Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 19(7): e1011489, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37399196

RESUMEN

The avian influenza virus (AIV) PA protein contributes to viral replication and pathogenicity; however, its interaction with innate immunity is not well understood. Here, we report that the H5 subtype AIV PA protein strongly suppresses host antiviral defense by interacting with and degrading a key protein in interferon (IFN) signaling, Janus kinase 1 (JAK1). Specifically, the AIV PA protein catalyzes the K48-linked polyubiquitination and degradation of JAK1 at lysine residue 249. Importantly, the AIV PA protein harboring 32T/550L degrades both avian and mammalian JAK1, while the AIV PA protein with residues 32M/550I degrades avian JAK1 only. Furthermore, the residues 32T/550L in PA protein confer optimum polymerase activity and AIV growth in mammalian cells. Notably, the replication and virulence of the AIV PA T32M/L550I mutant are attenuated in infected mice. Collectively, these data reveal an interference role for H5 subtype AIV PA protein in host innate immunity, which can be targeted for the development of specific and effective anti-influenza therapeutics.


Asunto(s)
Virus de la Influenza A , Gripe Aviar , Animales , Ratones , Virulencia , Pollos/metabolismo , Virus de la Influenza A/metabolismo , Proteínas no Estructurales Virales/metabolismo , Mamíferos
2.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35012978

RESUMEN

Preventing pathogenic viral and bacterial transmission in the human environment is critical, especially in potential outbreaks that may be caused by the release of ancient bacteria currently trapped in the permafrost. Existing commercial disinfectants present issues such as a high carbon footprint. This study proposes a sustainable alternative, a bioliquid derived from biomass prepared by hydrothermal liquefaction. Results indicate a high inactivation rate of pathogenic virus and bacteria by the as-prepared bioliquid, such as up to 99.99% for H1N1, H5N1, H7N9 influenza A virus, and Bacillus subtilis var. niger spores and 99.49% for Bacillus anthracis Inactivation of Escherichia coli and Staphylococcus epidermidis confirmed that low-molecular-weight and low-polarity compounds in bioliquid are potential antibacterial components. High temperatures promoted the production of antibacterial substances via depolymerization and dehydration reactions. Moreover, bioliquid was innoxious as confirmed by the rabbit skin test, and the cost per kilogram of the bioliquid was $0.04427, which is notably lower than that of commercial disinfectants. This study demonstrates the potential of biomass to support our biosafety with greater environmental sustainability.


Asunto(s)
Biomasa , Contención de Riesgos Biológicos , Ambiente , Energía Renovable , Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/ultraestructura , Humanos , Pruebas de Sensibilidad Microbiana , Peso Molecular , Pandemias , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/ultraestructura
3.
Int Immunol ; 35(5): 243-253, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36591893

RESUMEN

Immunogenicity can be evaluated by detecting antibodies (Abs) induced by an antigen. Presently deployed assays, however, do not consider the negative impacts of Ab poly-specificity, which is well established at the monoclonal antibody level. Here, we studied antibody poly-specificity at the serum level (i.e. nonspecific Ab-probe interactions, NSIs), and ended up establishing a new platform for viral peptide immunogenicity evaluation. We first selected three peptides of high, medium and low immunogenicity, using a 'vaccine serum response rate'-based approach (i.e. the gold standard). These three peptides (Pi) in the bovine serum albumin-Pi form were used to immunize chickens, resulting in longitudinal serum samples for screening with a non-cognate peptide library. The signal intensity of Ab-peptide specific binding and 'NSI count' was used to evaluate the viral peptides' immunogenicity. Only the NSI count agreed with the gold standard. The NSI count also provides more informative data on antibody production than the aggregated signal intensity by whole-protein-based indirect enzyme-linked immunosorbent assay.


Asunto(s)
Especificidad de Anticuerpos , Inmunoglobulinas , Péptidos , Proteínas Virales , Biblioteca de Péptidos , Inmunoglobulinas/sangre , Animales , Pollos , Virus de la Enfermedad de Newcastle/inmunología , Péptidos/inmunología , Ensayo de Inmunoadsorción Enzimática , Formación de Anticuerpos , Proteínas Virales/inmunología
4.
Sensors (Basel) ; 23(15)2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37571773

RESUMEN

Images captured under complex conditions frequently have low quality, and image performance obtained under low-light conditions is poor and does not satisfy subsequent engineering processing. The goal of low-light image enhancement is to restore low-light images to normal illumination levels. Although many methods have emerged in this field, they are inadequate for dealing with noise, color deviation, and exposure issues. To address these issues, we present CGAAN, a new unsupervised generative adversarial network that combines a new attention module and a new normalization function based on cycle generative adversarial networks and employs a global-local discriminator trained with unpaired low-light and normal-light images and stylized region loss. Our attention generates feature maps via global and average pooling, and the weights of different feature maps are calculated by multiplying learnable parameters and feature maps in the appropriate order. These weights indicate the significance of corresponding features. Specifically, our attention is a feature map attention mechanism that improves the network's feature-extraction ability by distinguishing the normal light domain from the low-light domain to obtain an attention map to solve the color bias and exposure problems. The style region loss guides the network to more effectively eliminate the effects of noise. The new normalization function we present preserves more semantic information while normalizing the image, which can guide the model to recover more details and improve image quality even further. The experimental results demonstrate that the proposed method can produce good results that are useful for practical applications.

5.
Emerg Infect Dis ; 28(8): 1664-1668, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35876682

RESUMEN

We investigated genetic and biologic characteristics of 2 Eurasian avian-like H1N1 swine influenza viruses from pigs in China that belong to the predominant G4 genotype. One swine isolate exhibited strikingly great homology to contemporaneous human Eurasian avian-like H1N1 isolates, preferential binding to the human-type receptor, and vigorous replication in mice without adaptation.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Enfermedades de los Porcinos , Animales , Aves , China/epidemiología , Genotipo , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/epidemiología , Ratones , Infecciones por Orthomyxoviridae/veterinaria , Filogenia , Virus Reordenados/genética , Porcinos , Enfermedades de los Porcinos/epidemiología
6.
Anal Chem ; 94(4): 1974-1982, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35044162

RESUMEN

Antibody-antigen (Ab-Ag) interactions are canonically described by a model that exclusively accommodates noninteraction (0) or reproducible interaction (RI) states, yet this model is inadequate to explain often-encountered nonreproducible signals. Here, by monitoring diverse experimental systems using a peptide-protein hybrid microarray, we observed that Ab-probe interactions comprise a substantial proportion of nonreproducible antibody-based results. This enabled our discovery and capacity to reliably identify nonreproducible Ab-probe interactions (NRIs), as well as our development of a powerful explanatory model ("0-NRI-RI-Hook four-state model") that is mAb concentration-dependent, regardless of specificity, which ultimately shows that both nonspecific interactions and NRIs are not predictable yet certain to happen. Our discoveries challenge the centrality of Ab-Ag interaction specificity data in serology and immunology.


Asunto(s)
Anticuerpos , Antígenos , Especificidad de Anticuerpos , Péptidos
7.
Ecotoxicol Environ Saf ; 244: 114032, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36084501

RESUMEN

The emergence of multidrug-resistant (MDR) bacteria harboring tet(X4), blaNDM or mcr-1 posed a serious threat to public health. Wild birds, especially migratory birds, were considered as one of important transmission vectors for antibiotic resistance genes (ARGs) globally, however, few studies were performed on the genomic epidemiology of critical resistance genes among them. Isolates harboring tet(X4), mcr-1 or blaNDM from migratory birds were identified and characterized by PCR, antimicrobial susceptibility testing, conjugation assays, whole genome sequencing and bioinformatics analysis. A total of 14 tet(X4)-bearing E. coli, 4 blaNDM-bearing E. coli and 23 mcr-1-bearing E. coli isolates were recovered from 1060 fecal samples of migratory birds. All isolates were MDR bacteria and most plasmids carrying tet(X4), blaNDM or mcr-1 were conjugative. We first identified an E. coli of migratory bird origin carrying blaNDM-4, which was located on a conjugative IncHI2 plasmid and embedded on a novel MDR region flanked by IS26 that could generate the circular intermediate. The emergency of E. coli isolates co-harboring mcr-1 and blaNDM-5 in migratory birds indicated the coexistence of ARGs in migratory birds was a novel threat. This study revealed the prevalence and molecular characteristics of three important ARGs in migratory birds, provided evidence that migratory birds were potential vectors of novel resistance genes and highlighted the monitoring of ARGs in migratory birds should be strengthened to prevent the spread of ARGs in a One Health strategy.


Asunto(s)
Infecciones por Escherichia coli , Proteínas de Escherichia coli , Animales , Antibacterianos , Aves , China , Colistina , Escherichia coli/genética , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/veterinaria , Proteínas de Escherichia coli/genética , Metagenómica , Pruebas de Sensibilidad Microbiana , Epidemiología Molecular , Plásmidos/genética , beta-Lactamasas/genética
8.
J Antimicrob Chemother ; 76(6): 1455-1458, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33758948

RESUMEN

BACKGROUND: Tigecycline and carbapenems are regarded as vital antimicrobials to treat serious bacterial infections. Co-occurrence of resistance genes conferring resistance to both tigecycline and carbapenems in Pseudomonas spp. was not investigated. OBJECTIVES: To characterize a megaplasmid co-harbouring tmexCD1-toprJ1 and blaVIM-2 in Pseudomonas putida of migratory bird origin. METHODS: One tigecycline- and carbapenem-resistant strain was isolated and characterized by antimicrobial susceptibility testing, conjugation assay, WGS and bioinformatics analysis. RESULTS: The strain P. putida ZXPA-20 resistant to meropenem and tigecycline was positive for blaVIM-2 and tmexCD1-toprJ1 genes. The gene blaVIM-2 was inserted into the backbone of the megaplasmid pZXPA-20 within a Tn5090-like structure. The genetic context of tmexCD1-toprJ1 in the megaplasmid was identical to many chromosomal tmexCD1-toprJ1 of Pseudomonas species. Plasmid-mediated tmexCD1-toprJ1 gene cluster in Pseudomonas spp. was more common than that in Klebsiella pneumoniae. To the best of our knowledge, this is the first report of co-occurrence of blaVIM-2 and tmexCD1-toprJ1 in one plasmid. CONCLUSIONS: Emergence of plasmid-mediated carbapenem and tigecycline resistance genes in P. putida from migratory birds highlighted the importance of surveillance of novel mobile resistance genes in migratory birds, which may play a vital role in global transmission of novel resistance genes.


Asunto(s)
Pseudomonas putida , Animales , Antibacterianos/farmacología , Aves , Carbapenémicos/farmacología , Resistencia a Múltiples Medicamentos , Pruebas de Sensibilidad Microbiana , Pseudomonas putida/genética , beta-Lactamasas
9.
FASEB J ; 34(8): 10132-10145, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32598086

RESUMEN

The newly reassorted IAV subtypes from zoonotic reservoirs respond poorly to current vaccines and antiviral therapy. There is an unmet need in developing novel antiviral drugs for better control of IAV infection. The cellular factors that are crucial for virus replication have been sought as novel molecular targets for antiviral therapy. Recent studies have shown that Janus kinases (JAK), JAK1, and JAK2, play an important role in IAV replication. Leflunomide is an anti-inflammatory drug primarily used for treating rheumatoid arthritis (RA). Prior studies suggest that A77 1726, the active metabolite of leflunomide, inhibits the activity of JAK1 and JAK3. Our current study aims to determine if A77 1726 can function as a JAK inhibitor to control IAV infection. Here, we report that A77 1726 inhibited the replication of three IAV subtypes(H5N1, H1N1, H9N2)in three cell types (chicken embryonic fibroblasts, A549, and MDCK). A77 1726 inhibited JAK1, JAK2, and STAT3 tyrosine phosphorylation. Similar observations were made with Ruxolitinib (Rux), a JAK-specific inhibitor. JAK2 overexpression enhanced H5N1 virus replication and compromised the antiviral activity of A77 1726. Leflunomide inhibited virus replication in the lungs of IAV-infected mice, alleviated their body weight loss, and prolonged their survival. Our study demonstrates for the first time the ability of A77 1726 to inhibit JAK2 activity and suggests that inhibition of JAK activity contributes to its antiviral activity.


Asunto(s)
Compuestos de Anilina/farmacología , Antirreumáticos/farmacología , Hidroxibutiratos/farmacología , Virus de la Influenza A/efectos de los fármacos , Quinasas Janus/antagonistas & inhibidores , Leflunamida/farmacología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Replicación Viral/efectos de los fármacos , Células A549 , Animales , Artritis Reumatoide/tratamiento farmacológico , Línea Celular , Línea Celular Tumoral , Crotonatos , Perros , Femenino , Humanos , Gripe Humana/tratamiento farmacológico , Gripe Humana/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Nitrilos , Infecciones por Orthomyxoviridae/metabolismo , Toluidinas
10.
Vet Res ; 52(1): 8, 2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33436086

RESUMEN

Since 2014, clade 2.3.4.4 has become the dominant epidemic branch of the Asian lineage H5 subtype highly pathogenic avian influenza virus (HPAIV) in southern and eastern China, while the H5N6 subtype is the most prevalent. We have shown earlier that lack of glycosylation at position 158 of the hemagglutinin (HA) glycoprotein due to the T160A mutation is a key determinant of the dual receptor binding property of clade 2.3.4.4 H5NX subtypes. Our present study aims to explore other effects of this site among H5N6 viruses. Here we report that N-linked glycosylation at site 158 facilitated the assembly of virus-like particles and enhanced virus replication in A549, MDCK, and chicken embryonic fibroblast (CEF) cells. Consistently, the HA-glycosylated H5N6 virus induced higher levels of inflammatory factors and resulted in stronger pathogenicity in mice than the virus without glycosylation at site 158. However, H5N6 viruses without glycosylation at site 158 were more resistant to heat and bound host cells better than the HA-glycosylated viruses. H5N6 virus without glycosylation at this site triggered the host immune response mechanism to antagonize the viral infection, making viral pathogenicity milder and favoring virus spread. These findings highlight the importance of glycosylation at site 158 of HA for the pathogenicity of the H5N6 viruses.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/metabolismo , Gripe Aviar/virología , Células A549/virología , Animales , Embrión de Pollo/virología , Pollos , Glicosilación , Pruebas de Hemaglutinación , Glicoproteínas Hemaglutininas del Virus de la Influenza/fisiología , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Gripe Aviar/inmunología , Gripe Humana/inmunología , Gripe Humana/virología , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Virus Genes ; 57(6): 521-528, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34519961

RESUMEN

H9N2 subtype avian influenza virus has dramatically evolved and undergone extensive reassortment since its emergence in early 1990s in China. The genotype S (G57), emerging in 2007 with the substitution of F98-like PB2 and M gene by G1-like ones, has become the overwhelming predominant genotype for the past 11 years since 2010. Here, we found that virus with G1-like PB2 were more efficient in protein expression and in infectious virus production than that with F98-like PB2 gene. By coinfected MDCK cells with the reassortant virus, more survival opportunity for viruses with G1-like PB2 than that of F/98-like was observed. Besides, in animal experiments, we found that the G1-like PB2 increases virus infectivity, replication, and virus shedding of H9N2 in chickens. Our results suggested that the substitution of G1-like PB2 play important role in promoting the fitness of genotype S H9N2 virus in China.


Asunto(s)
Subtipo H9N2 del Virus de la Influenza A , Virus Reordenados/genética , Proteínas Virales/genética , Animales , Pollos , Subtipo H9N2 del Virus de la Influenza A/genética , Replicación Viral
12.
BMC Vet Res ; 17(1): 22, 2021 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-33413356

RESUMEN

BACKGROUND: Hemagglutinin is a major surface protein in influenza A virus (IAV), and HA2 is relative conserved among different IAVs. It will be meaningful to identify broad-spectrum epitopes based on the HA2 protein. RESULTS: Overlapping peptides of the HA2 protein of the H5N1 IAV A/Mallard/Huadong/S/2005 were synthesized and loaded on modified silica gel film to form a microarray, and antisera against different subtypes of IAVs were used to screen universal epitopes. The selected epitope was further confirmed by western blotting using anti-peptide immune serum and viruses rescued with amino acid substitution. The results showed that 485-FYHKCDNECME-495 of the H5 14th peptide in HA2 had broad-spectrum binding activity with antisera against H1, H3, H4, H5, H6, H7, H8, H9, and H10 subtype IAV. Substitution of amino acids (K or D) in rescued viruses resulted in decreased serum binding, indicating that they were critical residues for serum binding activity. In Immune Epitope Database, some epitopes containing 14-4 peptide were confirmed as MHC-II-restricted CD4 T cell epitope and had effects on releasing IL-2 or IFN. CONCLUSION: The identified epitope should be a novel universal target for detection and vaccine design and its ability to generate immune protection needs further exploration.


Asunto(s)
Antígenos Virales/inmunología , Epítopos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Virus de la Influenza A/inmunología , Secuencia de Aminoácidos , Animales , Antígenos Virales/química , Embrión de Pollo , Pollos , Epítopos/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Sueros Inmunes , Subtipo H5N1 del Virus de la Influenza A/química , Virus de la Influenza A/química , Péptidos/química
13.
BMC Vet Res ; 17(1): 80, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33588843

RESUMEN

BACKGROUND: Genotype S H9N2 viruses have become predominant in poultry in China since 2010. These viruses frequently donate their whole internal gene segments to other emerging influenza A subtypes such as the novel H7N9, H5N6, and H10N8 viruses. We recently reported that the PB2 and M genes of the genotype S H9N2 virus, which are derived from the G1-like virus, enhance the fitness of H5Nx and H7N9 avian influenza viruses in chickens and mice. However, whether the G1-like PB2 and M genes are preferentially incorporated into progeny virions during virus reassortment remains unclear; whether the G1-like PB2 and M genes from different subtypes are differentially incorporated into new virion progeny remains unknown. RESULTS: We conducted a reassortment experiment with the use of a H7N9 virus as the backbone and found that G1-like M/PB2 genes were preferentially incorporated in progeny virions over F/98-like M/PB2 genes. Importantly, the preference varied among G1-like M/PB2 genes of different subtypes. When competing with F/98-like M/PB2 genes during reassortment, both the M and PB2 genes from the H7N9 virus GD15 showed an advantage, whereas only the PB2 gene from the H9N2 virus CZ73 and the M gene from the H9N2 virus AH320 displayed the advantage. CONCLUSION: Our findings highlight the preferential and variable advantages of H9N2-derived G1-like M and PB2 genes in incorporating them into H7N9 progeny virions over SH14-derived F/98-like M/PB2 genes.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/genética , Virus Reordenados/genética , Animales , Embrión de Pollo , Coinfección , Perros , Genotipo , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Virus Reordenados/crecimiento & desarrollo , Proteínas de la Matriz Viral/genética , Virión
14.
Virol J ; 17(1): 74, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32532301

RESUMEN

BACKGROUND: Although influenza A virus (IAV) employs diverse strategies to evade IFN responses by inhibiting the synthesis of IFN, how IAV regulates signaling downstream of IFN is incompletely understood. METHODS: In this study, we used Western blot-based protein analysis coupled with RT-qPCR, overexpression and RNA interference to investigate the regulation of JAK1 by IAV infection. RESULTS: The results indicated that JAK1 was ubiquitinated and degraded, resulting in inhibition of type I and type II IFN responses, demonstrating that IAV antagonizes the IFN-activated JAK/STAT signaling pathway by inducing the degradation of JAK1. Furthermore. IAV infection upregulated the suppressor of cytokine signaling (SOCS) protein SOCS1, and SOCS1 mediated the ubiquitination and degradation of JAK1. CONCLUSION: Collectively, our findings suggest that IAV infection induces SOCS1 expression to promote JAK1 degradation, which in turn inhibits host innate immune responses.


Asunto(s)
Virus de la Influenza A/inmunología , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Janus Quinasa 1/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Células A549 , Regulación hacia Abajo , Células HEK293 , Interacciones Microbiota-Huesped/inmunología , Humanos , Evasión Inmune , Inmunidad Innata , Transducción de Señal , Ubiquitinación
15.
Arch Virol ; 164(3): 807-817, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30671655

RESUMEN

The emergent highly pathogenic avian influenza A (H7N9) (HPAI) virus is a major public concern in China. Therefore, it is crucially important to develop an effective vaccine against this virus. In this study, we constructed a baculovirus vaccine expressing the hemagglutinin (HA) of H7N9 strain A/Chicken/Jiaxing/148/2014 (JX148). The recombinant baculovirus (rBac-JX148HA) generated in this study showed good growth in insect cells and good safety, and it stably expressed the HA protein. We compared the immunogenicity and efficacy of the inactivated whole-virus vaccine JX148 and rBac-JX148HA. One chicken in the JX148-treated group died on day 4 post-challenge, and three chickens had typical clinical symptoms (survival rate, 90%; morbidity, 40%). However, no chickens immunized with rBac-JX148HA showed clinical signs during the 14-day observation period. An analysis of viral shedding and viral replication demonstrated that rBac-JX148HA more efficiently inhibited viral shedding and viral replication than the inactivated whole-virus vaccine. Taken together, these results indicate that the inactivated recombinant baculovirus vaccine induces a high hemagglutination inhibition antibody titer, provides complete protection against challenge with the highly pathogenic H7N9 virus, and effectively inhibits viral shedding. Therefore, the candidate vaccine has potential utility in the prevention and control of H7N9 avian influenza and is also appropriate for veterinary vaccines using cell suspension culture technology.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/administración & dosificación , Subtipo H7N9 del Virus de la Influenza A/inmunología , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Animales , Anticuerpos Antivirales/inmunología , Baculoviridae/genética , Baculoviridae/metabolismo , Pollos , China , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/patogenicidad , Subtipo H7N9 del Virus de la Influenza A/fisiología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Vacunación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Virulencia , Esparcimiento de Virus
16.
Arch Virol ; 163(4): 947-960, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29307089

RESUMEN

During surveillance for avian influenza viruses, three H5N6 viruses were isolated in chickens obtained from live bird markets in eastern China, between January 2015 and April 2016. Sequence analysis revealed a high genomic homology between these poultry isolates and recent human H5N6 variants whose internal genes were derived from genotype S H9N2 avian influenza viruses. Glycan binding assays revealed that all avian H5N6 viruses were capable of binding to both human-type SAα-2,6Gal receptors and avian-type SAα-2,3Gal receptors. Their biological characteristics were further studied in BALB/c mice, specific-pathogen-free chickens, and mallard ducks. All three isolates had low pathogenicity in mice but were highly pathogenic to chickens, as evidenced by 100% mortality 36-120 hours post infection at a low dose of 103.0EID50 and through effective contact transmission. Moreover, all three poultry H5N6 isolates caused asymptomatic infections in ducks, which may serve as a reservoir host for their maintenance and dissemination; these migrating waterfowl could cause a potential global pandemic. Our study suggests that continuous epidemiological surveillance in poultry should be implemented for the early prevention of future influenza outbreaks.


Asunto(s)
Genes Virales , Subtipo H9N2 del Virus de la Influenza A/genética , Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Enfermedades de las Aves de Corral/epidemiología , Virus Reordenados/genética , Receptores Virales/genética , Animales , Enfermedades Asintomáticas , Pollos/virología , China/epidemiología , Patos/virología , Monitoreo Epidemiológico , Expresión Génica , Genotipo , Humanos , Subtipo H9N2 del Virus de la Influenza A/clasificación , Subtipo H9N2 del Virus de la Influenza A/inmunología , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/clasificación , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Gripe Aviar/transmisión , Gripe Aviar/virología , Ratones , Ratones Endogámicos BALB C , Filogenia , Polisacáridos/química , Polisacáridos/metabolismo , Aves de Corral/virología , Enfermedades de las Aves de Corral/transmisión , Enfermedades de las Aves de Corral/virología , Unión Proteica , Virus Reordenados/clasificación , Virus Reordenados/inmunología , Virus Reordenados/patogenicidad , Receptores Virales/inmunología
17.
Med Microbiol Immunol ; 206(2): 125-147, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28000052

RESUMEN

We previously reported a pair of H5N1 avian influenza viruses which are genetically similar but differ greatly in their virulence in mice. A/Chicken/Jiangsu/k0402/2010 (CK10) is highly lethal to mice, whereas A/Goose/Jiangsu/k0403/2010 (GS10) is avirulent. In this study, to investigate the host factors that account for their virulence discrepancy, we compared the pathology and host proteome of the CK10- or GS10-infected mouse lung. Moderate lung injury was observed from CK10-infected animals as early as the first day of infection, and the pathology steadily progressed at later time point. However, only mild lesions were observed in GS10-infected mouse lung at the late infection stage. Using the quantitative iTRAQ coupled LC-MS/MS method, we first found that more significantly differentially expressed (DE) proteins were stimulated by GS10 compared with CK10. However, bio-function analysis of the DE proteins suggested that CK10 induced much stronger inflammatory response-related functions than GS10. Canonical pathway analysis also demonstrated that CK10 highly activated the "Acute Phase Response Signaling," which results in a wide range of biological activities in response to viral infection, including many inflammatory processes. Further in-depth analysis showed that CK10 exacerbated acute lung injury-associated responses, including inflammatory response, cell death, reactive oxygen species production and complement response. In addition, some of these identified proteins that associated with the lung injury were further confirmed to be regulated in vitro. Therefore, our findings suggest that the early increased lung injury-associated host response induced by CK10 may contribute to the lung pathology and the high virulence of this virus in mice.


Asunto(s)
Interacciones Huésped-Patógeno , Subtipo H5N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Proteoma , Proteómica/métodos , Reacción de Fase Aguda/metabolismo , Reacción de Fase Aguda/virología , Animales , Comunicación Celular , Línea Celular , Cromatografía Liquida , Análisis por Conglomerados , Biología Computacional/métodos , Femenino , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Ratones , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/patología , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Reproducibilidad de los Resultados , Transducción de Señal , Espectrometría de Masas en Tándem
18.
Vet Res ; 48(1): 67, 2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-29070059

RESUMEN

Highly pathogenic avian influenza (HPAI) H5N8 virus has caused considerable economic losses to poultry industry and poses a great threat to public health. Our previous study revealed two genetically similar HPAI H5N8 viruses displaying completely different virulence in mice. However, the molecular basis for viral pathogenicity to mammals remains unknown. Herein, we generated a series of reassortants between the two viruses and evaluated their virulence in mice. We demonstrated that 283M in PB2 is a new mammalian virulence marker for H5 viruses and that synergistic effect of amino acid residues 283M and 526R in PB2 is responsible for high virulence of the HPAI H5N8 virus. Analysis of available PB2 sequences showed that PB2 283M is highly conserved among influenza A viruses, while PB2 526R presents in most of human H3N2 and H5N1 isolates. Further study confirmed that the residues 283M and 526R had similar impacts on an HPAI H5N1 virus, suggesting that influenza viruses with both residues may replicate well in mammalian hosts. Together, these results present new insights for synergistic effect of 283M and 526R in PB2 of H5 HPAI virus on virulence to mammalian host, furthering our understanding of the pathogenesis of influenza A virus.


Asunto(s)
Subtipo H5N8 del Virus de la Influenza A/genética , Subtipo H5N8 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Animales , Células Cultivadas , Pollos , Perros , Femenino , Fibroblastos , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Ratones , Virulencia
19.
Vet Res ; 48(1): 81, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162128

RESUMEN

A stem glycosylation site of hemagglutinin (HA) is important to the stability of the HA trimmer. A previous study shows that the stem 10/11 overlap glycosylation site of the H5 subtype avian influenza virus may influence the cleavage of HA, whereas the exact site and its effect on virulence remain unclear. In this study, site-directed mutagenesis was used to generate single or double mutant rSY-Δ10(10NNAT), rSY-Δ11(10NNSA), and rSY-Δ10/11(10NNAA) of the overlapping glycosylation site (10NNST) on the HA of A/Mallard/Huadong/S/2005(SY). By using Western blot analysis, we show that both rSY-Δ11 and rSY-Δ10/11 mutant viruses had significant delay on HA cleavage and a reduced HA molecular mass compared to the wild-type virus rSY, while the rSY-Δ10 mutant virus exhibited a similar HA molecular mass to that of the wild-type virus rSY. Interestingly, both rSY-Δ11 and rSY-Δ10/11 mutant viruses reverted their glycosylation sites at 11N after passage, indicating that 11N is a true and critical glycosylation site. Compared to the wild-type virus rSY, rSY-Δ11 and rSY-Δ10/11 mutant viruses had decreased growth rates, reduced thermo- and pH-stability, decreased pathogenicity, and limited systemic spread. Therefore, our study suggests that the 11N glycosylation site plays a key role in HA cleavage, structural stability and pathogenicity in H5 subtype avian influenza virus.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Animales , Línea Celular , Embrión de Pollo , Perros , Fibroblastos , Glicosilación , Células HEK293 , Humanos , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Células de Riñón Canino Madin Darby , Mutagénesis Sitio-Dirigida , Virulencia
20.
Vet Res ; 48(1): 7, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28166830

RESUMEN

We generated and characterized site-directed HA mutants on the genetic backbone of H5N1 clade 2.3.4 virus preferentially binding to α-2,3 receptors in order to identify the key determinants in hemagglutinin rendering the dual affinity to both α-2,3 (avian-type) and α-2,6 (human-type) linked sialic acid receptors of the current clade 2.3.4.4 H5NX subtype avian influenza reassortants. The results show that the T160A substitution resulted in the loss of a glycosylation site at 158N and led not only to enhanced binding specificity for human-type receptors but also transmissibility among guinea pigs, which could be considered as an important molecular marker for assessing pandemic potential of H5 subtype avian influenza isolates.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/virología , Sustitución de Aminoácidos/genética , Sustitución de Aminoácidos/fisiología , Animales , Femenino , Cobayas/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/transmisión , Receptores Virales/metabolismo , Replicación Viral/genética , Replicación Viral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA