Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(34): e2203919119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969783

RESUMEN

Previous studies have shown that the Eurasian avian-like H1N1 (EA H1N1) swine influenza viruses circulated widely in pigs around the world and formed multiple genotypes by acquiring non-hemagglutinin and neuraminidase segments derived from other swine influenza viruses. Swine influenza control is not a priority for the pig industry in many countries, and it is worrisome that some strains may become more pathogenic and/or transmissible during their circulation in nature. Our routine surveillance indicated that the EA H1N1 viruses obtained different internal genes from different swine influenza viruses and formed various new genotypes. In this study, we found that a naturally isolated swine influenza reassortant, A/swine/Liaoning/265/2017 (LN265), a representative strain of one of the predominant genotypes in recent years, is lethal in mice and transmissible in ferrets. LN265 contains the hemagglutinin, neuraminidase, and matrix of the EA H1N1 virus; the basic polymerase 2, basic polymerase 1, acidic polymerase (PA), and nucleoprotein of the 2009 H1N1 pandemic virus; and the nonstructural protein of the North American triple-reassortment H1N2 virus. By generating and testing a series of reassortants and mutants, we found that four gradually accumulated mutations in PA are responsible for the increased pathogenicity and transmissibility of LN265. We further revealed that these mutations increase the messenger RNA transcription of viral proteins by enhancing the endonuclease cleavage activity and viral RNA-binding ability of the PA protein. Our study demonstrates that EA H1N1 swine influenza virus became pathogenic and transmissible in ferrets by acquiring key mutations in PA and provides important insights for monitoring field strains with pandemic potential.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae , ARN Polimerasa Dependiente del ARN , Enfermedades de los Porcinos , Animales , Hurones , Genotipo , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Ratones , Mutación , Neuraminidasa/genética , Infecciones por Orthomyxoviridae/veterinaria , Infecciones por Orthomyxoviridae/virología , Filogenia , ARN Polimerasa Dependiente del ARN/genética , Virus Reordenados/genética , Porcinos , Enfermedades de los Porcinos/virología , Proteínas Virales/genética
2.
PLoS Pathog ; 18(4): e1010446, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35377920

RESUMEN

Host defense systems employ posttranslational modifications to protect against invading pathogens. Here, we found that protein inhibitor of activated STAT 1 (PIAS1) interacts with the nucleoprotein (NP), polymerase basic protein 1 (PB1), and polymerase basic protein 2 (PB2) of influenza A virus (IAV). Lentiviral-mediated stable overexpression of PIAS1 dramatically suppressed the replication of IAV, whereas siRNA knockdown or CRISPR/Cas9 knockout of PIAS1 expression significantly increased virus growth. The expression of PIAS1 was significantly induced upon IAV infection in both cell culture and mice, and PIAS1 was involved in the overall increase in cellular SUMOylation induced by IAV infection. We found that PIAS1 inhibited the activity of the viral RNP complex, whereas the C351S or W372A mutant of PIAS1, which lacks the SUMO E3 ligase activity, lost the ability to suppress the activity of the viral RNP complex. Notably, the SUMO E3 ligase activity of PIAS1 catalyzed robust SUMOylation of PB2, but had no role in PB1 SUMOylation and a minimal role in NP SUMOylation. Moreover, PIAS1-mediated SUMOylation remarkably reduced the stability of IAV PB2. When tested in vivo, we found that the downregulation of Pias1 expression in mice enhanced the growth and virulence of IAV. Together, our findings define PIAS1 as a restriction factor for the replication and pathogenesis of IAV.


Asunto(s)
Virus de la Influenza A , Proteínas Inhibidoras de STAT Activados , Sumoilación , Replicación Viral , Animales , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/fisiología , Ratones , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Virulencia
3.
Emerg Infect Dis ; 29(7): 1367-1375, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37347504

RESUMEN

Highly pathogenic avian influenza (HPAI) subtype H5N1 clade 2.3.4.4b virus has spread globally, causing unprecedented large-scale avian influenza outbreaks since 2020. In 2021, we isolated 17 highly pathogenic avian influenza H5N1 viruses from wild birds in China. To determine virus origin, we genetically analyzed 1,529 clade 2.3.4.4b H5N1 viruses reported globally since October 2020 and found that they formed 35 genotypes. The 17 viruses belonged to genotypes G07, which originated from eastern Asia, and G10, which originated from Russia. The viruses were moderately pathogenic in mice but were highly lethal in ducks. The viruses were in the same antigenic cluster as the current vaccine strain (H5-Re14) used in China. In chickens, the H5/H7 trivalent vaccine provided complete protection against clade 2.3.4.4b H5N1 virus challenge. Our data indicate that vaccination is an effective strategy for preventing and controlling the globally prevalent clade 2.3.4.4b H5N1 virus.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Animales , Ratones , Subtipo H5N1 del Virus de la Influenza A/genética , Pollos , Animales Salvajes , Virus de la Influenza A/genética , China/epidemiología , Filogenia
4.
PLoS Pathog ; 17(4): e1009561, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33905456

RESUMEN

The H7N9 avian influenza virus (AIV) that emerged in China have caused five waves of human infection. Further human cases have been successfully prevented since September 2017 through the use of an H7N9 vaccine in poultry. However, the H7N9 AIV has not been eradicated from poultry in China, and its evolution remains largely unexplored. In this study, we isolated 19 H7N9 AIVs during surveillance and diagnosis from February 2018 to December 2019, and genetic analysis showed that these viruses have formed two different genotypes. Animal studies indicated that the H7N9 viruses are highly lethal to chicken, cause mild infection in ducks, but have distinct pathotypes in mice. The viruses bound to avian-type receptors with high affinity, but gradually lost their ability to bind to human-type receptors. Importantly, we found that H7N9 AIVs isolated in 2019 were antigenically different from the H7N9 vaccine strain that was used for H7N9 influenza control in poultry, and that replication of these viruses cannot, therefore, be completely prevented in vaccinated chickens. We further revealed that two amino acid mutations at positions 135 and 160 in the HA protein added two glycosylation sites and facilitated the escape of the H7N9 viruses from the vaccine-induced immunity. Our study provides important insights into H7N9 virus evolution and control.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/genética , Subtipo H7N9 del Virus de la Influenza A/aislamiento & purificación , Vacunas contra la Influenza/uso terapéutico , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/virología , Animales , Animales de Zoológico/virología , Pollos/virología , China/epidemiología , Patos/virología , Control de Infecciones/métodos , Subtipo H7N9 del Virus de la Influenza A/clasificación , Subtipo H7N9 del Virus de la Influenza A/fisiología , Gripe Aviar/epidemiología , Gripe Aviar/virología , Ratones , Filogenia , Vigilancia de la Población , Aves de Corral , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/prevención & control
5.
PLoS Pathog ; 17(2): e1009336, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33571308

RESUMEN

Posttranslational modifications, such as SUMOylation, play specific roles in the life cycle of invading pathogens. However, the effect of SUMOylation on the adaptation, pathogenesis, and transmission of influenza A virus (IAV) remains largely unknown. Here, we found that a conserved lysine residue at position 612 (K612) of the polymerase basic protein 1 (PB1) of IAV is a bona fide SUMOylation site. SUMOylation of PB1 at K612 had no effect on the stability or cellular localization of PB1, but was critical for viral ribonucleoprotein (vRNP) complex activity and virus replication in vitro. When tested in vivo, we found that the virulence of SUMOylation-defective PB1/K612R mutant IAVs was highly attenuated in mice. Moreover, the airborne transmission of a 2009 pandemic H1N1 PB1/K612R mutant virus was impaired in ferrets, resulting in reversion to wild-type PB1 K612. Mechanistically, SUMOylation at K612 was essential for PB1 to act as the enzymatic core of the viral polymerase by preserving its ability to bind viral RNA. Our study reveals an essential role for PB1 K612 SUMOylation in the pathogenesis and transmission of IAVs, which can be targeted for the design of anti-influenza therapies.


Asunto(s)
Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/transmisión , ARN Viral/metabolismo , Sumoilación , Proteínas Virales/metabolismo , Replicación Viral , Animales , Perros , Femenino , Hurones , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , ARN Viral/genética , Proteínas Virales/química , Proteínas Virales/genética , Acoplamiento Viral
6.
Euro Surveill ; 28(41)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37824247

RESUMEN

BackgroundTwo human cases of avian influenza A (H3N8) virus infection were reported in China in 2022.AimTo characterise H3N8 viruses circulating in China in September 2021-May 2022.MethodsWe sampled poultry and poultry-related environments in 25 Chinese provinces. After isolating H3N8 viruses, whole genome sequences were obtained for molecular and phylogenetic analyses. The specificity of H3N8 viruses towards human or avian receptors was assessed in vitro. Their ability to replicate in chicken and mice, and to transmit between guinea pigs was also investigated.ResultsIn total, 98 H3N8 avian influenza virus isolates were retrieved from 38,639 samples; genetic analysis of 31 representative isolates revealed 17 genotypes. Viruses belonging to 10 of these genotypes had six internal genes originating from influenza A (H9N2) viruses. These reassorted viruses could be found in live poultry markets and comprised the strains responsible for the two human infections. A subset of nine H3N8 viruses (including six reassorted) that replicated efficiently in mice bound to both avian-type and human-type receptors in vitro. Three reassorted viruses were shed by chickens for up to 9 days, replicating efficiently in their upper respiratory tract. Five reassorted viruses tested on guinea pigs were transmissible among these by respiratory droplets.ConclusionAvian H3N8 viruses with H9N2 virus internal genes, causing two human infections, occurred in live poultry markets in China. The low pathogenicity of H3N8 viruses in poultry allows their continuous circulation with potential for reassortment. Careful monitoring of spill-over infections in humans is important to strengthen early-warning systems and maintain influenza pandemic preparedness.


Asunto(s)
Subtipo H3N8 del Virus de la Influenza A , Subtipo H9N2 del Virus de la Influenza A , Gripe Aviar , Gripe Humana , Enfermedades de las Aves de Corral , Animales , Humanos , Ratones , Cobayas , Gripe Humana/epidemiología , Aves de Corral , Gripe Aviar/epidemiología , Subtipo H9N2 del Virus de la Influenza A/genética , Filogenia , Pollos , China/epidemiología , Enfermedades de las Aves de Corral/epidemiología
7.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31694949

RESUMEN

Influenza A virus (IAV) coopts numerous host factors to complete its replication cycle. Here, we identify free fatty acid receptor 2 (FFAR2) as a cofactor for IAV entry into host cells. We found that downregulation of FFAR2 or Ffar2 expression significantly reduced the replication of IAV in A549 or RAW 264.7 cells. The treatment of A549 cells with small interfering RNA (siRNA) targeting FFAR2 or the FFAR2 pathway agonists 2-(4-chlorophenyl)-3-methyl-N-(thiazol-2-yl)butanamide (4-CMTB) and compound 58 (Cmp58) [(S)-2-(4-chlorophenyl)-3,3-dimethyl-N-(5-phenylthiazol-2-yl)butanamide] dramatically inhibited the nuclear accumulation of viral nucleoprotein (NP) at early time points postinfection, indicating that FFAR2 functions in the early stage of the IAV replication cycle. FFAR2 downregulation had no effect on the expression of sialic acid (SA) receptors on the cell membrane, the attachment of IAV to the SA receptors, or the activity of the viral ribonucleoprotein (vRNP) complex. Rather, the amount of internalized IAVs was significantly reduced in FFAR2-knocked-down or 4-CMTB- or Cmp58-treated A549 cells. Further studies showed that FFAR2 associated with ß-arrestin1 and that ß-arrestin1 interacted with the ß2-subunit of the AP-2 complex (AP2B1), the essential adaptor of the clathrin-mediated endocytosis pathway. Notably, siRNA knockdown of either ß-arrestin1 or AP2B1 dramatically impaired IAV replication, and AP2B1 knockdown or treatment with Barbadin, an inhibitor targeting the ß-arrestin1/AP2B1 complex, remarkably decreased the amount of internalized IAVs. Moreover, we found that FFAR2 interacted with three G protein-coupled receptor (GPCR) kinases (i.e., GRK2, GRK5, and GRK6) whose downregulation inhibited IAV replication. Together, our findings demonstrate that the FFAR2 signaling cascade is important for the efficient endocytosis of IAV into host cells.IMPORTANCE To complete its replication cycle, IAV hijacks the host endocytosis machinery to invade cells. However, the underlying mechanisms of how IAV is internalized into host cells remain poorly understood, emphasizing the need to elucidate the role of host factors in IAV entry into cells. In this study, we identified FFAR2 as an important host factor for the efficient replication of both low-pathogenic and highly pathogenic IAV. We revealed that FFAR2 facilitates the internalization of IAV into target cells during the early stage of infection. Upon further characterization of the role of FFAR2-associated proteins in virus replication, we found that the FFAR2-ß-arrestin1-AP2B1 signaling cascade is important for the efficient endocytosis of IAV. Our findings thus further our understanding of the biological details of IAV entry into host cells and establish FFAR2 as a potential target for antiviral drug development.


Asunto(s)
Endocitosis , Virus de la Influenza A/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Internalización del Virus , Células A549 , Subunidades beta de Complejo de Proteína Adaptadora/genética , Subunidades beta de Complejo de Proteína Adaptadora/metabolismo , Animales , Perros , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Ratones , Células RAW 264.7 , Receptores Acoplados a Proteínas G/genética , Replicación Viral/fisiología , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo
8.
J Immunol ; 200(9): 3188-3200, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29563178

RESUMEN

Sepsis commonly results in acute and chronic brain dysfunction, which dramatically increases the morbidity associated with this common disease. Chronic brain dysfunction in animal models of sepsis survival is linked to persistent neuroinflammation and expression of multiple cytokines. However, we have found previously that microglia predominantly upregulate the damage associated molecule S100A8/A9 after sepsis. In this article, we show that S100A8/A9 is increased in the brains of patients who died of sepsis and that S100A8 is expressed in astrocytes and myeloid cells. Using a mouse model of sepsis survival, we show that S100A8/A9 is persistently expressed in the brain after sepsis. S100A9 expression is necessary for recruitment of neutrophils to the brain and for priming production of reactive oxygen species and TNF-α secretion in microglia and macrophages. However, despite improving these indices of chronic inflammation, S100A9 deficiency results in worsened anxiety-like behavior 2 wk after sepsis. Taken together, these results indicate that S100A8/A9 contributes to several facets of neuroinflammation in sepsis survivor mice, including granulocyte recruitment and priming of microglial-reactive oxygen species and cytokine production, and that these processes may be protective against anxiety behavior in sepsis survivors.


Asunto(s)
Lesiones Encefálicas/etiología , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Neuroinmunomodulación/fisiología , Sepsis/complicaciones , Animales , Ansiedad/etiología , Ansiedad/metabolismo , Conducta Animal/fisiología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/metabolismo , Calgranulina A/inmunología , Calgranulina B/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Sepsis/inmunología , Sepsis/metabolismo
9.
Infect Immun ; 87(1)2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30323031

RESUMEN

Legionella pneumophila causes life-threatening pneumonia culminating in acute lung injury. Innate and adaptive cytokines play an important role in host defense against L. pneumophila infection. Interleukin-36 (IL-36) cytokines are recently described members of the larger IL-1 cytokine family known to exert potent inflammatory effects. In this study, we elucidated the role for IL-36 cytokines in experimental pneumonia caused by L. pneumophila Intratracheal (i.t.) administration of L. pneumophila induced the upregulation of both IL-36α and IL-36γ mRNA and protein production in the lung. Compared to the findings for L. pneumophila-infected wild-type (WT) mice, the i.t. administration of L. pneumophila to IL-36 receptor-deficient (IL-36R-/-) mice resulted in increased mortality, a delay in lung bacterial clearance, increased L. pneumophila dissemination to extrapulmonary organs, and impaired glucose homeostasis. Impaired lung bacterial clearance in IL-36R-/- mice was associated with a significantly reduced accumulation of inflammatory cells and the decreased production of proinflammatory cytokines and chemokines. Ex vivo, reduced expression of costimulatory molecules and impaired M1 polarization were observed in alveolar macrophages isolated from infected IL-36R-/- mice compared to macrophages from WT mice. While L. pneumophila-induced mortality in IL-36α- or IL-36γ-deficient mice was not different from that in WT animals, antibody-mediated neutralization of IL-36γ in IL-36α-/- mice resulted in mortality similar to that observed in IL-36R-/- mice, indicating redundant and overlapping roles for these cytokines in experimental murine L. pneumophila pneumonia.


Asunto(s)
Interleucina-1/metabolismo , Legionella pneumophila/inmunología , Enfermedad de los Legionarios/inmunología , Enfermedad de los Legionarios/patología , Animales , Modelos Animales de Enfermedad , Femenino , Interleucina-1/deficiencia , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Supervivencia
10.
Neurobiol Dis ; 127: 350-361, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30910747

RESUMEN

Congenital generalized lipodystrophy 2 (CGL2) is characterized by loss of adipose tissue, insulin resistance and cognitive deficits and caused by mutation of BSCL2/seipin gene. Seipin deletion in mice and rats causes severe lipodystrophy, insulin resistance, and cognitive impairment. Hippocampal neurons express seipin protein. This study aimed to investigate the influence of systemic seipin knockout (seipin-sKO), neuronal seipin knockout (seipin-nKO) or adipose seipin knockout (seipin-aKO) in hippocampal tau phosphorylation and aggregation. Levels of tau phosphorylation at Thr212/Ser214 and Ser202/Thr205 and oligomer tau protein were increased in seipin-sKO mice and seipin-nKO mice with a decrease in axonal density and expression of PPARγ. Neuronal seipin deletion increased activities of GSK3ß and Akt/mTOR signaling, which were corrected by the administration of PPARγ agonist rosiglitazone for 7 days. The autophagosome formation was reduced in seipin-sKO mice and seipin-nKO mice, which was rescued by the Akt and mTOR inhibitors. The administration of rosiglitazone or Akt, mTOR and GSK3ß inhibitors for 7 days could correct the hyperphosphorylation and aggregation of tau. On the other hand, seipin-sKO mice appeared insulin resistance and an increase in phosphorylation of tau at Ser396 and JNK, which were corrected by treatment with rosiglitazone for 30 days rather than 7 days. Inhibition of JNK in seipin-sKO mice corrected the hyperphosphorylated tau at Ser396. The results indicate that neuronal seipin deletion causes hyperphosphorylation and aggregation of tau protein leading to axonal atrophy through reduced PPARγ to enhance GSK3ß and Akt/mTOR signaling; systemic seipin deletion-induced insulin resistance causes tau hyperphosphorylation via cascading JNK pathway.


Asunto(s)
Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Hipocampo/metabolismo , Resistencia a la Insulina/fisiología , Neuronas/metabolismo , PPAR gamma/metabolismo , Proteínas tau/metabolismo , Animales , Axones/metabolismo , Axones/patología , Subunidades gamma de la Proteína de Unión al GTP/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Ratones , Ratones Noqueados , Neuronas/patología , PPAR gamma/genética , Fosforilación , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
11.
PLoS Pathog ; 13(11): e1006737, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29166668

RESUMEN

Pseudomonas aeruginosa is a Gram-negative pathogen that can lead to severe infection associated with lung injury and high mortality. The interleukin (IL)-36 cytokines (IL-36α, IL-36ß and IL-36γ) are newly described IL-1 like family cytokines that promote inflammatory response via binding to the IL-36 receptor (IL-36R). Here we investigated the functional role of IL-36 cytokines in the modulating of innate immune response against P. aeruginosa pulmonary infection. The intratracheal administration of flagellated cytotoxic P. aeruginosa (ATCC 19660) upregulated IL-36α and IL-36γ, but not IL-36ß, in the lungs. IL-36α and IL-36γ were expressed in pulmonary macrophages (PMs) and alveolar epithelial cells in response to P. aeruginosa in vitro. Mortality after bacterial challenge in IL-36 receptor deficient (IL-36R-/-) mice and IL-36γ deficient (IL-36γ-/-) mice, but not IL-36α deficient mice, was significantly lower than that of wild type mice. Decreased mortality in IL-36R-/- mice and IL-36γ-/- mice was associated with reduction in bacterial burden in the alveolar space, bacterial dissemination, production of inflammatory cytokines and lung injury, without changes in lung leukocyte influx. Interestingly, IL-36γ enhanced the production of prostaglandin E2 (PGE2) during P. aeruginosa infection in vivo and in vitro. Treatment of PMs with recombinant IL-36γ resulted in impaired bacterial killing via PGE2 and its receptor; EP2. P. aeruginosa infected EP2 deficient mice or WT mice treated with a COX-2-specific inhibitor showed decreased bacterial burden and dissemination, but no change in lung injury. Finally, we observed an increase in IL-36γ, but not IL-36α, in the airspace and plasma of patients with P. aeruginosa-induced acute respiratory distress syndrome. Thus, IL-36γ and its receptor signal not only impaired bacterial clearance in a possible PGE2 dependent fashion but also mediated lung injury during P. aeruginosa infection.


Asunto(s)
Dinoprostona/metabolismo , Inmunidad Innata/inmunología , Interleucina-1/metabolismo , Lesión Pulmonar/metabolismo , Infecciones por Pseudomonas/inmunología , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Animales , Citocinas/metabolismo , Interleucina-1/genética , Macrófagos Alveolares/inmunología , Ratones Noqueados , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Receptores de Interleucina-1/genética , Transducción de Señal/inmunología
12.
Virol J ; 16(1): 119, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31640801

RESUMEN

BACKGROUND: In 2017-2018, a new highly pathogenic H5N6 avian influenza virus (AIV) variant appeared in poultry and wild birds in Asian and European countries and caused multiple outbreaks. These variant strains are different from the H5N6 virus associated with human infection in previous years, and their genetic taxonomic status and antigenicity have changed. Therefore, revision of the primers and probes of fluorescent RT-PCR is important to detect the new H5N6 subtype AIV in poultry and reduce the risk of an epidemic in birds or humans. METHODS: In this study, the primers and probes including three groups of HA and four groups of NA for H5N6 influenza virus were evaluated. Then a set of ideal primer and probes were selected to further optimize the reaction system and established a method of double rRT-PCR assay. The specificity of this method was determined by using H1~H16 subtype AIV. RESULTS: The results showed that fluorescence signals were obtained for H5 virus in FAM channel and N6 virus in VIC channel, and no fluorescent signal was observed in other subtypes of avian influenza viruses. The detection limit of this assay was 69 copies for H5 and 83 copies for N6 gene. And, the variability tests of intra- and inter-assay showed excellent reproducibility. Moreover, this assay showed 100% agreement with virus isolation method in detecting samples from poultry. CONCLUSION: The duplex rRT-PCR assay presented here has high specificity, sensitivity and reproducibility, and can be used for laboratory surveillance and rapid diagnosis of newly emerged H5N6 subtype avian influenza viruses.


Asunto(s)
Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/diagnóstico , Técnicas Microbiológicas/veterinaria , Técnicas de Diagnóstico Molecular/veterinaria , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Animales , Pollos , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Virus de la Influenza A/genética , Gripe Aviar/virología , Técnicas Microbiológicas/normas , Neuraminidasa/genética , ARN Viral/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Proteínas Virales/genética
13.
J Virol ; 90(21): 9797-9805, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558424

RESUMEN

The H5N1 avian influenza viruses emerged in Southeast Asia in the late 20th century and have evolved into multiple phylogenetic clades based on their hemagglutinin (HA)-encoding genes. The clade 7.2 viruses were first detected in chickens in northern China in 2006, and vaccines specifically targeted to the clade were developed and have been used in poultry in China since 2006. During routine surveillance and disease diagnosis, we isolated seven H5 viruses between 2011 and 2014 that bear the clade 7.2 HA genes. Here, we performed extensive studies to understand how the clade 7.2 H5 viruses have evolved in chickens in China. Full genome sequence analysis revealed that the seven viruses formed two subtypes (four H5N1 viruses and three H5N2 viruses) and four genotypes by deriving genes from other influenza viruses. All of the viruses had antigenically drifted from the clade 7.2 viruses that were isolated in 2006. Pathogenicity studies of four viruses, one from each genotype, revealed that all of the viruses are highly pathogenic in chickens, but none of them could replicate in ducks. The four viruses exclusively bound to avian-type receptors and replicated only in the turbinates and/or lungs of mice; none of them were lethal to mice at a dosage of 106 50% egg infective doses (EID50). Our study indicates that although the clade 7.2 viruses have not been eradicated from poultry through vaccination, they have not become more dangerous to other animals (e.g., ducks and mice) and humans. IMPORTANCE: Animal influenza viruses can acquire the ability to infect and kill humans. The H5N1 viruses have been a concern in recent decades because of their clear pandemic potential. We sorted H5N1 influenza viruses into different phylogenetic clades based on their HA genes. The clade 7.2 viruses were detected in chickens in several provinces of northern China in 2006. Vaccines for these viruses were subsequently developed and have been used ever since to control infection of poultry. Here, we analyzed the genetic and biologic properties of seven clade 7.2 viruses that were isolated from chickens between 2011 and 2014. We found that after nearly 9 years of circulation in chickens, the clade 7.2 viruses still exclusively bind to avian-type receptors and are of low pathogenicity to mice, suggesting that these H5 viruses pose a low risk to human public health.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Animales , Pollos , China/epidemiología , Patos/virología , Genoma Viral/genética , Genotipo , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N2 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/inmunología , Filogenia , Aves de Corral , Vacunación/métodos
14.
J Immunol ; 194(4): 1894-904, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595781

RESUMEN

Idiopathic pulmonary fibrosis is a devastating lung disease characterized by inflammation and the development of excessive extracellular matrix deposition. Currently, there are only limited therapeutic intervenes to offer patients diagnosed with pulmonary fibrosis. Although previous studies focused on structural cells in promoting fibrosis, our study assessed the contribution of macrophages. Recently, TLR signaling has been identified as a regulator of pulmonary fibrosis. IL-1R-associated kinase-M (IRAK-M), a MyD88-dependent inhibitor of TLR signaling, suppresses deleterious inflammation, but may paradoxically promote fibrogenesis. Mice deficient in IRAK-M (IRAK-M(-/-)) were protected against bleomycin-induced fibrosis and displayed diminished collagen deposition in association with reduced production of IL-13 compared with wild-type (WT) control mice. Bone marrow chimera experiments indicated that IRAK-M expression by bone marrow-derived cells, rather than structural cells, promoted fibrosis. After bleomycin, WT macrophages displayed an alternatively activated phenotype, whereas IRAK-M(-/-) macrophages displayed higher expression of classically activated macrophage markers. Using an in vitro coculture system, macrophages isolated from in vivo bleomycin-challenged WT, but not IRAK-M(-/-), mice promoted increased collagen and α-smooth muscle actin expression from lung fibroblasts in an IL-13-dependent fashion. Finally, IRAK-M expression is upregulated in peripheral blood cells from idiopathic pulmonary fibrosis patients and correlated with markers of alternative macrophage activation. These data indicate expression of IRAK-M skews lung macrophages toward an alternatively activated profibrotic phenotype, which promotes collagen production, leading to the progression of experimental pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Activación de Macrófagos/fisiología , Animales , Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Western Blotting , Separación Celular , Técnicas de Cocultivo , Colágeno , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibroblastos/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
15.
Anal Bioanal Chem ; 409(19): 4517-4528, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28646298

RESUMEN

This work established a fluorescence approach for detecting ascorbic acid (AA) based on Cu-ZnCdS quantum dots (Cu-ZnCdS QDs) and α-MnO2 nanorods. Cu-ZnCdS QDs and α-MnO2 nanorods were characterized by high-resolution transmission electron microscopy (HRTEM), fluorescence spectroscopy, inductively coupled plasma optical emission spectroscopy (ICP-OES) and X-ray diffraction (XRD). In the presence of α-MnO2 nanorods, the fluorescence of Cu-ZnCdS QDs was greatly quenched through the inner filter effect (IFE). Subsequently, AA can trigger the decomposition of the α-MnO2 nanorods which can reduce α-MnO2 to Mn2+ and recover the fluorescence. Under optimal conditions, a linear relation was obtained over the range 5.02-401.77 µM with a 31.62 µM detection limit. Through applying the fluorescent sensing system for detecting AA, a satisfactory result is obtained with recoveries ranging from 89.23% to 110.99%.

16.
Am J Respir Cell Mol Biol ; 53(6): 872-81, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25965198

RESUMEN

To protect the host against exuberant inflammation and injury responses, cells have the ability to become hyporesponsive or "tolerized" to repeated stimulation by microbial and nonmicrobial insults. The lung airspace is constantly exposed to a variety of exogenous and endogenous Toll-like receptor (TLR) ligands, yet the ability of alveolar epithelial cells (AECs) to be tolerized has yet to be examined. We hypothesize that type II AECs will develop a tolerance phenotype upon repeated TLR agonist exposure. To test this hypothesis, primary AECs isolated from the lungs of mice and a murine AEC cell line (MLE-12) were stimulated with either a vehicle control or a TLR ligand for 18 hours, washed, then restimulated with either vehicle or TLR ligand for an additional 6 hours. Tolerance was assessed by measurement of TLR ligand-stimulated chemokine production (monocyte chemoattractant protein [MCP]-1/CCL2, keratinocyte chemoattractant [KC]/CXCL1, and macrophage inflammatory protein [MIP]-2/CXCL2). Sequential treatment of primary AECs or MLE-12 cells with TLR agonists resulted in induction of either tolerance or cross-tolerance. The induction of tolerance was not due to expression of specific negative regulators of TLR signaling (interleukin-1 receptor associated kinase [IRAK]-M, Toll-interacting protein [Tollip], single Ig IL-1-related receptor [SIGIRR], or suppressor of cytokine signaling [SOCS]), inhibitory microRNAs (miRs; specifically, miR-155 and miR146a), or secretion of inhibitory or regulatory soluble mediators (prostaglandin E2, IL-10, transforming growth factor-ß, or IFN-α/ß). Moreover, inhibition of histone demethylation or DNA methylation did not prevent the development of tolerance. However, treatment of AECs with the histone deacetylase inhibitors trichostatin A or suberoylanilide hyrozamine resulted in reversal of the tolerance phenotype. These findings indicate a novel mechanism by which epigenetic modification regulates the induction of tolerance in AECs.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Epigénesis Genética/inmunología , Receptores Toll-Like/fisiología , Células Epiteliales Alveolares/inmunología , Animales , Células Cultivadas , Quimiocinas/biosíntesis , Quimiocinas/genética , Expresión Génica/inmunología , Tolerancia Inmunológica , Ligandos , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Toll-Like/agonistas
17.
BMC Microbiol ; 15: 168, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26289074

RESUMEN

BACKGROUND: The NS1 protein of avian influenza virus (AIV) is an important virulent factor of AIV. It has been shown to counteract host type I interferon response, to mediate host cell apoptosis, and to regulate the process of protein synthesis. The identification of AIV epitopes on NS1 protein is important for understanding influenza virus pathogenesis. RESULTS: In this paper, we describe the generation, identification, and epitope mapping of a NS1 protein-specific monoclonal antibody (MAb) D9. First, to induce the production of MAbs, BALB/c mice were immunized with a purified recombinant NS1 expressed in E. coli. The spleen cells from the immunized mice were fused with myeloma cells SP2/0, and through screening via indirect ELISAs, a MAb, named D9, was identified. Western blot assay results showed that MAb D9 reacted strongly with the recombinant NS1. Confocal laser scanning microscopy showed that this MAb also reacts with NS1 expressed in 293T cells that had been transfected with eukaryotic recombinant plasmids. Results from screening a phage display random 7-mer peptide library with MAb D9 demonstrated that it recognizes phages displaying peptides with the consensus peptide WNLNTV--VS, which closely matches the (182)WNDNTVRVS(190) of AIV NS1. Further identification of the displayed epitope was performed with a set of truncated polypeptides expressed as glutathione S-transferase fusion proteins, and the motif (182)WNDNT(186) was defined as the minimal unit of the linear B cell epitope recognized by MAb D9 in western blot assays. Moreover, homology analysis showed that this epitope is a conserved motif among AIV. CONCLUSIONS: We identified a conserved linear epitope, WNDNT, on the AIV NS1 protein that is recognized by MAb D9. This MAb and its epitope may facilitate future studies on NS1 function and aid the development of new diagnostic methods for AIV detection.


Asunto(s)
Mapeo Epitopo , Epítopos de Linfocito B/inmunología , Proteínas no Estructurales Virales/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/aislamiento & purificación , Aves , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Gripe Aviar/virología , Ratones Endogámicos BALB C , Biblioteca de Péptidos
18.
Virol J ; 12: 69, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25925390

RESUMEN

BACKGROUND: A novel reassortant H7N9 influenza A virus has crossed the species barrier from poultry to cause human infections in China in 2013 and 2014. Rapid detection of the novel H7N9 virus is important to detect this virus in poultry and reduce the risk of an epidemic in birds or humans. FINDINGS: In this study, a multiplex real-time RT-PCR (rRT-PCR) assay for rapid detection of H7N9 and other influenza A viruses was developed. To evaluate the assay, various influenza A viruses, other avian respiratory viruses, and 1,070 samples from poultry were tested. Fluorescence signals corresponding to H7 and N9 subtypes were detected only when H7 and N9 subtypes were present, while the fluorescence signal for the influenza A M gene was detected in all specimens with influenza A strains. The fluorescent signal can be detected in dilutions as low as 56 copies per reaction for the H7, N9 and M genes. Intra- and inter-assay variability tests showed a reliable assay. In poultry samples, a comparison of rRT-PCR with virus isolation showed a high level of agreement. CONCLUSIONS: The multiplex rRT-PCR assay in this study has good specificity, sensitivity and reproducibility, and will be useful for laboratory surveillance and rapid diagnosis of H7N9 and other influenza A viruses.


Asunto(s)
Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/diagnóstico , Gripe Aviar/virología , Técnicas de Diagnóstico Molecular/métodos , Reacción en Cadena de la Polimerasa Multiplex/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Animales , China , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Virus de la Influenza A/genética , Neuraminidasa/genética , Aves de Corral , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Medicina Veterinaria/métodos , Proteínas de la Matriz Viral/genética , Proteínas Virales/genética
19.
Arch Virol ; 160(10): 2471-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26179621

RESUMEN

In China, a novel reassortant influenza A (H7N9) virus, which has caused 435 cases of human infection, has recently emerged. Most cases of human infections with the H7N9 virus are known to be associated with a poultry farm and live-poultry markets. In this study, a one-step duplex real-time reverse transcription polymerase chain reaction (RRT-PCR) assay was developed for the simultaneous detection of the hemagglutinin (HA) and neuraminidase (NA) genes of the H7N9 virus for effective surveillance and early diagnosis of cases from clinical samples collected from live-poultry markets or poultry farms. The detection limit of this assay was as low as 0.1 EID50 of H7N9 viruses, which is similar to the detection limit of the real-time RT-PCR assay released by the Word Health Organization. The coefficients of variation (CVs) of both inter-assay and intra-assay reproducibility were less than 1.55 %, showing good reproducibility. No cross-reactivity was observed with RNA of other subtypes of influenza virus or other avian respiratory viruses. The assay can effectively detect H7N9 influenza virus RNA from multiple sources, including chickens, pigeons, ducks, humans, and the environment. Furthermore, the RRT-PCR assay was evaluated with more than 700 clinical samples collected from live-poultry markets and 120 experimentally infected chicken samples. Together, these results indicate that the duplex RRT-PCR assay is a specific, sensitive, and efficient diagnostic method for the epidemiological surveillance and diagnosis of H7N9 virus from different sources, particularly poultry samples.


Asunto(s)
Subtipo H7N9 del Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Enfermedades de las Aves de Corral/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Animales , Pollos , China , Columbidae , Patos , Humanos , Subtipo H7N9 del Virus de la Influenza A/genética , Gripe Aviar/diagnóstico , Gripe Humana/virología , Enfermedades de las Aves de Corral/diagnóstico
20.
J Immunol ; 190(1): 349-56, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23180821

RESUMEN

Hypersensitivity pneumonitis (HP) is an inflammatory lung disease that develops after repeated exposure to inhaled particulate Ag. Stachybotrys chartarum is a dimorphic fungus that has been implicated in a number of respiratory illnesses, including HP. In this study, we have developed a murine model of S. chartarum-induced HP that reproduces pathology observed in human HP, and we have hypothesized that TLR9-mediated IL-23 and IL-17 responses are required for the generation of granulomatous inflammation induced by inhaled S. chartarum. Mice that undergo i.p. sensitization and intratracheal challenge with 10(6) S. chartarum spores developed granulomatous inflammation with multinucleate giant cells, accompanied by increased accumulation of T cells. S. chartarum sensitization and challenge resulted in robust pulmonary expression of IL-17 and IL-23. S. chartarum-mediated granulomatous inflammation required intact IL-23 or IL-17 responses and required TLR9, because TLR9(-/-) mice displayed reduced IL-17 and IL-23 expression in whole lung associated with decreased accumulation of IL-17 expressing CD4(+) and γδ T cells. Compared with S. chartarum-sensitized dendritic cells (DC) isolated from WT mice, DCs isolated from TLR9(-/-) mice had a reduced ability to produce IL-23 in responses to S. chartarum. Moreover, shRNA knockdown of IL-23 in DCs abolished IL-17 production from splenocytes in response to Ag challenge. Finally, the intratracheal reconstitution of IL-23 in TLR9(-/-) mice recapitulated the immunopathology observed in WT mice. In conclusion, our studies suggest that TLR9 is critical for the development of Th17-mediated granulomatous inflammation in the lung in response to S. chartarum.


Asunto(s)
Alveolitis Alérgica Extrínseca/inmunología , Alveolitis Alérgica Extrínseca/patología , Interleucina-17/fisiología , Interleucina-23/fisiología , Stachybotrys/inmunología , Receptor Toll-Like 9/fisiología , Alveolitis Alérgica Extrínseca/genética , Animales , Antígenos Fúngicos/administración & dosificación , Antígenos Fúngicos/inmunología , Modelos Animales de Enfermedad , Humanos , Interleucina-17/biosíntesis , Interleucina-17/genética , Interleucina-23/administración & dosificación , Interleucina-23/genética , Intubación Intratraqueal , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Recombinantes/administración & dosificación , Stachybotrys/patogenicidad , Receptor Toll-Like 9/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA