTING, duTBK1, or duIRF7 in the duck RIG-I pathway. However, duRNF216 targeted duTRAF3 and inhibited duMAVS in the recruitment of duTRAF3 in a dose-dependent manner. duRNF216 catalyzed K48-linked polyubiquitination of duck TRAF3, which was degraded by the proteasome pathway. Additionally, AIV PB1 protein competed with duTRAF3 for binding to duRNF216 to reduce degradation of TRAF3 by proteasomes in the cytoplasm, thereby slightly weakening duRNF216-mediated downregulation of IFN-I. Moreover, although duRNF216 downregulated the IFN-ß expression during virus infection, the expression level of IFN-ß in AIV-infected duck embryonic fibroblasts overexpressing duRNF216 was still higher than that in uninfected cells, which would hinder the viral replication. During AIV infection, duRNF216 protein targeted the core protein PB1 of viral polymerase to hinder viral polymerase activity and viral RNA synthesis in the nucleus, ultimately strongly restricting viral replication. Thus, our study reveals a new mechanism by which duRNF216 downregulates innate immunity and inhibits AIV replication in ducks. These findings broaden our understanding of the mechanisms by which the duRNF216 protein affects AIV replication in ducks.


Asunto(s)
Patos , Inmunidad Innata , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar , Transducción de Señal , Ubiquitina-Proteína Ligasas , Replicación Viral , Animales , Patos/inmunología , Patos/virología , Replicación Viral/inmunología , Transducción de Señal/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/fisiología , Inmunidad Innata/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/inmunología , Fibroblastos/inmunología , Fibroblastos/virología , Proteínas Aviares/inmunología , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Ubiquitinación , Proteína 58 DEAD Box/metabolismo , Proteína 58 DEAD Box/inmunología
9.
Virology ; 595: 110094, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38692133

RESUMEN

Stress-induced immunosuppression (SIIS) is one of common problems in the intensive poultry industry, affecting the effect of vaccine immunization and leading to high incidences of diseases. In this study, the expression characteristics and regulatory mechanisms of miR-214 in the processes of SIIS and its influence on the immune response to avian influenza virus (AIV) vaccine in chicken were explored. The qRT-PCR results showed that serum circulating miR-214 was significantly differentially expressed (especially on 2, 5, and 28 days post immunization (dpi)) in the processes, so had the potential as a molecular marker. MiR-214 expressions from multiple tissues were closely associated with the changes in circulating miR-214 expression levels. MiR-214-PTEN regulatory network was a potential key regulatory mechanism for the heart, bursa of Fabricius, and glandular stomach to participate in the process of SIIS affecting AIV immune response. This study can provide references for further understanding of stress affecting immune response.


Asunto(s)
Pollos , Vacunas contra la Influenza , Gripe Aviar , MicroARNs , Fosfohidrolasa PTEN , Estrés Fisiológico , Animales , MicroARNs/genética , MicroARNs/metabolismo , Pollos/virología , Vacunas contra la Influenza/inmunología , Gripe Aviar/virología , Gripe Aviar/inmunología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/inmunología , Tolerancia Inmunológica , Transducción de Señal , Virus de la Influenza A/inmunología
10.
Int J Mol Sci ; 25(5)2024 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-38473897

RESUMEN

The H9N2 avian influenza virus causes reduced production performance and immunosuppression in chickens. The chicken yolk sac immunoglobulins (IgY) receptor (FcRY) transports from the yolk into the embryo, providing offspring with passive immunity to infection against common poultry pathogens. FcRY is expressed in many tissues/organs of the chicken; however, there are no reports investigating FcRY expression in chicken macrophage cells, and how H9N2-infected HD11 cells (a chicken macrophage-like cell line) regulate FcRY expression remains uninvestigated. This study used the H9N2 virus as a model pathogen to explore the regulation of FcRY expression in avian macrophages. FcRY was highly expressed in HD11 cells, as shown by reverse transcription polymerase chain reactions, and indirect immunofluorescence indicated that FcRY was widely expressed in HD11 cells. HD11 cells infected with live H9N2 virus exhibited downregulated FcRY expression. Transfection of eukaryotic expression plasmids encoding each viral protein of H9N2 into HD11 cells revealed that nonstructural protein (NS1) and matrix protein (M1) downregulated FcRY expression. In addition, the use of a c-jun N-terminal kinase (JNK) activator inhibited the expression of FcRY, while a JNK inhibitor antagonized the downregulation of FcRY expression by live H9N2 virus, NS1 and M1 proteins. Finally, a dual luciferase reporter system showed that both the M1 protein and the transcription factor c-jun inhibited FcRY expression at the transcriptional level. Taken together, the transcription factor c-jun was a negative regulator of FcRY, while the live H9N2 virus, NS1, and M1 proteins downregulated the FcRY expression through activating the JNK signaling pathway. This provides an experimental basis for a novel mechanism of immunosuppression in the H9N2 avian influenza virus.


Asunto(s)
Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Animales , Pollos/metabolismo , Subtipo H9N2 del Virus de la Influenza A/fisiología , Sistema de Señalización de MAP Quinasas , Línea Celular , Macrófagos/metabolismo , Factores de Transcripción/metabolismo
11.
Poult Sci ; 103(4): 103496, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38330890

RESUMEN

The avian influenza virus is infected through the mucosal route, thus mucosal barrier defense is very important. While the inactivated H9N2 vaccine cannot achieve sufficient mucosal immunity, adjuvants are needed to induce mucosal and systemic immunity to prevent poultry from H9N2 influenza virus infection. Our previous study found that polysaccharide from Atractylodes macrocephala Koidz binding with zinc oxide nanoparticles (AMP-ZnONPs) had immune-enhancing effects in vitro. This study aimed to evaluate the mucosal immune responses of oral whole-inactivated H9N2 virus (WIV)+AMP-ZnONPs and its impact on the animal challenge protection, and the corresponding changes of pulmonary metabolomics after the second immunization. The results showed that compared to the WIV, the combined treatment of WIV and AMP-ZnONPs significantly enhanced the HI titer, IgG and specific sIgA levels, the number of goblet cells and intestinal epithelial lymphocytes (iIELs) as well as the expression of J-chain, polymeric immunoglobulin receptor (pIgR), interleukin-10 (IL-10), tumor necrosis factor-α (TNF-α) and transforming growth factor-ß (TGF-ß). In viral attack experiments, WIV combing with AMP-ZnONPs effectively reduced lung damage and viral titers in throat swabs. Interestingly, significant changes of both the IgA intestinal immune network and PPAR pathway could also be found in the WIV+AMP-ZnONPs group compared to the non-infected group. Taken together, these findings suggest that AMP-ZnONPs can serve as a potential mucosal vaccine adjuvant, thereby avoiding adverse stress and corresponding costs caused by vaccine injection.


Asunto(s)
Subtipo H9N2 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Aviar , Vacunas , Animales , Inmunidad Mucosa , Pollos , Anticuerpos Antivirales , Adyuvantes Inmunológicos/farmacología , Administración Oral , Vacunas de Productos Inactivados , Gripe Aviar/prevención & control
12.
Front Biosci (Landmark Ed) ; 29(1): 11, 2024 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-38287809

RESUMEN

BACKGROUND: Highly pathogenic H5Nx viruses cause avian influenza, a zoonotic disease that can infect humans. The vaccine can facilitate the prevention of human infections from infected poultry. Our previous study showed that an H5 cleavage-site peptide vaccine containing the polybasic amino acid RRRK could protect chickens from lethal infections of the highly pathogenic H5N6 avian influenza virus. METHODS: Chickens immunized with the various polybasic amino combinations (RRRK, RRR, RR, R, RK, and K) of H5 cleavage-site peptides were challenged with highly pathogenic H5N6 avian influenza viruses. The challenged chickens were monitored for survival rate, and viral titers in swabs and tissue samples were measured in Madin-Darby canine kidney (MDCK) cells using the median tissue culture infectious dose 50 (log10 TCID50/mL). RESULTS: Most H5 cleavage-site vaccines containing various combinations of polybasic amino acids protected chickens from lethal infection. Chickens immunized with the RK-containing peptide combination of the H5 cleavage site were not protected. CONCLUSIONS: The polybasic amino acids (RRRK) of H5 cleavage cleavage-site peptide vaccines are important for protecting chickens against HP H5N6 avian influenza virus. The H5 cleavage cleavage-site peptide containing RK did not protect chickens against the virus.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Animales , Perros , Humanos , Pollos/metabolismo , Gripe Aviar/prevención & control , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Aminoácidos/metabolismo , Péptidos
13.
Microbes Infect ; 26(1-2): 105231, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37777054

RESUMEN

Broad-spectrum antiviral activities of interferon-induced transmembrane proteins (IFITMs) are primarily attributed to in vitro inhibition of viral entry. Here, we used an avian sarcoma-leukosis virus (RCAS)-based gene transfer system and successfully generated chicks that constitutively express chicken IFITM3 (chIFITM3). The chIFITM3-overexpressing chicks showed significant protection and disease tolerance against highly pathogenic avian influenza virus (HPAIV) H5N1 (Clade 2.2.1.2). The chicks, overexpressing chIFITM3, also showed delayed onset of clinical symptoms, reduced viral shedding, and alleviated histopathologic alterations compared to control and challenged chicks. These findings highlight that overexpression of chIFITM3 provide a substantial defense against zoonotic H5N1 in vivo.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Sarcoma Aviar , Animales , Pollos , Gripe Aviar/prevención & control , Subtipo H5N1 del Virus de la Influenza A/genética
14.
Poult Sci ; 103(2): 103294, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38042040

RESUMEN

The bursa of Fabricius (BF) is pivotal for B lymphocyte ontogenesis. In the present investigation, a novel bursal peptide, designated BP7, was extracted from BF and was found to stimulate colony-forming unit pre-B (CFU pre-B) formation at various concentrations (1 µg/mL, P < 0.05; 5 µg/mL, P < 0.05; 25 µg/mL, P < 0.05). Moreover, BP7 modulated B cell differentiation pathways. The immunoregulatory potential of BP7 was further assessed in avian and murine models subjected to immunization with inactivated avian influenza virus (AIV, H9N2 subtype). BP7 significantly augmented AIV-specific antibody levels (Prime immunization: 5 mg/kg, P < 0.05; Boost immunization: 0.4, 1, and 5 mg/kg, P < 0.05) and cytokine secretion in the avian model (IL-4 and IFN-γ: 0.4, 1, and 5 mg/kg, P < 0.05). Similarly, in the murine model, AIV-specific antibody levels (Prime and Boost immunization: 0.4, 1, and 5 mg/kg, P < 0.05) and cytokine production (IL-4 and IFN-γ: 0.4, 1, and 5 mg/kg, P < 0.05) were notably enhanced. This study offers novel insights into the mechanisms underlying B cell maturation and holds implications for future immunopharmacological interventions.


Asunto(s)
Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Enfermedades de los Roedores , Animales , Ratones , Interleucina-4/metabolismo , Pollos , Péptidos/metabolismo , Bolsa de Fabricio , Enfermedades de los Roedores/metabolismo
15.
Vet Rec ; 194(2): e3616, 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38012027

RESUMEN

BACKGROUND: High-pathogenicity avian influenza (HPAI) has become a conservation threat to wild birds. Therefore, suitable vaccine technology and practical application methods require investigation. METHODS: Twenty-four African penguins (Spheniscus demersus) were vaccinated with either a conventional inactivated clade 2.3.4.4b H5N8 HPAI whole virus or a tobacco leaf-produced H5 haemagglutinin-based virus-like particle (VLP). Six birds received a second dose of the inactivated vaccine. Antibody responses were assessed and compared by employing haemagglutination inhibition tests. RESULTS: A second dose of inactivated vaccine was required to induce antibody titres above the level required to suppress virus shedding, while a single dose of VLP vaccine produced these levels by day 14, and one bird still had antibodies on day 430. LIMITATIONS: Bacterial contamination of the VLP vaccine limited the monitoring period and sample size in that treatment group, and it was not possible to perform a challenge study with field virus. CONCLUSION: VLP vaccines offer a more practical option than inactivated whole viruses, especially in logistically challenging situations involving wild birds.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Subtipo H5N8 del Virus de la Influenza A , Vacunas contra la Influenza , Gripe Aviar , Spheniscidae , Animales , Gripe Aviar/prevención & control , Virulencia , Pollos , Vacunación/veterinaria , Vacunas de Productos Inactivados
16.
PLoS Pathog ; 19(10): e1011685, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37819993

RESUMEN

Chicken lung is an important target organ of avian influenza virus (AIV) infection, and different pathogenic virus strains lead to opposite prognosis. Using a single-cell RNA sequencing (scRNA-seq) assay, we systematically and sequentially analyzed the transcriptome of 16 cell types (19 clusters) in the lung tissue of chickens infected with H5N1 highly pathogenic avian influenza virus (HPAIV) and H9N2 low pathogenic avian influenza virus (LPAIV), respectively. Notably, we developed a valuable catalog of marker genes for these cell types. Compared to H9N2 AIV infection, H5N1 AIV infection induced extensive virus replication and the immune reaction across most cell types simultaneously. More importantly, we propose that infiltrating inflammatory macrophages (clusters 0, 1, and 14) with massive viral replication, pro-inflammatory cytokines (IFN-ß, IL1ß, IL6 and IL8), and emerging interaction of various cell populations through CCL4, CCL19 and CXCL13, potentially contributed to the H5N1 AIV driven inflammatory lung injury. Our data revealed complex but distinct immune response landscapes in the lung tissue of chickens after H5N1 and H9N2 AIV infection, and deciphered the potential mechanisms underlying AIV-driven inflammatory reactions in chicken. Furthermore, this article provides a rich database for the molecular basis of different cell-type responses to AIV infection.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Lesión Pulmonar , Animales , Pollos/metabolismo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/genética , Análisis de la Célula Individual
17.
Virus Genes ; 59(5): 716-722, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37395889

RESUMEN

This study aims to screen and identify specific cluster miRNAs of H7N9 virus-infected N2a cells and explore the possible pathogenesis of these miRNAs. The N2a cells are infected with H7N9 and H1N1 influenza viruses, and the cells are collected at 12, 24 and 48 h to extract total RNA. To sequence miRNAs and identify different virus-specific miRNAs, high-throughput sequencing technology is used. Fifteen H7N9 virus-specific cluster miRNAs are screened, and eight of them are included in the miRBase database. These cluster-specific miRNAs regulate many signaling pathways, such as the PI3K-Akt signaling pathway, the RAS signaling pathway, the cAMP signaling pathway, actin cytoskeleton regulation and cancer-related genes. The study provides a scientific basis for the pathogenesis of H7N9 avian influenza, which is regulated by miRNAs.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Subtipo H7N9 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , MicroARNs , Animales , Humanos , Subtipo H7N9 del Virus de la Influenza A/genética , MicroARNs/genética , Subtipo H1N1 del Virus de la Influenza A/genética , Fosfatidilinositol 3-Quinasas , Gripe Humana/genética
18.
Biosens Bioelectron ; 237: 115423, 2023 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-37311406

RESUMEN

The rapid and sensitive detection of pathogenic viruses is important for controlling pandemics. Herein, a rapid, ultrasensitive, optical biosensing scheme was developed to detect avian influenza virus H9N2 using a genetically engineered filamentous M13 phage probe. The M13 phage was genetically engineered to bear an H9N2-binding peptide (H9N2BP) at the tip and a gold nanoparticle (AuNP)-binding peptide (AuBP) on the sidewall to form an engineered phage nanofiber, M13@H9N2BP@AuBP. Simulated modelling showed that M13@H9N2BP@AuBP enabled a 40-fold enhancement of the electric field enhancement in surface plasmon resonance (SPR) compared to conventional AuNPs. Experimentally, this signal enhancement scheme was employed for detecting H9N2 particles with a sensitivity down to 6.3 copies/mL (1.04 × 10-5 fM). The phage-based SPR scheme can detect H9N2 viruses in real allantoic samples within 10 min, even at very low concentrations beyond the detection limit of quantitative polymerase chain reaction (qPCR). Moreover, after capturing the H9N2 viruses on the sensor chip, the H9N2-binding phage nanofibers can be quantitatively converted into plaques that are visible to the naked eye for further quantification, thereby allowing us to enumerate the H9N2 virus particles through a second mode to cross-validate the SPR results. This novel phage-based biosensing strategy can be employed to detect other pathogens because the H9N2-binding peptides can be easily switched with other pathogen-binding peptides using phage display technology.


Asunto(s)
Bacteriófagos , Técnicas Biosensibles , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Nanopartículas del Metal , Nanofibras , Animales , Oro , Gripe Aviar/diagnóstico , Péptidos
19.
Rev. chil. infectol ; Rev. chil. infectol;40(3): 266-269, jun. 2023.
Artículo en Español | LILACS | ID: biblio-1515133

RESUMEN

Desde la segunda mitad de 2022 se ha reportado un aumento de casos de influenza en aves migratorias en Latinoamérica. Los virus influenza A y B son los principales agentes asociados a influenza estacional epidémica en humanos. Los virus influenza A circulan no solo en humanos sino también en animales, incluyendo aves migratorias. El intercambio de segmentos de ARN genómico entre dos virus del mismo tipo aumenta la diversidad de los subtipos circulantes e incluso puede facilitar la generación de progenie viral potencialmente pandémica. La naturaleza zoonótica del virus influenza A puede generar infecciones en humanos con virus de origen animal. El virus influenza A de origen aviar ha ocasionado transmisiones en humanos, incluyendo casos graves y muertes, siendo la influenza A H5N1 la más destacada. Es importante tomar medidas de prevención y control en caso de aumento de casos de influenza en aves migratorias para prevenir posibles pandemias en Chile y el mundo.


Since the second half of 2022, an increase in influenza cases in migratory birds has been reported in Latin America. Influenza A and B viruses are the main agents associated with seasonal epidemic influenza in humans. Influenza A viruses circulate not only in humans but also in animals, including migratory birds. The exchange of genomic RNA segments among two viruses increases the diversity of circulating subtypes and may even facilitate the generation of potentially pandemic viral progeny. The zoonotic nature of influenza A virus can generate infections in humans with animal-origin viruses. Avian-origin influenza A virus has caused transmissions in humans, including severe cases and deaths, with influenza A H5N1 being the most prominent. It is important to take preventive and control measures in case of an increase in influenza cases in migratory birds to prevent possible pandemics in Chile and the world.


Asunto(s)
Humanos , Animales , Gripe Humana/epidemiología , Subtipo H5N1 del Virus de la Influenza A , Gripe Aviar/epidemiología , Aves , Infecciones por Orthomyxoviridae , Gripe Humana/prevención & control , Gripe Humana/transmisión , Pandemias/prevención & control , Gripe Aviar/prevención & control , Gripe Aviar/transmisión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA