Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Virol ; 92(11)2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29563291

RESUMEN

Nonstructural protein 1 (NS1) of influenza A virus regulates innate immune responses via various mechanisms. We previously showed that a naturally occurring deletion (the EALQR motif) in the NS1 effector domain of an H5N1 swine-origin avian influenza virus impairs the inhibition of type I interferon (IFN) in chicken fibroblasts and attenuates virulence in chickens. Here we found that the virus bearing this deletion in its NS1 effector domain showed diminished inhibition of IFN-related cytokine expression and attenuated virulence in mice. We further showed that deletion of the EALQR motif disrupted NS1 dimerization, impairing double-stranded RNA (dsRNA) sequestration and competitive binding with RIG-I. In addition, the EALQR-deleted NS1 protein could not bind to TRIM25, unlike full-length NS1, and was less able to block TRIM25 oligomerization and self-ubiquitination, further impairing the inhibition of TRIM25-mediated RIG-I ubiquitination compared to that with full-length NS1. Our data demonstrate that the EALQR deletion prevents NS1 from blocking RIG-I-mediated IFN induction via a novel mechanism to attenuate viral replication and virulence in mammalian cells and animals.IMPORTANCE H5 highly pathogenic avian influenza viruses have infected more than 800 individuals across 16 countries, with an overall case fatality rate of 53%. Among viral proteins, nonstructural protein 1 (NS1) of influenza virus is considered a key determinant for type I interferon (IFN) antagonism, pathogenicity, and host range. However, precisely how NS1 modulates virus-host interaction, facilitating virus survival, is not fully understood. Here we report that a naturally occurring deletion (of the EALQR motif) in the NS1 effector domain of an H5N1 swine-origin avian influenza virus disrupted NS1 dimerization, which diminished the blockade of IFN induction via the RIG-I signaling pathway, thereby impairing virus replication and virulence in the host. Our study demonstrates that the EALQR motif of NS1 regulates virus fitness to attain a virus-host compromise state in animals and identifies this critical motif as a potential target for the future development of small molecular drugs and attenuated vaccines.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Interferón Tipo I/inmunología , Proteínas no Estructurales Virales/genética , Células A549 , Animales , Línea Celular Tumoral , Embrión de Pollo , Chlorocebus aethiops , Proteínas de Unión al ADN/metabolismo , Femenino , Células HEK293 , Humanos , Inmunidad Innata/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/genética , Dominios Proteicos/genética , Receptores de Superficie Celular , Eliminación de Secuencia/genética , Células THP-1 , Factores de Transcripción/metabolismo , Ubiquitinación , Células Vero , Proteínas no Estructurales Virales/metabolismo
2.
J Virol ; 91(12)2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28404845

RESUMEN

H5N6 avian influenza virus (AIV) has posed a potential threat to public health since its emergence in China in 2013. To understand the evolution and emergence of H5N6 AIV in the avian population, we performed molecular surveillance of live poultry markets (LPMs) in Wugang Prefecture, Hunan Province, in central China, during 2014 and 2015. Wugang Prefecture is located on the Eastern Asian-Australian migratory bird flyway, and a human death due to an H5N6 virus was reported in the prefecture on 21 November 2016. In total, we sampled and sequenced the complete genomes of 175 H5N6 AIVs. Notably, our analysis revealed that H5N6 AIVs contain at least six genotypes arising from segment reassortment, including a rare variant that possesses an HA gene derived from H5N1 clade 2.3.2 and a novel NP gene that has its origins with H7N3 viruses. In addition, phylogenetic analysis revealed that genetically similar H5N6 AIVs tend to cluster according to their geographic regions of origin. These results help to reveal the evolutionary behavior of influenza viruses prior to their emergence in humans.IMPORTANCE The newly emerged H5N6 influenza A virus has caused more than 10 human deaths in China since 2013. In November 2016, a human death due to an H5N6 virus, in Wugang Prefecture, Hunan Province, was confirmed by the WHO. To better understand the evolution and emergence of H5N6 viruses, we surveyed live poultry markets (LPMs) in Wugang Prefecture before the reported human death, with a focus on revealing the diversity and genomic origins of H5N6 in birds during 2014 and 2015. In general, H5N6 viruses in this region were most closely related to H5N1 clade 2.3.4.4, with the exception of one virus with an HA gene derived from clade 2.3.2 such that it represents a novel reassortant. Clearly, the ongoing surveillance of LPMs is central to monitoring the emergence of pathogenic influenza viruses.


Asunto(s)
Aves/virología , Evolución Molecular , Genoma Viral , Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Gripe Aviar/virología , Animales , China/epidemiología , Monitoreo Epidemiológico , Variación Genética , Genotipo , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Gripe Humana/virología , Filogenia , Aves de Corral/virología , ARN Viral/genética , Virus Reordenados
3.
Arch Virol ; 162(5): 1349-1353, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28116526

RESUMEN

In this study, we analyzed the genome of a H10N5 influenza virus from wild birds. This virus was identified as a novel reassortant virus with internal genes from multiple subtypes and of distinct origins. After sequential passage in mice, mouse-adapted viruses bearing mutations PB2-E627K and HA-G218E were generated. These viruses caused dramatic body weight loss and death, and also replicated in mouse brain, suggesting that the pathogenicity of low pathogenic H10N5 in chickens can be enhanced after passage in mammals. Our data imply that H10N5 viruses might be a potential risk to human health therefore it is important to undertake continued surveillance and biosecurity evaluation of these viruses.


Asunto(s)
Enfermedades de las Aves/virología , Aves/virología , Hemaglutininas Virales/genética , Gripe Aviar/virología , Virus Reordenados/clasificación , Virus Reordenados/genética , Animales , Animales Salvajes/virología , Embrión de Pollo , Femenino , Marcadores Genéticos/genética , Ratones , Ratones Endogámicos BALB C , Virus Reordenados/aislamiento & purificación
4.
Emerg Microbes Infect ; 12(2): 2244091, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37526446

RESUMEN

High pathogenicity avian influenza (HPAI) H5N1 is a subtype of the influenza A virus primarily found in birds. The subtype emerged in China in 1996 and has spread globally, causing significant morbidity and mortality in birds and humans. In Cambodia, a lethal case was reported in February 2023 involving an 11-year-old girl, marking the first human HPAI H5N1 infection in the country since 2014. This research examined the zoonotic potential of the human H5N1 isolate, A/Cambodia/NPH230032/2023 (KHM/23), by assessing its receptor binding, fusion pH, HA thermal stability, and antigenicity. Results showed that KHM/23 exhibits similar receptor binding and antigenicity as the early clade 2.3.2.1c HPAI H5N1 strain, and it does not bind to human-like receptors. Despite showing limited zoonotic risk, the increased thermal stability and reduced pH of fusion in KHM/23 indicate a potential threat to poultry, emphasizing the need for vigilant monitoring.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Gripe Humana , Animales , Femenino , Humanos , Niño , Gripe Aviar/epidemiología , Hemaglutininas , Gripe Humana/epidemiología , Cambodia/epidemiología
5.
Viruses ; 13(2)2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33546185

RESUMEN

The long-term control strategy of SARS-CoV-2 and other major respiratory viruses needs to include antivirals to treat acute infections, in addition to the judicious use of effective vaccines. Whilst COVID-19 vaccines are being rolled out for mass vaccination, the modest number of antivirals in use or development for any disease bears testament to the challenges of antiviral development. We recently showed that non-cytotoxic levels of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, induces a potent host innate immune antiviral response that blocks influenza A virus replication. Here we show that TG is also highly effective in blocking the replication of respiratory syncytial virus (RSV), common cold coronavirus OC43, SARS-CoV-2 and influenza A virus in immortalized or primary human cells. TG's antiviral performance was significantly better than remdesivir and ribavirin in their respective inhibition of OC43 and RSV. Notably, TG was just as inhibitory to coronaviruses (OC43 and SARS-CoV-2) and influenza viruses (USSR H1N1 and pdm 2009 H1N1) in separate infections as in co-infections. Post-infection oral gavage of acid-stable TG protected mice against a lethal influenza virus challenge. Together with its ability to inhibit the different viruses before or during active infection, and with an antiviral duration of at least 48 h post-TG exposure, we propose that TG (or its derivatives) is a promising broad-spectrum inhibitor against SARS-CoV-2, OC43, RSV and influenza virus.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Coronavirus Humano OC43/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Virus Sincitial Respiratorio Humano/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Tapsigargina/farmacología , Animales , Antivirales/uso terapéutico , Betacoronavirus/fisiología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Coronavirus Humano OC43/fisiología , Estrés del Retículo Endoplásmico , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Ratones , Pruebas de Sensibilidad Microbiana , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , Virus Sincitial Respiratorio Humano/fisiología , Ribavirina/farmacología , SARS-CoV-2/fisiología , Tapsigargina/uso terapéutico , Replicación Viral/efectos de los fármacos
6.
Viruses ; 12(10)2020 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-32992478

RESUMEN

Influenza A virus is a major global pathogen of humans, and there is an unmet need for effective antivirals. Current antivirals against influenza A virus directly target the virus and are vulnerable to mutational resistance. Harnessing an effective host antiviral response is an attractive alternative. We show that brief exposure to low, non-toxic doses of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, promptly elicits an extended antiviral state that dramatically blocks influenza A virus production. Crucially, oral administration of TG protected mice against lethal virus infection and reduced virus titres in the lungs of treated mice. TG-induced ER stress unfolded protein response appears as a key driver responsible for activating a spectrum of host antiviral defences that include an enhanced type I/III interferon response. Our findings suggest that TG is potentially a viable host-centric antiviral for the treatment of influenza A virus infection without the inherent problem of drug resistance.


Asunto(s)
Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H3N8 del Virus de la Influenza A/crecimiento & desarrollo , Tapsigargina/farmacología , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Embrión de Pollo , Chlorocebus aethiops , Perros , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Gripe Humana/tratamiento farmacológico , Interferón Tipo I/efectos de los fármacos , Interferón Tipo I/inmunología , Interferones/efectos de los fármacos , Interferones/inmunología , Ratones , Ratones Endogámicos BALB C , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/antagonistas & inhibidores , Porcinos , Respuesta de Proteína Desplegada/efectos de los fármacos , Células Vero , Interferón lambda
7.
Vet Microbiol ; 235: 234-242, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31383307

RESUMEN

During 2012-2015, six H5N1 avian influenza viruses were isolated from domestic birds and the environment around Qinghai Lake. Phylogenetic analysis of HA genes revealed that A/chicken/Gansu/XG2/2012 (CK/GS/XG2/12) belonged to clade 2.3.2.1a, while A/environment/Qinghai/1/2013 (EN/QH/1/13), A/chicken/Qinghai/QH1/2015 (CK/QH/QH1/15), A/chicken/Qinghai/QH2/2015 (CK/QH/QH2/15), A/chicken/Qinghai/QH3/2015 (CK/QH/QH3/15), and A/goose/Qinghai/QH6/2015 (GS/QH/QH6/15) belonged to clade 2.3.2.1c. Further analysis of the internal genes of the isolates found that the PB2 gene of EN/QH/1/13 had 99.6% nucleotide identity with that of A/tiger/Jiangsu/1/2013 (H5N1), which clustered into an independent branch with PB2 from multiple subtypes. PB2, PB1, and M genes of CK/QH/QH3/15 were from H9N2, suggesting it was a reassortant of H5N1 and H9N2. Animal studies of three selected viruses revealed that CK/GS/XG2/12, EN/QH/1/13, and CK/QH/QH3/15 were highly lethal to chickens, with intravenous pathogenicity indexes (IVPIs) of 2.97, 2.81, and 3.00, respectively, and systemically replicated in chickens. In a mouse study, three selected H5N1 viruses were highly pathogenic to mice and readily replicated in the lungs, nasal turbinates, kidneys, spleens, and brains. Therefore, isolates in this study appear to be novel reassortants that were circulating at the interface of wild and domestic birds around Qinghai Lake and are lethal to chickens and mice. These data suggest that more extensive surveillance should be implemented, and matched vaccines should be chosen for the domestic birds in this area.


Asunto(s)
Animales Domésticos/virología , Subtipo H5N1 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Lagos/virología , Células A549 , Animales , Pollos/virología , China/epidemiología , Perros , Patos/virología , Evolución Molecular , Femenino , Humanos , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Subtipo H9N2 del Virus de la Influenza A/genética , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Gripe Aviar/mortalidad , Gripe Aviar/virología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Filogenia , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Replicación Viral
8.
Front Microbiol ; 10: 2955, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921093

RESUMEN

Influenza A viruses (IAVs) continuously challenge the poultry industry and human health. Studies of IAVs are still hampered by the availability of suitable animal models. Chinese tree shrews (Tupaia belangeri chinensis) are closely related to primates physiologically and genetically, which make them a potential animal model for human diseases. In this study, we comprehensively evaluated infectivity and transmissibility in Chinese tree shrews by using pandemic H1N1 (A/Sichuan/1/2009, pdmH1N1), avian-origin H5N1 (A/Chicken/Gansu/2/2012, H5N1) and early human-origin H7N9 (A/Suzhou/SZ19/2014, H7N9) IAVs. We found that these viruses replicated efficiently in primary tree shrew cells and tree shrews without prior adaption. Pathological lesions in the lungs of the infected tree shrews were severe on day 3 post-inoculation, although clinic symptoms were self-limiting. The pdmH1N1 and H7N9 viruses, but not the H5N1 virus, transmitted among tree shrews by direct contact. Interestingly, we also observed that unadapted H7N9 virus could transmit from tree shrews to naïve guinea pigs. Virus-inoculated tree shrews generated a strong humoral immune response and were protected from challenge with homologous virus. Taken together, our findings suggest the Chinese tree shrew would be a useful mammalian model to study the pathogenesis and transmission of IAVs.

9.
Virology ; 496: 131-137, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27310357

RESUMEN

We have previously reported that phosphoinositide-specific phospholipase γ1 (PLC-γ1) signaling is activated by influenza virus H1N1 infection and mediates efficient viral entry in human epithelial cells. In this study, we show that H1N1 also activates PLCγ-1 signaling in human promonocytic cell line -derived macrophages. Surprisingly, the activated PLCγ-1 signaling is not important for viral replication in macrophages, but is involved in the virus-induced inflammatory responses. PLC-γ1-specific inhibitor U73122 strongly inhibits the H1N1 virus-induced NF-κB signaling, blocking the up-regulation of TNF-α, IL-6, MIP-1α, and reactive oxidative species. In a positive feedback loop, IL-1ß and TNF-α activate the PLCγ-1 signaling in both epithelial and macrophage cell lines. In summary, we have shown for the first time that the PLCγ-1 signaling plays an important role in the H1N1-induced inflammatory responses. Our study suggests that targeting the PLCγ-1 signaling is a potential antiviral therapy against H1N1 by inhibiting both viral replication and excessive inflammation.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/virología , Fosfolipasa C gamma/metabolismo , Animales , Línea Celular , Citocinas/metabolismo , Estrenos/farmacología , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/virología , FN-kappa B/metabolismo , Fosfolipasa C gamma/antagonistas & inhibidores , Pirrolidinonas/farmacología , Especies Reactivas de Oxígeno , Transducción de Señal/efectos de los fármacos , Células U937
10.
Sci Rep ; 6: 25549, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-27151540

RESUMEN

Multiple infections of avian influenza viruses (AIVs) in poultry or wild birds contribute to the continued evolution of H5 subtype viruses in nature and provide potential recombination of AIVs of different origins. In this study, we carried out surveillance of AIVs in ducks, geese and the environment of a community in Hunan province, China, from 2014-2015. We isolated multiple co-circulated AIVs including H3N2, H3N8, and H5N6, and, most importantly, a novel reassortant: H3N6. Phylogenetic analyses suggest that H3N6 is highly likely derived from H5N6, which has recently been shown to have zoonotic potential with human infections. Studies with mammalian cell lines and a mouse model indicate that four selected AIVs of duck or goose origin can infect MDCK and A549 cells but have low pathogenicity in mice. We propose that a potential co-circulation of multiple subtypes including H5N6 in local area may result in the production of novel subtypes such as H3N6 by gene reassortment.


Asunto(s)
Microbiología Ambiental , Virus de la Influenza A/clasificación , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/epidemiología , Gripe Aviar/virología , Virus Reordenados/clasificación , Virus Reordenados/aislamiento & purificación , Animales , Línea Celular , China/epidemiología , Modelos Animales de Enfermedad , Perros , Patos , Evolución Molecular , Gansos , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Ratones , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Filogenia , Virus Reordenados/genética , Virus Reordenados/patogenicidad , Análisis de Secuencia de ADN , Virulencia
11.
Infect Genet Evol ; 36: 462-466, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26296602

RESUMEN

Novel H5N6 influenza A viruses have infected birds and human beings and caused four human clinical cases in China since 2014. The pig, as a mixing vessel, plays an important role for influenza virus reassortment and transmission. Towards this, routine surveillance for swine influenza in Guangdong province was conducted in 2014. In this study, we reported the biological characterization of two H5N6 influenza viruses isolated from healthy pigs in Guangdong province. Genetic analysis indicates that the two viruses are reassortants of 2.3.4.4 H5N1 and H6N6 avian influenza viruses with a high similarity to duck and human H5N6 influenza viruses isolated from Guangdong province. The data from chicken and mouse experiments show that the viruses are highly pathogenic in chickens and result in a systemic infection, and replicate in the mouse lung accompanying with a clinical inflammatory pathology. The results of the study demonstrate that the two H5N6 influenza viruses isolated from swine are the avian-originated viruses and have not adapted to swine population yet. However, they might keep evolving and pose a potential risk to public health and the continued surveillance of swine influenza should be strengthened.


Asunto(s)
Genotipo , Virus de la Influenza A/clasificación , Virus de la Influenza A/fisiología , Gripe Aviar/virología , Infecciones por Orthomyxoviridae/virología , Virus Reordenados , Animales , Pollos , Femenino , Genes Virales , Virus de la Influenza A/aislamiento & purificación , Ratones , Infecciones por Orthomyxoviridae/patología , Filogenia , Porcinos , Factores de Virulencia/genética , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda