Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.778
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 25(12)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38928244

RESUMEN

Obesity and metabolic syndrome alter serum lipid profiles. They also increase vulnerability to viral infections and worsen the survival rate and symptoms after infection. How serum lipids affect influenza virus proliferation is unclear. Here, we investigated the effects of lysophosphatidylcholines on influenza A virus (IAV) proliferation. IAV particles in the culture medium were titrated using extraction-free quantitative PCR, and viral RNA and protein levels were assessed using real-time PCR and Western blot, respectively. RNA sequencing data were analyzed using PCA and heatmap analysis, and pathway analysis was performed using the KEGG mapper and PathIN tools. Statistical analysis was conducted using SPSS21.0. LPC treatment of THP-1 cells significantly increased IAV proliferation and IAV RNA and protein levels, and saturated LPC was more active in IAV RNA expression than unsaturated LPC was. The functional analysis of genes affected by LPCs showed that the expression of genes involved in IAV signaling, such as suppressor of cytokine signaling 3 (SOCS3), phosphoinositide-3-kinase regulatory subunit 3 (PI3K) and AKT serine/threonine kinase 3 (AKT3), Toll-like receptor 7 (TKR7), and interferon gamma receptor 1 (IFNGR1), was changed by LPC. Altered influenza A pathways were linked with MAPK and PI3K/AKT signaling. Treatment with inhibitors of MAPK or PI3K attenuated viral gene expression changes induced by LPCs. The present study shows that LPCs stimulated virus reproduction by modifying the cellular environment to one in which viruses proliferated better. This was mediated by the MAPK, JNK, and PI3K/AKT pathways. Further animal studies are needed to confirm the link between LPCs from serum or the respiratory system and IAV proliferation.


Asunto(s)
Virus de la Influenza A , Lisofosfatidilcolinas , Sistema de Señalización de MAP Quinasas , Replicación Viral , Humanos , Lisofosfatidilcolinas/farmacología , Lisofosfatidilcolinas/metabolismo , Replicación Viral/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Virus de la Influenza A/fisiología , Macrófagos/metabolismo , Macrófagos/virología , Macrófagos/efectos de los fármacos , Células THP-1 , Diferenciación Celular/efectos de los fármacos , Gripe Humana/virología , Gripe Humana/metabolismo , Transducción de Señal/efectos de los fármacos , Animales
2.
Int J Mol Sci ; 25(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38928290

RESUMEN

Influenza virus infection poses a great threat to human health globally each year. Non-coding RNAs (ncRNAs) in the human genome have been reported to participate in the replication process of the influenza virus, among which there are still many unknowns about Long Intergenic Non-Coding RNAs (LincRNAs) in the cell cycle of viral infections. Here, we observed an increased expression of Linc01615 in A549 cells upon influenza virus PR8 infection, accompanied by the successful activation of the intracellular immune system. The knockdown of Linc01615 using the shRNAs promoted the proliferation of the influenza A virus, and the intracellular immune system was inhibited, in which the expressions of IFN-ß, IL-28A, IL-29, ISG-15, MX1, and MX2 were decreased. Predictions from the catRAPID website suggested a potential interaction between Linc01615 and DHX9. Also, knocking down Linc01615 promoted influenza virus proliferation. The subsequent transcriptome sequencing results indicated a decrease in Linc01615 expression after influenza virus infection when DHX9 was knocked down. Further analysis through cross-linking immunoprecipitation and high-throughput sequencing (CLIP-seq) in HEK293 cells stably expressing DHX9 confirmed the interaction between DHX9 and Linc01615. We speculate that DHX9 may interact with Linc01615 to partake in influenza virus replication and that Linc01615 helps to activate the intracellular immune system. These findings suggest a deeper connection between DHX9 and Linc01615, which highlights the significant role of Linc01615 in the influenza virus replication process. This research provides valuable insights into understanding influenza virus replication and offers new targets for preventing influenza virus infections.


Asunto(s)
ARN Helicasas DEAD-box , Gripe Humana , ARN Largo no Codificante , Replicación Viral , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células A549 , Células HEK293 , Gripe Humana/virología , Gripe Humana/genética , Gripe Humana/inmunología , Gripe Humana/metabolismo , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Virus de la Influenza A/fisiología , Animales , Perros , Técnicas de Silenciamiento del Gen , Proteínas de Neoplasias
3.
J Transl Med ; 22(1): 570, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879538

RESUMEN

BACKGROUND: Gut microbiota (GM) have been implicated as important regulators of gastrointestinal symptom which is commonly occurred along with respiratory influenza A virus (IAV) infection, suggesting the involvement of the gut-to-lung axis in a host's response to IAV. IAV primarily destroys airway epithelium tight junctions (TJs) and consequently causes acute respiratory disease syndrome. It is known that GM and their metabolism produce an anti-influenza effect, but their role in IAV-induced airway epithelial integrity remains unknown. METHODS: A mouse model of IAV infection was established. GM were analyzed using 16S rRNA gene sequencing, and short-chain fatty acids (SCFAs) levels were measured. GM depletion and fecal microbiota transplantation (FMT) were conducted to validate the role of GM in IAV infection. A pair-feeding experiment was conducted to reveal whether IAV-induced GM dysbiosis is attributed to impaired food intake. Furthermore, human bronchial epithelial (HBE) cells were cocultured with IAV in the presence or absence of acetate. TJs function was analyzed by paracellular permeability and transepithelial electronic resistance (TEER). The mechanism of how acetate affects TJs integrity was evaluated in HBE cells transfected with G protein-coupled receptor 43 (GPR43) short hairpin RNA (shRNA). RESULTS: IAV-infected mice exhibited lower relative abundance of acetate-producing bacteria (Bacteroides, Bifidobacterium, and Akkermansia) and decreased acetate levels in gut and serum. These changes were partly caused by a decrease in food consumption (due to anorexia). GM depletion exacerbated and FMT restored IAV-induced lung inflammatory injury. IAV infection suppressed expressions of TJs (occludin, ZO-1) leading to disrupted airway epithelial barrier function as evidenced by decreased TEER and increased permeability. Acetate pretreatment activated GPR43, partially restored IAV-induced airway epithelial barrier function, and reduced inflammatory cytokines levels (TNF-α, IL-6, and IL-1ß). Such protective effects of acetate were absent in HBE cells transfected with GPR43 shRNA. Acetate and GPR43 improved TJs in an AMP-activated protein kinase (AMPK)-dependent manner. CONCLUSION: Collectively, our results demonstrated that GM protected airway TJs by modulating GPR43-AMPK signaling in IAV-induced lung injury. Therefore, improving GM dysbiosis may be a potential therapeutic target for patients with IAV infection.


Asunto(s)
Acetatos , Microbioma Gastrointestinal , Lesión Pulmonar , Infecciones por Orthomyxoviridae , Uniones Estrechas , Animales , Uniones Estrechas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Acetatos/metabolismo , Humanos , Infecciones por Orthomyxoviridae/complicaciones , Ratones Endogámicos C57BL , Virus de la Influenza A , Trasplante de Microbiota Fecal , Receptores Acoplados a Proteínas G/metabolismo , Ratones , Células Epiteliales/metabolismo , Disbiosis , Ácidos Grasos Volátiles/metabolismo
4.
Curr Microbiol ; 81(7): 198, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38819647

RESUMEN

Although usefulness of masks for protection against respiratory pathogens, accumulation of pathogens on their surface represents a source of infection spread. Here we prepared a plant extract-based disinfecting layer to be used in coating masks thus inhibiting their capacity to transmit airborne pathogens. To reach this, a polypropylene membrane base was coated with a layer of polyvinyledine difluoride polymer containing 500 µg/ml of Camellia sinensis (Black tea) methanolic extract. Direct inhibitory effects of C. sinensis were initially demonstrated against Staphylococcus aureus (respiratory bacteria), influenza A virus (enveloped virus) and adenovirus 1 (non-enveloped virus) which were directly proportional to both extract concentration and incubation time with the pathogen. This was later confirmed by the capacity of the supplemented membrane with the plant extract to block infectivity of the above mentioned pathogens, recorded % inhibition values were 61, 72 and 50 for S. aureus, influenza and adenovirus, respectively. In addition to the disinfecting capacity of the membrane its hydrophobic nature and pore size (154 nm) prevented penetration of dust particles or water droplets carrying respiratory pathogens. In summary, introducing this layer could protect users from infection and decrease infection risk upon handling contaminated masks surfaces.


Asunto(s)
Camellia sinensis , Máscaras , Extractos Vegetales , Staphylococcus aureus , Camellia sinensis/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , Staphylococcus aureus/efectos de los fármacos , Máscaras/virología , Desinfectantes/farmacología , Virus de la Influenza A/efectos de los fármacos , Humanos
5.
Nat Commun ; 15(1): 4500, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802391

RESUMEN

Jamaican fruit bats (Artibeus jamaicensis) naturally harbor a wide range of viruses of human relevance. These infections are typically mild in bats, suggesting unique features of their immune system. To better understand the immune response to viral infections in bats, we infected male Jamaican fruit bats with the bat-derived influenza A virus (IAV) H18N11. Using comparative single-cell RNA sequencing, we generated single-cell atlases of the Jamaican fruit bat intestine and mesentery. Gene expression profiling showed that H18N11 infection resulted in a moderate induction of interferon-stimulated genes and transcriptional activation of immune cells. H18N11 infection was predominant in various leukocytes, including macrophages, B cells, and NK/T cells. Confirming these findings, human leukocytes, particularly macrophages, were also susceptible to H18N11, highlighting the zoonotic potential of this bat-derived IAV. Our study provides insight into a natural virus-host relationship and thus serves as a fundamental resource for future in-depth characterization of bat immunology.


Asunto(s)
Quirópteros , Infecciones por Orthomyxoviridae , Análisis de la Célula Individual , Animales , Quirópteros/virología , Quirópteros/inmunología , Quirópteros/genética , Masculino , Humanos , Infecciones por Orthomyxoviridae/virología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/veterinaria , Macrófagos/inmunología , Macrófagos/virología , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Perfilación de la Expresión Génica
6.
Viruses ; 16(5)2024 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-38793579

RESUMEN

Acute respiratory infections are a major global burden in resource-limited countries, including countries in Africa. Although COVID-19 has been well studied since the pandemic emerged in Gabon, Central Africa, less attention has been paid to other respiratory viral diseases, and very little data are available. Herein, we provide the first data on the genetic diversity and detection of 18 major respiratory viruses in Gabon during the COVID-19 pandemic. Of 582 nasopharyngeal swab specimens collected from March 2020 to July 2021, which were SARS-CoV-2 negative, 156 were positive (26%) for the following viruses: enterovirus (20.3%), human rhinovirus (HRV) (4.6%), human coronavirus OC43 (1.2%), human adenovirus (0.9%), human metapneumovirus (hMPV) (0.5%), influenza A virus (IAV) (0.3%), and human parainfluenza viruses (0.5%). To determine the genetic diversity and transmission route of the viruses, phylogenetic analyses were performed using genome sequences of the detected viruses. The IAV strain detected in this study was genetically similar to strains isolated in the USA, whereas the hMPV strain belonging to the A2b subtype formed a cluster with Kenyan strains. This study provides the first complete genomic sequences of HRV, IAV, and hMPV detected in Gabon, and provides insight into the circulation of respiratory viruses in the country.


Asunto(s)
COVID-19 , Variación Genética , Filogenia , Infecciones del Sistema Respiratorio , Humanos , Gabón/epidemiología , COVID-19/epidemiología , COVID-19/virología , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/epidemiología , SARS-CoV-2/genética , SARS-CoV-2/clasificación , SARS-CoV-2/aislamiento & purificación , Masculino , Adulto , Femenino , Niño , Persona de Mediana Edad , Adolescente , Preescolar , Adulto Joven , Rhinovirus/genética , Rhinovirus/aislamiento & purificación , Rhinovirus/clasificación , Virus/genética , Virus/clasificación , Virus/aislamiento & purificación , Metapneumovirus/genética , Metapneumovirus/aislamiento & purificación , Metapneumovirus/clasificación , Genoma Viral , Nasofaringe/virología , Lactante , Anciano , Pandemias , Virus de la Influenza A/genética , Virus de la Influenza A/aislamiento & purificación , Virus de la Influenza A/clasificación
7.
PLoS Pathog ; 20(5): e1012279, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38814988

RESUMEN

The influenza A virus (IAV) consists of 8 single-stranded, negative-sense viral RNA (vRNA) segments. After infection, vRNA is transcribed, replicated, and wrapped by viral nucleoprotein (NP) to form viral ribonucleoprotein (vRNP). The transcription, replication, and nuclear export of the viral genome are regulated by the IAV protein, NS2, which is translated from spliced mRNA transcribed from viral NS vRNA. This splicing is inefficient, explaining why NS2 is present in low abundance after IAV infection. The levels of NS2 and its subsequent accumulation are thought to influence viral RNA replication and vRNP nuclear export. Here we show that NS2 is ubiquitinated at the K64 and K88 residues by K48-linked and K63-linked polyubiquitin (polyUb) chains, leading to the degradation of NS2 by the proteasome. Additionally, we show that a host deubiquitinase, OTUB1, can remove polyUb chains conjugated to NS2, thereby stabilizing NS2. Accordingly, knock down of OTUB1 by siRNA reduces the nuclear export of vRNP, and reduces the overall production of IAV. These results collectively demonstrate that the levels of NS2 in IAV-infected cells are regulated by a ubiquitination-deubiquitination system involving OTUB1 that is necessary for optimal IAV replication.


Asunto(s)
Cisteína Endopeptidasas , Virus de la Influenza A , Proteínas no Estructurales Virales , Replicación Viral , Animales , Perros , Humanos , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Enzimas Desubicuitinizantes/metabolismo , Células HEK293 , Virus de la Influenza A/metabolismo , Gripe Humana/metabolismo , Gripe Humana/virología , ARN Viral/metabolismo , ARN Viral/genética , Ubiquitinación , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética , Replicación Viral/fisiología , Línea Celular , Células Vero , Chlorocebus aethiops
8.
Virology ; 595: 110094, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38692133

RESUMEN

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


Asunto(s)
Pollos , Vacunas contra la Influenza , Gripe Aviar , MicroARNs , Fosfohidrolasa PTEN , Estrés Fisiológico , Animales , MicroARNs/genética , MicroARNs/metabolismo , Pollos/virología , Vacunas contra la Influenza/inmunología , Gripe Aviar/virología , Gripe Aviar/inmunología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Enfermedades de las Aves de Corral/virología , Enfermedades de las Aves de Corral/inmunología , Tolerancia Inmunológica , Transducción de Señal , Virus de la Influenza A/inmunología
9.
Diagn Microbiol Infect Dis ; 109(3): 116293, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38598951

RESUMEN

Data on people suspected with acute respiratory infections (ARIs) from August 2022 to December 2023 in southern China were analyzed. Following the COVID-19 pandemic, the positive detection rates of respiratory pathogens increased to 56.9%. Influenza A virus (IAV) emerged as the predominant prevalence pathogen (52.1%), followed by Mycoplasma pneumoniae (Mp: 21.2%), and SARS-CoV-2 (11.6%). Mp, IAV, and Human rhinovirus (HRV) infection were the primary etiologies of ARIs patients under age 18, accounting for 49.4%, 48.6%, and 21.7%, respectively. Mp, HRV, Respiratory syncytial virus (RSV), and Adenovirus (ADV) contributed to ARIs cases in virtually every month in this group, with Mp being particularly notable for its consistent presence and high co-infection rate (31.0%). IAV was predominant in the 19 to 59 age group (88.6%), SARS-CoV-2 was responsible for most of ARIs in the elderly group (82.5%). This study provides valuable insights into the dynamic nature of respiratory pathogens post COVID-19 era.


Asunto(s)
COVID-19 , Coinfección , Infecciones del Sistema Respiratorio , Humanos , China/epidemiología , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/microbiología , Persona de Mediana Edad , Adulto , Adolescente , Niño , Preescolar , COVID-19/epidemiología , Adulto Joven , Lactante , Anciano , Masculino , Femenino , Coinfección/epidemiología , Coinfección/microbiología , Coinfección/virología , SARS-CoV-2 , Recién Nacido , Prevalencia , Virus de la Influenza A/aislamiento & purificación , Mycoplasma pneumoniae , Anciano de 80 o más Años , Enfermedad Aguda/epidemiología
10.
J Virol Methods ; 327: 114943, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38679164

RESUMEN

We established primary porcine nasal, tracheal, and bronchial epithelial cells that recapitulate the physical and functional properties of the respiratory tract and have the ability to fully differentiate. Trans-well cultures demonstrated increased transepithelial electrical resistance over time the presence of tight junctions as demonstrated by immunohistochemistry. The nasal, tracheal, and bronchial epithelial cells developed cilia, secreted mucus, and expressed sialic acids on surface glycoproteins, the latter which are required for influenza A virus infection. Swine influenza viruses were shown to replicate efficiently in the primary epithelial cell cultures, supporting the use of these culture models to assess swine influenza and other virus infection. Primary porcine nasal, tracheal, and bronchial epithelial cell culture models enable assessment of emerging and novel influenza viruses for pandemic potential as well as mechanistic studies to understand mechanisms of infection, reassortment, and generation of novel virus. As swine are susceptible to infection with multiple viral and bacterial respiratory pathogens, these primary airway cell models may enable study of the cellular response to infection by pathogens associated with Porcine Respiratory Disease Complex.


Asunto(s)
Células Epiteliales , Animales , Porcinos , Células Epiteliales/virología , Tráquea/virología , Tráquea/citología , Bronquios/virología , Bronquios/citología , Células Cultivadas , Técnicas de Cultivo de Célula/métodos , Virus de la Influenza A/fisiología , Replicación Viral
11.
Chemistry ; 30(32): e202401108, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38567703

RESUMEN

Sialyl-Lewisx (SLex) is involved in immune regulation, human fertilization, cancer, and bacterial and viral diseases. The influence of the complex glycan structures, which can present SLex epitopes, on binding is largely unknown. We report here a chemoenzymatic strategy for the preparation of a panel of twenty-two isomeric asymmetrical tri-antennary N-glycans presenting SLex-Lex epitopes on either the MGAT4 or MGAT5 arm that include putative high-affinity ligands for E-selectin. The N-glycans were prepared starting from a sialoglycopeptide isolated from egg yolk powder and took advantage of inherent substrate preferences of glycosyltransferases and the use of 5'-diphospho-N-trifluoracetylglucosamine (UDP-GlcNHTFA) that can be transferred by branching N-acetylglucosaminyltransferases to give, after base treatment, GlcNH2-containing glycans that temporarily disable an antenna from enzymatic modification. Glycan microarray binding studies showed that E-selectin bound equally well to linear glycans and tri-antennary N-glycans presenting SLex-Lex. On the other hand, it was found that hemagglutinins (HA) of H5 influenza A viruses (IAV) preferentially bound the tri-antennary N-glycans. Furthermore, several H5 HAs preferentially bound to N-glycan presenting SLex on the MGAT4 arm. SLex is displayed in the respiratory tract of several avian species, demonstrating the relevance of investigating the binding of, among others IAVs, to complex N-glycans presenting SLex.


Asunto(s)
Selectina E , Virus de la Influenza A , Polisacáridos , Antígeno Sialil Lewis X , Polisacáridos/química , Polisacáridos/metabolismo , Virus de la Influenza A/metabolismo , Antígeno Sialil Lewis X/metabolismo , Antígeno Sialil Lewis X/química , Selectina E/metabolismo , Selectina E/química , Humanos , Oligosacáridos/química , Oligosacáridos/síntesis química , Oligosacáridos/metabolismo , Receptores Virales/metabolismo , Receptores Virales/química , Epítopos/química , Epítopos/metabolismo , Animales
12.
Nature ; 628(8009): 835-843, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38600381

RESUMEN

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Asunto(s)
Lesión Pulmonar , Necroptosis , Infecciones por Orthomyxoviridae , Inhibidores de Proteínas Quinasas , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Animales , Femenino , Humanos , Masculino , Ratones , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/virología , Células Epiteliales Alveolares/metabolismo , Virus de la Influenza A/clasificación , Virus de la Influenza A/efectos de los fármacos , Virus de la Influenza A/inmunología , Virus de la Influenza A/patogenicidad , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Lesión Pulmonar/virología , Ratones Endogámicos C57BL , Necroptosis/efectos de los fármacos , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/mortalidad , Infecciones por Orthomyxoviridae/virología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Síndrome de Dificultad Respiratoria/complicaciones , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/prevención & control , Síndrome de Dificultad Respiratoria/virología
13.
Sci Signal ; 17(831): eadg7867, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38593156

RESUMEN

Type I interferons (IFNs) are critical for the antiviral immune response, and fine-tuning type I IFN production is critical to effectively clearing viruses without causing harmful immunopathology. We showed that the transcription factor Miz1 epigenetically repressed the expression of genes encoding type I IFNs in mouse lung epithelial cells by recruiting histone deacetylase 1 (HDAC1) to the promoters of Ifna and Ifnb. Loss of function of Miz1 resulted in augmented production of these type I IFNs during influenza A virus (IAV) infection, leading to improved viral clearance in vitro and in vivo. IAV infection induced Miz1 accumulation by promoting the cullin-4B (CUL4B)-mediated ubiquitylation and degradation of the E3 ubiquitin ligase Mule (Mcl-1 ubiquitin ligase E3; also known as Huwe1 or Arf-BP1), which targets Miz1 for degradation. As a result, Miz1 accumulation limited type I IFN production and favored viral replication. This study reveals a previously unrecognized function of Miz1 in regulating antiviral defense and a potential mechanism for influenza viruses to evade host immune defense.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Interferón Tipo I , Ratones , Animales , Humanos , Virus de la Influenza A/fisiología , Ubiquitinación , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Replicación Viral , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Gripe Humana/genética , Interferones/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo
14.
J Med Virol ; 96(4): e29602, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38597349

RESUMEN

China experienced severe epidemics of multiple respiratory pathogens in 2023 after lifting "Zero-COVID" policy. The present study aims to investigate the changing circulation and infection patterns of respiratory pathogens in 2023. The 160 436 laboratory results of influenza virus and respiratory syncytial virus (RSV) from February 2020 to December 2023, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) from June 2020 to December 2023, Mycoplasma pneumoniae, adenovirus, and human rhinovirus from January 2023 to December 2023 were analyzed. We observed the alternating epidemics of SARS-CoV-2 and influenza A virus (IAV), as well as the out-of-season epidemic of RSV during the spring and summer of 2023. Cocirculation of multiple respiratory pathogens was observed during the autumn and winter of 2023. The susceptible age range of RSV in this winter epidemic (10.5, interquartile range [IQR]: 5-30) was significantly higher than previously (4, IQR: 3-34). The coinfection rate of IAV and RSV in this winter epidemic (0.695%) was significantly higher than that of the last cocirculation period (0.027%) (p < 0.001). Similar trend was also found in the coinfection of IAV and SARS-CoV-2. The present study observed the cocirculation of multiple respiratory pathogens, changing age range of susceptible population, and increasing coinfection rates during the autumn and winter of 2023, in Beijing, China.


Asunto(s)
Coinfección , Virus de la Influenza A , Gripe Humana , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio , Humanos , Estudios Retrospectivos , Infecciones del Sistema Respiratorio/epidemiología , Beijing/epidemiología , Estaciones del Año , Coinfección/epidemiología , China/epidemiología , SARS-CoV-2 , Gripe Humana/epidemiología , Infecciones por Virus Sincitial Respiratorio/epidemiología
15.
Curr Opin Virol ; 66: 101409, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38564993

RESUMEN

Influenza A virus (IAV) infections pose a global health challenge that necessitates a comprehensive understanding of the host immune response to devise effective therapeutic interventions. As monocytes and macrophages play crucial roles in host defence, inflammation, and repair, this review explores the intricate journey of these cells during and after IAV infection. First, we highlight the dynamics and functions of lung-resident macrophage populations post-IAV. Second, we review the current knowledge of recruited monocytes and monocyte-derived cells, emphasising their roles in viral clearance, inflammation, immunomodulation, and tissue repair. Third, we shed light on the consequences of IAV-induced macrophage alterations on long-term lung immunity. We conclude by underscoring current knowledge gaps and exciting prospects for future research in unravelling the complexities of macrophage responses to respiratory viral infections.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Macrófagos , Monocitos , Humanos , Monocitos/inmunología , Monocitos/virología , Gripe Humana/inmunología , Gripe Humana/virología , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Macrófagos/inmunología , Macrófagos/virología , Animales , Pulmón/virología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Inflamación/inmunología , Inflamación/virología
16.
Phytother Res ; 38(6): 3080-3121, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38613154

RESUMEN

Zoonotic diseases are major public health concerns and undeniable threats to human health. Among Zoonotic diseases, zoonotic viruses and prions are much more difficult to eradicate, as they result in higher infections and mortality rates. Several investigations have shown curcumin, the active ingredient of turmeric, to have wide spectrum properties such as anti-microbial, anti-vascular, anti-inflammatory, anti-tumor, anti-neoplastic, anti-oxidant, and immune system modulator properties. In the present study, we performed a comprehensive review of existing in silico, in vitro, and in vivo evidence on the antiviral (54 important zoonotic viruses) and anti-prion properties of curcumin and curcuminoids in PubMed, Google Scholar, Science Direct, Scopus, and Web of Science databases. Database searches yielded 13,380 results, out of which 216 studies were eligible according to inclusion criteria. Of 216 studies, 135 (62.5%), 24 (11.1%), and 19 (8.8%) were conducted on the effect of curcumin and curcuminoids against SARS-CoV-2, Influenza A virus, and dengue virus, respectively. This review suggests curcumin and curcuminoids as promising therapeutic agents against a wide range of viral zoonoses by targeting different proteins and signaling pathways.


Asunto(s)
Antivirales , Curcumina , Curcumina/farmacología , Animales , Humanos , Antivirales/farmacología , Zoonosis/tratamiento farmacológico , Zoonosis/virología , SARS-CoV-2/efectos de los fármacos , Priones/efectos de los fármacos , Virus de la Influenza A/efectos de los fármacos , Virus del Dengue/efectos de los fármacos , Tratamiento Farmacológico de COVID-19 , COVID-19/virología
17.
Front Immunol ; 15: 1378610, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38638436

RESUMEN

Influenza A virus (IAV) infection in pregnancy resembles a preeclamptic phenotype characterised by vascular dysfunction and foetal growth retardation. Given that low dose aspirin (ASA) is safe in pregnancy and is used to prevent preeclampsia, we investigated whether ASA or NO-conjugated aspirin, NCX4016, resolve vascular inflammation and function to improve offspring outcomes following IAV infection in pregnant mice. Pregnant mice were intranasally infected with a mouse adapted IAV strain (Hkx31; 104 plaque forming units) and received daily treatments with either 200µg/kg ASA or NCX4016 via oral gavage. Mice were then culled and the maternal lungs and aortas collected for qPCR analysis, and wire myography was performed on aortic rings to assess endothelial and vascular smooth muscle functionality. Pup and placentas were weighed and pup growth rates and survival assessed. IAV infected mice had an impaired endothelial dependent relaxation response to ACh in the aorta, which was prevented by ASA and NCX4016 treatment. ASA and NCX4016 treatment prevented IAV dissemination and inflammation of the aorta as well as improving the pup placental ratios in utero, survival and growth rates at post-natal day 5. Low dose ASA is safe to use during pregnancy for preeclampsia and this study demonstrates that ASA may prove a promising treatment for averting the significant vascular complications associated with influenza infection during pregnancy.


Asunto(s)
Aspirina/análogos & derivados , Virus de la Influenza A , Gripe Humana , Nitratos , Preeclampsia , Enfermedades Vasculares , Humanos , Ratones , Femenino , Embarazo , Animales , Placenta , Aspirina/farmacología , Inflamación , Aorta
18.
J Biol Chem ; 300(4): 107153, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38462163

RESUMEN

The innate immune system features a web of interacting pathways that require exquisite regulation. To identify novel nodes in this immune landscape, we conducted a gain-of-function, genome-wide CRISPR activation screen with influenza A virus. We identified both appreciated and novel antiviral genes, including Jade family PHD zinc finger 3 (JADE3) a protein involved in directing the histone acetyltransferase histone acetyltransferase binding to ORC1 complex to modify chromatin and regulate transcription. JADE3 is both necessary and sufficient to restrict influenza A virus infection. Our results suggest a distinct function for JADE3 as expression of the closely related paralogs JADE1 and JADE2 does not confer resistance to influenza A virus infection. JADE3 is required for both constitutive and inducible expression of the well-characterized antiviral gene interferon-induced transmembrane protein 3 (IFITM3). Furthermore, we find JADE3 activates the NF-kB signaling pathway, which is required for the promotion of IFITM3 expression by JADE3. Therefore, we propose JADE3 activates an antiviral genetic program involving NF-kB-dependent IFITM3 expression to restrict influenza A virus infection.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata , Proteínas de la Membrana , FN-kappa B , Proteínas Oncogénicas , Proteínas de Unión al ARN , Animales , Humanos , Sistemas CRISPR-Cas , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Células HEK293 , Inmunidad Innata/genética , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Transducción de Señal , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/inmunología
19.
Curr Pharm Des ; 30(12): 912-920, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38482627

RESUMEN

The discussion has revolved around the derivatives of amino acids and peptides containing carbocycles and their potential antiviral activity in vitro against influenza A, hepatitis C viruses, and coronavirus. Studies conducted on cell cultures reveal that aminoadamantane amino acid derivatives exhibit the capacity to hinder the replication of viruses containing viroporins. Furthermore, certain compounds demonstrate potent virucidal activity with respect to influenza A/H5N1 and hepatitis C virus particles. A conceptual framework for viroporin inhibitors has been introduced, incorporating carbocyclic motifs as membranotropic carriers in the structure, alongside a functional segment comprised of amino acids and peptides. These components correspond to the interaction with the inner surface of the channel's pore or another target protein.


Asunto(s)
Aminoácidos , Antivirales , Péptidos , Antivirales/farmacología , Antivirales/química , Antivirales/síntesis química , Humanos , Péptidos/farmacología , Péptidos/química , Péptidos/síntesis química , Aminoácidos/química , Aminoácidos/farmacología , Animales , Hepacivirus/efectos de los fármacos , Proteínas Viroporinas/antagonistas & inhibidores , Proteínas Viroporinas/metabolismo , Proteínas Viroporinas/química , Virus de la Influenza A/efectos de los fármacos
20.
Arch Virol ; 169(4): 74, 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38480558

RESUMEN

Triple motif protein 21 (TRIM21) has an antiviral function that inhibits various viral infections. However, its role in the progress of influenza A virus (IAV) infection is unclear. In this study, we investigated the role and molecular mechanism of TRIM21 in IAV infection. RT-qPCR was used to determine the level of TRIM21 mRNA, and ELISA was used to measure the levels of IFN-α, IFN-ß, IL-6, and TNF-α. The levels of the TRIM21, NP, TBK1, IRF3, p-TBK1, and p-IRF3 proteins were estimated by Western blot. The results showed that, after IAV infection, TRIM21 was upregulated in clinical patient serum and A549 cells, and this was correlated with the IFN response. Overexpression of TRIM21 reduced IAV replication and transcription in in vitro cell experiments. TRIM21 also increased IFN-α and IFN-ß levels and decreased IL-6 and TNF-α levels in A549 cells. In addition, overexpression of TRIM21 inhibited IAV-induced apoptosis. Further experiments demonstrated that TBK1-IRF3 signaling was activated by TRIM21 and was involved in the inhibitory effect of TRIM21 on virus replication. In summary, our study suggests that TRIM21 inhibits viral replication by activating the TBK1-IRF3 signaling pathway, further inhibiting the infection process of IAV.


Asunto(s)
Subtipo H1N1 del Virus de la Influenza A , Virus de la Influenza A , Gripe Humana , Humanos , Células A549 , Inflamación , Virus de la Influenza A/metabolismo , Subtipo H1N1 del Virus de la Influenza A/metabolismo , Gripe Humana/genética , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Interferón-alfa/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA