Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 869
Filtrar
1.
Infect Immun ; 92(5): e0045323, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38602405

RESUMEN

Nontypeable Haemophilus influenzae (NTHi) is a major otitis media (OM) pathogen, with colonization a prerequisite for disease development. Most acute OM is in children <5 years old, with recurrent and chronic OM impacting hearing and learning. Therapies to prevent NTHi colonization and/or disease are needed, especially for young children. Respiratory viruses are implicated in driving the development of bacterial OM in children. We have developed an infant mouse model of influenza-driven NTHi OM, as a preclinical tool for the evaluation of safety and efficacy of clinical therapies to prevent NTHi colonization and the development of OM. In this model, 100% of infant BALB/cARC mice were colonized with NTHi, and all developed NTHi OM. Influenza A virus (IAV) facilitated the establishment of dense (1 × 105 CFU/mL) and long-lasting (6 days) NTHi colonization. IAV was essential for the development of NTHi OM, with 100% of mice in the IAV/NTHi group developing NTHi OM compared with 8% of mice in the NTHi only group. Histological analysis and cytokine measurements revealed that the inflammation observed in the middle ear of the infant mice with OM reflected inflammation observed in children with OM. We have developed the first infant mouse model of NTHi colonization and OM. This ascension model uses influenza-driven establishment of OM and reflects the clinical pathology of bacterial OM developing after a respiratory virus infection. This model provides a valuable tool for testing therapies to prevent or treat NTHi colonization and disease in young children.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Haemophilus , Haemophilus influenzae , Virus de la Influenza A , Otitis Media , Animales , Otitis Media/microbiología , Haemophilus influenzae/crecimiento & desarrollo , Haemophilus influenzae/patogenicidad , Haemophilus influenzae/fisiología , Infecciones por Haemophilus/microbiología , Ratones , Virus de la Influenza A/patogenicidad , Virus de la Influenza A/crecimiento & desarrollo , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/complicaciones , Humanos , Animales Recién Nacidos
2.
J Virol ; 98(4): e0010224, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38470058

RESUMEN

The transmembrane serine protease 2 (TMPRSS2) activates the outer structural proteins of a number of respiratory viruses including influenza A virus (IAV), parainfluenza viruses, and various coronaviruses for membrane fusion. Previous studies showed that TMPRSS2 interacts with the carboxypeptidase angiotensin-converting enzyme 2 (ACE2), a cell surface protein that serves as an entry receptor for some coronaviruses. Here, by using protease activity assays, we determine that ACE2 increases the enzymatic activity of TMPRSS2 in a non-catalytic manner. Furthermore, we demonstrate that ACE2 knockdown inhibits TMPRSS2-mediated cleavage of IAV hemagglutinin (HA) in Calu-3 human airway cells and suppresses virus titers 100- to 1.000-fold. Transient expression of ACE2 in ACE2-deficient cells increased TMPRSS2-mediated HA cleavage and IAV replication. ACE2 knockdown also reduced titers of MERS-CoV and prevented S cleavage by TMPRSS2 in Calu-3 cells. By contrast, proteolytic activation and multicycle replication of IAV with multibasic HA cleavage site typically cleaved by furin were not affected by ACE2 knockdown. Co-immunoprecipitation analysis revealed that ACE2-TMPRSS2 interaction requires the enzymatic activity of TMPRSS2 and the carboxypeptidase domain of ACE2. Together, our data identify ACE2 as a new co-factor or stabilizer of TMPRSS2 activity and as a novel host cell factor involved in proteolytic activation and spread of IAV in human airway cells. Furthermore, our data indicate that ACE2 is involved in the TMPRSS2-catalyzed activation of additional respiratory viruses including MERS-CoV.IMPORTANCEProteolytic cleavage of viral envelope proteins by host cell proteases is essential for the infectivity of many viruses and relevant proteases provide promising drug targets. The transmembrane serine protease 2 (TMPRSS2) has been identified as a major activating protease of several respiratory viruses, including influenza A virus. TMPRSS2 was previously shown to interact with angiotensin-converting enzyme 2 (ACE2). Here, we report the mechanistic details of this interaction. We demonstrate that ACE2 increases or stabilizes the enzymatic activity of TMPRSS2. Furthermore, we describe ACE2 involvement in TMPRSS2-catalyzed cleavage of the influenza A virus hemagglutinin and MERS-CoV spike protein in human airway cells. These findings expand our knowledge of the activation of respiratory viruses by TMPRSS2 and the host cell factors involved. In addition, our results could help to elucidate a physiological role for TMPRSS2.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Virus de la Influenza A , Pulmón , Proteolisis , Serina Endopeptidasas , Animales , Perros , Humanos , Enzima Convertidora de Angiotensina 2/deficiencia , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Biocatálisis , Línea Celular , Furina/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/metabolismo , Pulmón/citología , Pulmón/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/metabolismo , Unión Proteica , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Replicación Viral
3.
J Virol ; 98(2): e0149423, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38294251

RESUMEN

Influenza B viruses (IBV) cocirculate with influenza A viruses (IAV) and cause periodic epidemics of disease, yet antibody and cellular responses following IBV infection are less well understood. Using the ferret model for antisera generation for influenza surveillance purposes, IAV resulted in robust antibody responses following infection, whereas IBV required an additional booster dose, over 85% of the time, to generate equivalent antibody titers. In this study, we utilized primary differentiated ferret nasal epithelial cells (FNECs) which were inoculated with IAV and IBV to study differences in innate immune responses which may result in differences in adaptive immune responses in the host. FNECs were inoculated with IAV (H1N1pdm09 and H3N2 subtypes) or IBV (B/Victoria and B/Yamagata lineages) and assessed for 72 h. Cells were analyzed for gene expression by quantitative real-time PCR, and apical and basolateral supernatants were assessed for virus kinetics and interferon (IFN), respectively. Similar virus kinetics were observed with IAV and IBV in FNECs. A comparison of gene expression and protein secretion profiles demonstrated that IBV-inoculated FNEC expressed delayed type-I/II IFN responses and reduced type-III IFN secretion compared to IAV-inoculated cells. Concurrently, gene expression of Thymic Stromal Lymphopoietin (TSLP), a type-III IFN-induced gene that enhances adaptive immune responses, was significantly downregulated in IBV-inoculated FNECs. Significant differences in other proinflammatory and adaptive genes were suppressed and delayed following IBV inoculation. Following IBV infection, ex vivo cell cultures derived from the ferret upper respiratory tract exhibited reduced and delayed innate responses which may contribute to reduced antibody responses in vivo.IMPORTANCEInfluenza B viruses (IBV) represent nearly one-quarter of all human influenza cases and are responsible for significant clinical and socioeconomic impacts but do not pose the same pandemic risks as influenza A viruses (IAV) and have thus received much less attention. IBV accounts for greater severity and deaths in children, and vaccine efficacy remains low. The ferret can be readily infected with human clinical isolates and demonstrates a similar course of disease and immune responses. IBV, however, generates lower antibodies in ferrets than IAV following the challenge. To determine whether differences in initial innate responses following infection may affect the development of robust adaptive immune responses, ferret respiratory tract cells were isolated, infected with IAV/IBV, and compared. Understanding the differences in the initial innate immune responses to IAV and IBV may be important in the development of more effective vaccines and interventions to generate more robust protective immune responses.


Asunto(s)
Inmunidad Adaptativa , Células Epiteliales , Hurones , Inmunidad Innata , Virus de la Influenza A , Virus de la Influenza B , Interferones , Mucosa Nasal , Animales , Niño , Humanos , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/virología , Hurones/inmunología , Hurones/virología , Virus de la Influenza A/clasificación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Virus de la Influenza B/clasificación , Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/inmunología , Vacunas contra la Influenza , Gripe Humana/virología , Interferones/inmunología , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Linfopoyetina del Estroma Tímico/genética , Linfopoyetina del Estroma Tímico/inmunología , Células Cultivadas
4.
Nature ; 619(7969): 338-347, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37380775

RESUMEN

Spillover events of avian influenza A viruses (IAVs) to humans could represent the first step in a future pandemic1. Several factors that limit the transmission and replication of avian IAVs in mammals have been identified. There are several gaps in our understanding to predict which virus lineages are more likely to cross the species barrier and cause disease in humans1. Here, we identified human BTN3A3 (butyrophilin subfamily 3 member A3)2 as a potent inhibitor of avian IAVs but not human IAVs. We determined that BTN3A3 is expressed in human airways and its antiviral activity evolved in primates. We show that BTN3A3 restriction acts primarily at the early stages of the virus life cycle by inhibiting avian IAV RNA replication. We identified residue 313 in the viral nucleoprotein (NP) as the genetic determinant of BTN3A3 sensitivity (313F or, rarely, 313L in avian viruses) or evasion (313Y or 313V in human viruses). However, avian IAV serotypes, such as H7 and H9, that spilled over into humans also evade BTN3A3 restriction. In these cases, BTN3A3 evasion is due to substitutions (N, H or Q) in NP residue 52 that is adjacent to residue 313 in the NP structure3. Thus, sensitivity or resistance to BTN3A3 is another factor to consider in the risk assessment of the zoonotic potential of avian influenza viruses.


Asunto(s)
Aves , Interacciones Microbiota-Huesped , Virus de la Influenza A , Gripe Aviar , Gripe Humana , Zoonosis Virales , Animales , Humanos , Aves/virología , Virus de la Influenza A/clasificación , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/transmisión , Gripe Aviar/virología , Gripe Humana/prevención & control , Gripe Humana/transmisión , Gripe Humana/virología , Primates , Sistema Respiratorio/metabolismo , Sistema Respiratorio/virología , Medición de Riesgo , Zoonosis Virales/prevención & control , Zoonosis Virales/transmisión , Zoonosis Virales/virología , Replicación Viral
5.
Onderstepoort J Vet Res ; 89(1): e1-e6, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36546513

RESUMEN

Influenza A viruses (IAVs) are typically isolated and cultured by successive passages using 9- to 11-day-old embryonated chicken eggs (ECEs) and in 14-day old ECEs for virus mutational studies. Real-time reverse transcription-polymerase chain reaction tests (RT-PCRs) are commonly used for IAV diagnosis, but virus isolation remains invaluable in terms of its high sensitivity, providing viable isolates for further studies and the ability to distinguish between viable and nonviable virus. Efforts at isolating ostrich-origin IAVs from RT-PCR positive specimens using ECEs have often been unsuccessful, raising the possibility of a species bottleneck, whereby ostrich-adapted IAVs may not readily infect and replicate in ECEs, yet the capacity of an ostrich embryo to support the replication of influenza viruses has not been previously demonstrated. This study describes an optimised method for H5 and H7 subtype IAV isolation and propagation in 28-day old embryonated ostrich eggs (EOEs), the biological equivalent of 14-day old ECEs. The viability of EOEs transported from breeding sites could be maximised by pre-incubating the eggs for 12 to 14 days prior to long-distance transportation. This method applied to studies for ostrich-adapted virus isolation and in ovo studies will enable better understanding of the virus-host interaction in ostriches and the emergence of potentially zoonotic diseases.


Asunto(s)
Virus de la Influenza A , Struthioniformes , Animales , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/aislamiento & purificación , Gripe Aviar/virología , Cigoto/virología , Virología/métodos
6.
J Virol ; 96(15): e0078622, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35861516

RESUMEN

The M1 of influenza A virus (IAV) is important for the virus life cycle, especially for the assembly and budding of viruses, which is a multistep process that requires host factors. Identifying novel host proteins that interact with M1 and understanding their functions in IAV replication are of great interest in antiviral drug development. In this study, we identified 19 host proteins in DF1 cells suspected to interact with the M1 protein of an H5N6 virus through immunoprecipitation (IP)/mass spectrometry. Among them, PSMD12, a 26S proteasome regulatory subunit, was shown to interact with influenza M1, acting as a positive host factor in IAV replication in avian and human cells. The data showed that PSMD12 promoted K63-linked ubiquitination of M1 at the K102 site. H5N6 and PR8 with an M1-K102 site mutant displayed a significantly weaker replication ability than the wild-type viruses. Mechanistically, PSMD12 promoted M1-M2 virus-like particle (VLP) release, and an M1-K102 mutation disrupted the formation of supernatant M1-M2 VLPs. An H5N6 M1-K102 site mutation or knockdown PSMD12 disrupted the budding release of the virus in chicken embryo fibroblast (CEF) cells, which was confirmed by transmission electron microscopy. Further study confirmed that M1-K102 site mutation significantly affected the virulence of H5N6 and PR8 viruses in mice. In conclusion, we report the novel host factor PSMD12 which affects the replication of influenza virus by mediating K63-linked ubiquitination of M1 at K102. These findings provide novel insight into the interactions between IAV and host cells, while suggesting an important target for anti-influenza virus drug research. IMPORTANCE M1 is proposed to play multiple biologically important roles in the life cycle of IAV, which relies largely on host factors. This study is the first one to identify that PSMD12 interacts with M1, mediates K63-linked ubiquitination of M1 at the K102 site, and thus positively regulates influenza virus proliferation. PSMD12 promoted M1-M2 VLP egress, and an M1-K102 mutation affected the M1-M2 VLP formation. Furthermore, we demonstrate the importance of this site to the morphology and budding of influenza viruses by obtaining mutant viruses, and the M1 ubiquitination regulator PSMD12 has a similar function to the M1 K102 mutation in regulating virus release and virus morphology. Additionally, we confirm the reduced virulence of H5N6 and PR8 (H1N1) viruses carrying the M1-K102 site mutation in mice. These findings provide novel insights into IAV interactions with host cells and suggest a valid and highly conserved candidate target for antiviral drug development.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Influenza A , Complejo de la Endopetidasa Proteasomal , Ubiquitinación , Proteínas de la Matriz Viral , Replicación Viral , Animales , Antivirales , Línea Celular , Embrión de Pollo , Fibroblastos , Humanos , Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/metabolismo , Virus de la Influenza A/patogenicidad , Ratones , Mutación , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Virulencia/genética
7.
J Virol ; 96(15): e0041622, 2022 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-35862707

RESUMEN

Avian or human influenza A viruses bind preferentially to avian- or human-type sialic acid receptors, respectively, indicating that receptor tropism is an important factor for determining the viral host range. However, there are currently no reliable methods for analyzing receptor tropism biologically under physiological conditions. In this study, we established a novel system using MDCK cells with avian- or human-type sialic acid receptors and with both sialic acid receptors knocked out (KO). When we examined the replication of human and avian influenza viruses in these KO cells, we observed unique viral receptor tropism that could not be detected using a conventional solid-phase sialylglycan binding assay, which directly assesses physical binding between the virus and sialic acids. Furthermore, we serially passaged an engineered avian-derived H4N5 influenza virus, whose PB2 gene was deleted, in avian-type receptor KO cells stably expressing PB2 to select a mutant with enhanced replication in KO cells; however, its binding to human-type sialylglycan was undetectable using the solid-phase binding assay. These data indicate that a panel of sialic acid receptor KO cells could be a useful tool for determining the biological receptor tropism of influenza A viruses. Moreover, the PB2KO virus experimental system could help to safely and efficiently identify the mutations required for avian influenza viruses to adapt to human cells that could trigger a new influenza pandemic. IMPORTANCE The acquisition of mutations that allow avian influenza A virus hemagglutinins to recognize human-type receptors is mandatory for the transmission of avian viruses to humans, which could lead to a pandemic. In this study, we established a novel system using a set of genetically engineered MDCK cells with knocked out sialic acid receptors to biologically evaluate the receptor tropism for influenza A viruses. Using this system, we observed unique receptor tropism in several virus strains that was undetectable using conventional solid-phase binding assays that measure physical binding between the virus and artificially synthesized sialylglycans. This study contributes to elucidation of the relationship between the physical binding of virus and receptor and viral infectivity. Furthermore, the system using sialic acid knockout cells could provide a useful tool to explore the sialic acid-independent entry mechanism. In addition, our system could be safely used to identify mutations that could acquire human-type receptor tropism.


Asunto(s)
Virus de la Influenza A , Ácido N-Acetilneuramínico , Receptores de Superficie Celular , Receptores Virales , Tropismo Viral , Internalización del Virus , Animales , Aves/virología , Perros , Técnicas de Inactivación de Genes , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/metabolismo , Gripe Aviar/virología , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Ácido N-Acetilneuramínico/metabolismo , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Virales/química , Receptores Virales/genética , Receptores Virales/metabolismo
8.
Biochem Biophys Res Commun ; 587: 63-68, 2022 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-34864548

RESUMEN

The concept of tRNA recycling has recently emerged from the studies of ribosome-associated quality control. Therein tRNase ZS removes the 2', 3'>p from the ANKZF1-cleaved tRNA and the subsequent TRNT1 action re-generates the intact tRNA. To know the roles of the tRNA recycling in vivo, we investigated how viral infection affects the tRNA recycling system by analyzing the mRNA levels of tRNase ZS and TRNT1. We found that both genes in HeLa cells are upregulated in response to infection of Theiler's mouse encephalitis virus but not to that of an influenza A virus. Upregulation was also observed in cells infected with encephalomyocarditis virus with reduced efficiency. The levels of the IFN-ß mRNA appeared to positively correlate with those of the tRNase ZS and TRNT1 mRNAs. The tRNase ZS gene may be regulated post-transcriptionally in the cells infected with Theiler's mouse encephalitis virus.


Asunto(s)
Endorribonucleasas/genética , Interacciones Huésped-Patógeno/genética , Nucleotidiltransferasas/genética , Procesamiento Postranscripcional del ARN , ARN de Transferencia/genética , Theilovirus/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Virus de la Encefalomiocarditis/genética , Virus de la Encefalomiocarditis/crecimiento & desarrollo , Virus de la Encefalomiocarditis/metabolismo , Endorribonucleasas/metabolismo , Células HeLa , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Nucleotidiltransferasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Theilovirus/crecimiento & desarrollo , Theilovirus/metabolismo , Carga Viral
9.
Microbiol Spectr ; 9(3): e0109121, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34935417

RESUMEN

Chemical methods of virus inactivation are used routinely to prevent viral transmission in both a personal hygiene capacity but also in at-risk environments like hospitals. Several virucidal products exist, including hand soaps, gels, and surface disinfectants. Resin acids, which can be derived from tall oil, produced from trees, have been shown to exhibit antibacterial activity. However, whether these products or their derivatives have virucidal activity is unknown. Here, we assessed the capacity of rosin soap to inactivate a panel of pathogenic mammalian viruses in vitro. We show that rosin soap can inactivate human enveloped viruses: influenza A virus (IAV), respiratory syncytial virus, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). For IAV, rosin soap could provide a 100,000-fold reduction in infectivity. However, rosin soap failed to affect the nonenveloped encephalomyocarditis virus (EMCV). The inhibitory effect of rosin soap against IAV infectivity was dependent on its concentration but not on the incubation time or temperature. In all, we demonstrate a novel chemical inactivation method against enveloped viruses, which could be of use for preventing virus infections in certain settings. IMPORTANCE Viruses remain a significant cause of human disease and death, most notably illustrated through the current coronavirus disease 2019 (COVID-19) pandemic. Control of virus infection continues to pose a significant global health challenge to the human population. Viruses can spread through multiple routes, including via environmental and surface contamination, where viruses can remain infectious for days. Methods for inactivating viruses on such surfaces may help mitigate infection. Here, we present evidence identifying a novel virucidal product, rosin soap, which is produced from tall oil from coniferous trees. Rosin soap was able to rapidly and potently inactivate influenza virus and other enveloped viruses.


Asunto(s)
Antivirales/farmacología , Resinas de Plantas/farmacología , Jabones/farmacología , Antivirales/análisis , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/crecimiento & desarrollo , Aceites de Plantas/análisis , Aceites de Plantas/farmacología , Resinas de Plantas/análisis , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , Jabones/análisis , Inactivación de Virus/efectos de los fármacos
10.
Nutrients ; 13(11)2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34836165

RESUMEN

It is difficult to match annual vaccines against the exact influenza strain that is spreading in any given flu season. Owing to the emergence of drug-resistant viral strains, new approaches for treating influenza are needed. Euglena gracilis (hereinafter Euglena), microalga, used as functional foods and supplements, have been shown to alleviate symptoms of influenza virus infection in mice. However, the mechanism underlying the inhibitory action of microalgae against the influenza virus is unknown. Here, we aimed to study the antiviral activity of Euglena extract against the influenza virus and the underlying action mechanism using Madin-Darby canine kidney (MDCK) cells. Euglena extract strongly inhibited infection by all influenza virus strains examined, including those resistant to the anti-influenza drugs oseltamivir and amantadine. A time-of-addition assay revealed that Euglena extract did not affect the cycle of virus replication, and cell pretreatment or prolonged treatment of infected cells reduced the virus titer. Thus, Euglena extract may activate the host cell defense mechanisms, rather than directly acting on the influenza virus. Moreover, various minerals, mainly zinc, in Euglena extract were found to be involved in the antiviral activity of the extract. In conclusion, Euglena extract could be a potent agent for preventing and treating influenza.


Asunto(s)
Antivirales , Mezclas Complejas/farmacología , Euglena , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza B/crecimiento & desarrollo , Animales , Perros , Euglena/química , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza B/efectos de los fármacos , Células de Riñón Canino Madin Darby , Replicación Viral/efectos de los fármacos , Zinc/análisis , Acetato de Zinc/farmacología
11.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830321

RESUMEN

Conformational conversion of the cellular isoform of prion protein, PrPC, into the abnormally folded, amyloidogenic isoform, PrPSc, is an underlying pathogenic mechanism in prion diseases. The diseases manifest as sporadic, hereditary, and acquired disorders. Etiological mechanisms driving the conversion of PrPC into PrPSc are unknown in sporadic prion diseases, while prion infection and specific mutations in the PrP gene are known to cause the conversion of PrPC into PrPSc in acquired and hereditary prion diseases, respectively. We recently reported that a neurotropic strain of influenza A virus (IAV) induced the conversion of PrPC into PrPSc as well as formation of infectious prions in mouse neuroblastoma cells after infection, suggesting the causative role of the neuronal infection of IAV in sporadic prion diseases. Here, we discuss the conversion mechanism of PrPC into PrPSc in different types of prion diseases, by presenting our findings of the IAV infection-induced conversion of PrPC into PrPSc and by reviewing the so far reported transgenic animal models of hereditary prion diseases and the reverse genetic studies, which have revealed the structure-function relationship for PrPC to convert into PrPSc after prion infection.


Asunto(s)
Síndrome de Creutzfeldt-Jakob/genética , Enfermedad de Gerstmann-Straussler-Scheinker/genética , Gripe Humana/genética , Insomnio Familiar Fatal/genética , Proteínas PrPC/genética , Proteínas PrPSc/genética , Proteínas Priónicas/genética , Animales , Línea Celular Tumoral , Síndrome de Creutzfeldt-Jakob/metabolismo , Síndrome de Creutzfeldt-Jakob/patología , Síndrome de Creutzfeldt-Jakob/virología , Enfermedad de Gerstmann-Straussler-Scheinker/metabolismo , Enfermedad de Gerstmann-Straussler-Scheinker/patología , Enfermedad de Gerstmann-Straussler-Scheinker/virología , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/patogenicidad , Gripe Humana/metabolismo , Gripe Humana/patología , Gripe Humana/virología , Insomnio Familiar Fatal/metabolismo , Insomnio Familiar Fatal/patología , Insomnio Familiar Fatal/virología , Ratones , Ratones Transgénicos , Mutación , Neuronas/metabolismo , Neuronas/patología , Neuronas/virología , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/metabolismo , Proteínas Priónicas/química , Proteínas Priónicas/metabolismo , Conformación Proteica , Genética Inversa/métodos
12.
Emerg Microbes Infect ; 10(1): 1896-1907, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34498543

RESUMEN

BACKGROUND: There is great interest in understanding the viral genomic predictors of phenotypic traits that allow influenza A viruses to adapt to or become more virulent in different hosts. Machine learning techniques have demonstrated promise in addressing this critical need for other pathogens because the underlying algorithms are especially well equipped to uncover complex patterns in large datasets and produce generalizable predictions for new data. As the body of research where these techniques are applied for influenza A virus phenotype prediction continues to grow, it is useful to consider the strengths and weaknesses of these approaches to understand what has prevented these models from seeing widespread use by surveillance laboratories and to identify gaps that are underexplored with this technology. METHODS AND RESULTS: We present a systematic review of English literature published through 15 April 2021 of studies employing machine learning methods to generate predictions of influenza A virus phenotypes from genomic or proteomic input. Forty-nine studies were included in this review, spanning the topics of host discrimination, human adaptability, subtype and clade assignment, pandemic lineage assignment, characteristics of infection, and antiviral drug resistance. CONCLUSIONS: Our findings suggest that biases in model design and a dearth of wet laboratory follow-up may explain why these models often go underused. We, therefore, offer guidance to overcome these limitations, aid in improving predictive models of previously studied influenza A virus phenotypes, and extend those models to unexplored phenotypes in the ultimate pursuit of tools to enable the characterization of virus isolates across surveillance laboratories.


Asunto(s)
Algoritmos , Farmacorresistencia Viral/genética , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/genética , Aprendizaje Automático , Genoma Viral/genética , Genotipo , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Fenotipo
13.
Biochem Biophys Res Commun ; 575: 36-41, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34455219

RESUMEN

Air spaces and material surfaces in a pathogen-contaminated environment can often be a source of infection to humans, and disinfection has become a common intervention focused on reducing the contamination levels. In this study, we examined the efficacy of SAIW, a unique electrolyzed water with chlorine-free, high pH, high concentration of dissolved hydrogen, and low oxygen reduction potential, for the inactivation of several viruses and bacteria. Infectivity assays revealed that initial viral titers of enveloped and non-enveloped viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza A virus, herpes simplex virus type 1, human coronavirus, feline calicivirus, and canine parvovirus, were reduced by 2.9- to 5.5-log10 within 30 s of SAIW exposure. Similarly, the culturability of three Gram-negative bacteria (Escherichia coli, Salmonella, and Legionella) dropped down by 1.9- to 4.9-log10 within 30 s of SAIW treatment. Mechanistically, treatment with SAIW was found to significantly decrease the binding and subsequent entry efficiencies of SARS-CoV-2 on Vero cells. Finally, we showed that this chlorine-free electrolytic ion water had no acute inhalation toxicity in mice, demonstrating that SAIW holds promise for a safer antiviral and antibacterial disinfectant.


Asunto(s)
Antiinfecciosos/farmacología , Desinfectantes/farmacología , Desinfección/métodos , SARS-CoV-2/efectos de los fármacos , Inactivación de Virus/efectos de los fármacos , Agua/farmacología , Animales , Calicivirus Felino/efectos de los fármacos , Calicivirus Felino/crecimiento & desarrollo , Chlorocebus aethiops , Recuento de Colonia Microbiana , Electrólisis , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/crecimiento & desarrollo , Humanos , Concentración de Iones de Hidrógeno , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/crecimiento & desarrollo , Legionella/efectos de los fármacos , Legionella/crecimiento & desarrollo , Ratones , Parvovirus Canino/efectos de los fármacos , Parvovirus Canino/crecimiento & desarrollo , SARS-CoV-2/crecimiento & desarrollo , Salmonella/efectos de los fármacos , Salmonella/crecimiento & desarrollo , Piel/efectos de los fármacos , Células Vero , Carga Viral
14.
Cells ; 10(8)2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34440633

RESUMEN

Viral invasion of target cells triggers an immediate intracellular host defense system aimed at preventing further propagation of the virus. Viral genomes or early products of viral replication are sensed by a number of pattern recognition receptors, leading to the synthesis and production of type I interferons (IFNs) that, in turn, activate a cascade of IFN-stimulated genes (ISGs) with antiviral functions. Among these, several members of the tripartite motif (TRIM) family are antiviral executors. This article will focus, in particular, on TRIM22 as an example of a multitarget antiviral member of the TRIM family. The antiviral activities of TRIM22 against different DNA and RNA viruses, particularly human immunodeficiency virus type 1 (HIV-1) and influenza A virus (IAV), will be discussed. TRIM22 restriction of virus replication can involve either direct interaction of TRIM22 E3 ubiquitin ligase activity with viral proteins, or indirect protein-protein interactions resulting in control of viral gene transcription, but also epigenetic effects exerted at the chromatin level.


Asunto(s)
Infecciones por VIH/virología , VIH-1/patogenicidad , Inmunidad Innata , Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Antígenos de Histocompatibilidad Menor/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Replicación Viral , Animales , Regulación Viral de la Expresión Génica , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , VIH-1/genética , VIH-1/crecimiento & desarrollo , VIH-1/inmunología , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Gripe Humana/metabolismo , Transducción de Señal
15.
J Virol ; 95(18): e0087821, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34190596

RESUMEN

The influenza A virus genome is comprised of eight single-stranded negative-sense viral RNA (vRNA) segments. Each of the eight vRNA segments contains segment-specific nonconserved noncoding regions (NCRs) of similar sequence and length in different influenza A virus strains. However, in the subtype-determinant segments, encoding hemagglutinin (HA) and neuraminidase (NA), the segment-specific noncoding regions are subtype specific, varying significantly in sequence and length at both the 3' and 5' termini among different subtypes. The significance of these subtype-specific noncoding regions (ssNCR) in the influenza virus replication cycle is not fully understood. In this study, we show that truncations of the 3'-end H1-subtype-specific noncoding region (H1-ssNCR) resulted in recombinant viruses with decreased HA vRNA replication and attenuated growth phenotype, although the vRNA replication was not affected in single-template RNP reconstitution assays. The attenuated viruses were unstable, and point mutations at nucleotide position 76 or 56 in the adjacent coding region of HA vRNA were found after serial passage. The mutations restored the HA vRNA replication and reversed the attenuated virus growth phenotype. We propose that the terminal noncoding and adjacent coding regions act synergistically to ensure optimal levels of HA vRNA replication in a multisegment environment. These results provide novel insights into the role of the 3'-end nonconserved noncoding regions and adjacent coding regions on template preference in multiple-segmented negative-strand RNA viruses. IMPORTANCE While most influenza A virus vRNA segments contain segment-specific nonconserved noncoding regions of similar length and sequence, these regions vary considerably both in length and sequence in the segments encoding HA and NA, the two major antigenic determinants of influenza A viruses. In this study, we investigated the function of the 3'-end H1-ssNCR and observed a synergistic effect between the 3'-end H1-ssNCR nucleotides and adjacent coding nucleotide(s) of the HA segment on template preference in a multisegment environment. The results unravel an additional level of complexity in the regulation of RNA replication in multiple-segmented negative-strand RNA viruses.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Virus de la Influenza A/crecimiento & desarrollo , Gripe Humana/virología , Sistemas de Lectura Abierta/genética , ARN Viral/metabolismo , Regiones no Traducidas/genética , Proteínas Virales/metabolismo , Replicación Viral , Células A549 , Secuencia de Bases , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Gripe Humana/genética , Gripe Humana/metabolismo , ARN Viral/genética , Proteínas Virales/genética , Ensamble de Virus
16.
PLoS One ; 16(3): e0248960, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33770117

RESUMEN

Novel antiviral agents for influenza, which poses a substantial threat to humans, are required. Cyclobakuchiols A and B have been isolated from Psoralea glandulosa, and cyclobakuchiol C has been isolated from P. corylifolia. The structural differences between cyclobakuchiol A and C arise due to the oxidation state of isopropyl group, and these compounds can be derived from (+)-(S)-bakuchiol, a phenolic isoprenoid compound present in P. corylifolia seeds. We previously reported that bakuchiol induces enantiospecific anti-influenza A virus activity involving nuclear factor erythroid 2-related factor 2 (Nrf2) activation. However, it remains unclear whether cyclobakuchiols A-C induce anti-influenza A virus activity. In this study, cyclobakuchiols A, B, and C along with cyclobakuchiol D, a new artificial compound derived from cyclobakuchiol B, were synthesized and examined for their anti-influenza A virus activities using Madin-Darby canine kidney cells. As a result, cyclobakuchiols A-D were found to inhibit influenza A viral infection, growth, and the reduction of expression of viral mRNAs and proteins in influenza A virus-infected cells. Additionally, these compounds markedly reduced the mRNA expression of the host cell influenza A virus-induced immune response genes, interferon-ß and myxovirus-resistant protein 1. In addition, cyclobakuchiols A-D upregulated the mRNA levels of NAD(P)H quinone oxidoreductase 1, an Nrf2-induced gene, in influenza A virus-infected cells. Notably, cyclobakuchiols A, B, and C, but not D, induced the Nrf2 activation pathway. These findings demonstrate that cyclobakuchiols have anti-influenza viral activity involving host cell oxidative stress response. In addition, our results suggest that the suitably spatial configuration between oxidized isopropyl group and phenol moiety in the structure of cyclobakuchiols is required for their effect.


Asunto(s)
Antivirales/síntesis química , Antivirales/farmacología , Técnicas de Química Sintética , Ciclohexanos/síntesis química , Ciclohexanos/farmacología , Virus de la Influenza A/efectos de los fármacos , Animales , Antivirales/química , Supervivencia Celular/efectos de los fármacos , Ciclohexanos/química , Ciclohexanos/toxicidad , Perros , Regulación Viral de la Expresión Génica/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Procesamiento de Imagen Asistido por Computador , Virus de la Influenza A/crecimiento & desarrollo , Interferón beta/genética , Interferón beta/metabolismo , Células de Riñón Canino Madin Darby , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Proteínas Virales/metabolismo
17.
PLoS One ; 16(1): e0244885, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33449947

RESUMEN

Human influenza virus infections occur annually worldwide and are associated with high morbidity and mortality. Hence, development of novel anti-influenza drugs is urgently required. Rice Power® extract developed by the Yushin Brewer Co. Ltd. is a novel aqueous extract of rice obtained via saccharization and fermentation with various microorganisms, such as Aspergillus oryzae, yeast [such as Saccharomyces cerevisiae], and lactic acid bacteria, possessing various biological and pharmacological properties. In our previous experimental screening with thirty types of Rice Power® extracts, we observed that the 30th Rice Power® (Y30) extract promoted the survival of influenza A virus-infected Madin-Darby canine kidney (MDCK) cells. Therefore, to identify compounds for the development of novel anti-influenza drugs, we aimed to investigate whether the Y30 extract exhibits anti-influenza A virus activity. In the present study, we demonstrated that the Y30 extract strongly promoted the survival of influenza A H1N1 Puerto Rico 8/34 (A/PR/8/34), California 7/09, or H3N2 Aichi 2/68 (A/Aichi/2/68) viruses-infected MDCK cells and inhibited A/PR/8/34 or A/Aichi/2/68 viruses infection and growth in the co-treatment and pre-infection experiments. The pre-treatment of Y30 extract on MDCK cells did not induce anti-influenza activity in the cell. The Y30 extract did not significantly affect influenza A virus hemagglutination, and neuraminidase and RNA-dependent RNA polymerase activities. Interestingly, the electron microscopy experiment revealed that the Y30 extract disrupts the integrity of influenza A virus particles by permeabilizing the viral membrane envelope, suggesting that Y30 extract has a direct virucidal effect against influenza A virus. Furthermore, we observed that compared to the ethyl acetate (EtOAc) extract, the water extract of Y30 extract considerably promoted the survival of cells infected with A/PR/8/34 virus. These results indicated that more anti-influenza components were present in the water extract of Y30 extract than in the EtOAc extract. Our results highlight the potential of a rice extract fermented with A. oryzae and S. cerevisiae as an anti-influenza medicine and a drug source for the development of anti-influenza compounds.


Asunto(s)
Aspergillus oryzae/metabolismo , Virus de la Influenza A/efectos de los fármacos , Oryza/química , Oryza/microbiología , Extractos Vegetales/farmacología , Saccharomyces cerevisiae/metabolismo , Agua/química , Acetatos/química , Animales , Antivirales/farmacología , Perros , Fermentación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/fisiología , Células de Riñón Canino Madin Darby , Viabilidad Microbiana/efectos de los fármacos
18.
Vet Microbiol ; 253: 108970, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33421685

RESUMEN

H3N2 canine influenza virus (CIV) has been circulating in many countries since 2008. The epidemic spread of CIV could be a concern for public health because of the close contact between humans and companion animals. In this study, we used Madin-Darby canine kidney (MDCK) cells as a coinfection model of H3N2 CIV and the pandemic (2009) H1N1 influenza virus to investigate the possibility of genetic mutation or recombination. One of the resultant progeny viruses, designated as CP15, was identified with a significantly increased replication ability. For this viral strain all segments exhibit a homology close to 100 % with its parental strain A/Canine/Jiangsu/06/2010 (JS/10), except for two site mutations K156E and R201 K which occur in the receptor-binding sites of hemagglutinin (HA) and antigen binding sites of neuraminidase (NA), respectively. Virus growth in MDCK cells showed that CP15 had a higher virus titer (more than 10 times) than JS/10. Consistent with this, CP15 exhibited extensive tissue tropism and higher viral RNA loads in the spleen, kidney and lung of mice challenged with this virus compared to JS/10. However, body weight loss and lung injure score due to CP15 infection were greatly reduced. Importantly, anti-CP15 serum antibodies could confer a high neutralization activity against JS/10. These findings indicated that the CP15 strain of high replication ability represents a promising candidate to develop an efficient CIV vaccine.


Asunto(s)
Enfermedades de los Perros/virología , Subtipo H3N2 del Virus de la Influenza A/fisiología , Virus de la Influenza A/aislamiento & purificación , Replicación Viral , Animales , Perros , Femenino , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H1N1 del Virus de la Influenza A/fisiología , Subtipo H3N2 del Virus de la Influenza A/genética , Virus de la Influenza A/genética , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/virología
19.
Elife ; 92020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33269701

RESUMEN

Viral infection induces the expression of numerous host genes that impact the outcome of infection. Here, we show that infection of human lung epithelial cells with influenza A virus (IAV) also induces a broad program of alternative splicing of host genes. Although these splicing-regulated genes are not enriched for canonical regulators of viral infection, we find that many of these genes do impact replication of IAV. Moreover, in several cases, specific inhibition of the IAV-induced splicing pattern also attenuates viral infection. We further show that approximately a quarter of the IAV-induced splicing events are regulated by hnRNP K, a host protein required for efficient splicing of the IAV M transcript in nuclear speckles. Finally, we find an increase in hnRNP K in nuclear speckles upon IAV infection, which may alter accessibility of hnRNP K for host transcripts thereby leading to a program of host splicing changes that promote IAV replication.


Asunto(s)
Empalme Alternativo , Núcleo Celular/virología , Células Epiteliales/virología , Virus de la Influenza A/crecimiento & desarrollo , Pulmón/virología , Replicación Viral , Células A549 , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Epiteliales/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Interacciones Huésped-Patógeno , Humanos , Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Pulmón/metabolismo
20.
J Virol ; 94(24)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32967956

RESUMEN

Wild aquatic birds maintain a large, genetically diverse pool of influenza A viruses (IAVs), which can be transmitted to lower mammals and, ultimately, humans. Through phenotypic analyses of viral replication efficiency, only a small set of avian IAVs were found to replicate well in epithelial cells of the swine upper respiratory tract, and these viruses were shown to infect and cause virus shedding in pigs. Such a phenotypic trait of the viral replication efficiency appears to emerge randomly and is distributed among IAVs across multiple avian species and geographic and temporal orders. It is not determined by receptor binding preference but is determined by other markers across genomic segments, such as those in the ribonucleoprotein complex. This study demonstrates that phenotypic variants of viral replication efficiency exist among avian IAVs but that only a few of these may result in viral shedding in pigs upon infection, providing opportunities for these viruses to become adapted to pigs, thus posing a higher potential risk for creating novel variants or detrimental reassortants within pig populations.IMPORTANCE Swine serve as a mixing vessel for generating pandemic strains of human influenza virus. All hemagglutinin subtypes of IAVs can infect swine; however, only sporadic cases of infection with avian IAVs are reported in domestic swine. The molecular mechanisms affecting the ability of avian IAVs to infect swine are still not fully understood. From the findings of phenotypic analyses, this study suggests that the tissue tropisms (i.e., in swine upper respiratory tracts) of avian IAVs affect their spillovers from wild birds to pigs. It was found that this phenotype is determined not by receptor binding preference but is determined by other markers across genomic segments, such as those in the ribonucleoprotein complex. In addition, our results show that such a phenotypic trait was sporadically and randomly distributed among IAVs across multiple avian species and geographic and temporal orders. This study suggests an efficient way for assessment of the risk posed by avian IAVs, such as in evaluating their potentials to be transmitted from birds to pigs.


Asunto(s)
Animales Salvajes/virología , Aves/virología , Virus de la Influenza A/genética , Gripe Aviar/transmisión , Gripe Aviar/virología , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Tropismo , Animales , Línea Celular , Células Epiteliales/virología , Células HEK293 , Hemaglutininas , Humanos , Virus de la Influenza A/crecimiento & desarrollo , Pandemias , Filogenia , Sistema Respiratorio/virología , Porcinos , Replicación Viral , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...