Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Vet Res ; 55(1): 63, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760810

RESUMEN

The maintenance of viral protein homeostasis depends on the interaction between host cell proteins and viral proteins. As a molecular chaperone, heat shock protein 70 (HSP70) has been shown to play an important role in viral infection. Our results showed that HSP70 can affect translation, replication, assembly, and release during the life cycle of duck hepatitis A virus type 1 (DHAV-1). We demonstrated that HSP70 can regulate viral translation by interacting with the DHAV-1 internal ribosome entry site (IRES). In addition, HSP70 interacts with the viral capsid proteins VP1 and VP3 and promotes their stability by inhibiting proteasomal degradation, thereby facilitating the assembly of DHAV-1 virions. This study demonstrates the specific role of HSP70 in regulating DHAV-1 replication, which are helpful for understanding the pathogenesis of DHAV-1 infection and provide additional information about the role of HSP70 in infection by different kinds of picornaviruses, as well as the interaction between picornaviruses and host cells.


Asunto(s)
Proteínas HSP70 de Choque Térmico , Virus de la Hepatitis del Pato , Sitios Internos de Entrada al Ribosoma , Replicación Viral , Virus de la Hepatitis del Pato/fisiología , Virus de la Hepatitis del Pato/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Animales , Proteínas Estructurales Virales/metabolismo , Proteínas Estructurales Virales/genética , Patos , Enfermedades de las Aves de Corral/virología , Infecciones por Picornaviridae/veterinaria , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Hepatitis Viral Animal/virología , Hepatitis Viral Animal/metabolismo , Biosíntesis de Proteínas
2.
Nat Commun ; 15(1): 3469, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658526

RESUMEN

Human parechoviruses (PeV-A) are increasingly being recognized as a cause of infection in neonates and young infants, leading to a spectrum of clinical manifestations ranging from mild gastrointestinal and respiratory illnesses to severe sepsis and meningitis. However, the host factors required for parechovirus entry and infection remain poorly characterized. Here, using genome-wide CRISPR/Cas9 loss-of-function screens, we identify myeloid-associated differentiation marker (MYADM) as a host factor essential for the entry of several human parechovirus genotypes including PeV-A1, PeV-A2 and PeV-A3. Genetic knockout of MYADM confers resistance to PeV-A infection in cell lines and in human gastrointestinal epithelial organoids. Using immunoprecipitation, we show that MYADM binds to PeV-A1 particles via its fourth extracellular loop, and we identify critical amino acid residues within the loop that mediate binding and infection. The demonstrated interaction between MYADM and PeV-A1, and its importance specifically for viral entry, suggest that MYADM is a virus receptor. Knockout of MYADM does not reduce PeV-A1 attachment to cells pointing to a role at the post-attachment stage. Our study suggests that MYADM is a multi-genotype receptor for human parechoviruses with potential as an antiviral target to combat disease associated with emerging parechoviruses.


Asunto(s)
Parechovirus , Infecciones por Picornaviridae , Internalización del Virus , Humanos , Línea Celular , Sistemas CRISPR-Cas , Células HEK293 , Organoides/virología , Organoides/metabolismo , Parechovirus/genética , Parechovirus/metabolismo , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/metabolismo , Unión Proteica , Receptores Virales/metabolismo , Receptores Virales/genética
3.
Int J Mol Sci ; 24(4)2023 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-36835041

RESUMEN

Deficiencies in epithelial barrier integrity are involved in the pathogenesis of chronic rhinosinusitis (CRS). This study aimed to investigate the role of ephrinA1/ephA2 signaling on sinonasal epithelial permeability and rhinovirus-induced epithelial permeability. This role in the process of epithelial permeability was evaluated by stimulating ephA2 with ephrinA1 and inactivating ephA2 with ephA2 siRNA or inhibitor in cells exposed to rhinovirus infection. EphrinA1 treatment increased epithelial permeability, which was associated with decreased expression of ZO-1, ZO-2, and occludin. These effects of ephrinA1 were attenuated by blocking the action of ephA2 with ephA2 siRNA or inhibitor. Furthermore, rhinovirus infection upregulated the expression levels of ephrinA1 and ephA2, increasing epithelial permeability, which was suppressed in ephA2-deficient cells. These results suggest a novel role of ephrinA1/ephA2 signaling in epithelial barrier integrity in the sinonasal epithelium, suggesting their participation in rhinovirus-induced epithelial dysfunction.


Asunto(s)
Permeabilidad de la Membrana Celular , Células Epiteliales , Receptor EphA1 , Receptor EphA2 , Humanos , Permeabilidad de la Membrana Celular/genética , Permeabilidad de la Membrana Celular/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Infecciones por Picornaviridae/metabolismo , Receptor EphA2/metabolismo , Rhinovirus/patogenicidad , ARN Bicatenario , ARN Interferente Pequeño/metabolismo , Transducción de Señal/fisiología
4.
Front Immunol ; 13: 782936, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35242128

RESUMEN

BACKGROUND: Rhinovirus (RV) infections are a major cause of asthma exacerbations. Unlike other respiratory viruses, RV causes minimal cytotoxic effects on airway epithelial cells and cytokines play a critical role in its pathogenesis. However, previous findings on RV-induced cytokine responses were largely inconsistent. Thus, this study sought to identify the cytokine/chemokine profiles induced by RV infection and their correlations with airway inflammatory responses and/or respiratory symptoms using systematic review, and to determine whether a quantitative difference exists in cytokine levels between asthmatic and healthy individuals via meta-analysis. METHODS: Relevant articles were obtained from PubMed, Scopus, and ScienceDirect databases. Studies that compared RV-induced cytokine responses between asthmatic and healthy individuals were included in the systematic review, and their findings were categorized based on the study designs, which were ex vivo primary bronchial epithelial cells (PBECs), ex vivo peripheral blood mononuclear cells (PBMCs), and human experimental studies. Data on cytokine levels were also extracted and analyzed using Review Manager 5.4. RESULTS: Thirty-four articles were included in the systematic review, with 18 of these further subjected to meta-analysis. Several studies reported the correlations between the levels of cytokines, such as IL-8, IL-4, IL-5, and IL-13, and respiratory symptoms. Evidence suggests that IL-25 and IL-33 may be the cytokines that promote type 2 inflammation in asthmatics after RV infection. Besides that, a meta-analysis revealed that PBECs from children with atopic asthma produced significantly lower levels of IFN-ß [Effect size (ES): -0.84, p = 0.030] and IFN-λ (ES: -1.00, p = 0.002), and PBECs from adult atopic asthmatics produced significantly lower levels of IFN-ß (ES: -0.68, p = 0.009), compared to healthy subjects after RV infection. A trend towards a deficient production of IFN-γ (ES: -0.56, p = 0.060) in PBMCs from adult atopic asthmatics was observed. In lower airways, asthmatics also had significantly lower baseline IL-15 (ES: -0.69, p = 0.020) levels. CONCLUSION: Overall, RV-induced asthma exacerbations are potentially caused by an imbalance between Th1 and Th2 cytokines, which may be contributed by defective innate immune responses at cellular levels. Exogenous IFNs delivery may be beneficial as a prophylactic approach for RV-induced asthma exacerbations. SYSTEMATIC REVIEW REGISTRATION: https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=184119, identifier CRD42020184119.


Asunto(s)
Asma , Citocinas , Infecciones por Enterovirus , Hipersensibilidad Inmediata , Infecciones por Picornaviridae , Adulto , Niño , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Leucocitos Mononucleares , Infecciones por Picornaviridae/complicaciones , Infecciones por Picornaviridae/metabolismo , Rhinovirus
5.
J Virol ; 96(2): e0155021, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34757844

RESUMEN

Seneca Valley virus (SVV), a member of the Picornaviridae family, can activate autophagy via the PERK and ATF6 unfolded protein response pathways and facilitate viral replication; however, the precise molecular mechanism that regulates SVV-induced autophagy remains unclear. Here, we revealed that SVV infection inhibited the phosphorylation of mechanistic target of rapamycin kinase (MTOR) and activated phosphorylation of the serine/threonine kinase AKT. We observed that activating AMP-activated protein kinase (AMPK), extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (MAPK), and p38 MAPK signaling by SVV infection promoted autophagy induction and viral replication; additionally, the SVV-induced autophagy was independent of the ULK1 complex. We further evaluated the role of viral protein(s) in the AKT-AMPK-MAPK-MTOR pathway during SVV-induced autophagy and found that VP1 induced autophagy, as evidenced by puncta colocalization with microtubule-associated protein 1 light chain 3 (LC3) in the cytoplasm and enhanced LC3-II levels. This might be associated with the interaction of VP1 with sequestosome 1 and promoting its degradation. In addition, the expression of VP1 enhanced AKT phosphorylation and AMPK phosphorylation, while MTOR phosphorylation was inhibited. These results indicate that VP1 induces autophagy by the AKT-AMPK-MTOR pathway. Additionally, expression of VP3 and 3C was found to activate autophagy induction via the ERK1/2 MAPK-MTOR and p38 MAPK-MTOR pathway. Taken together, our data suggest that SVV-induced autophagy has finely tuned molecular mechanisms in which VP1, VP3, and 3C contribute synergistically to the AKT-AMPK-MAPK-MTOR pathway. IMPORTANCE Autophagy is an essential cellular catabolic process to sustain normal physiological processes that are modulated by a variety of signaling pathways. Invading virus is a stimulus to induce autophagy that regulates viral replication. It has been demonstrated that Seneca Valley virus (SVV) induced autophagy via the PERK and ATF6 unfolded protein response pathways. However, the precise signaling pathway involved in autophagy is still poorly understood. In this study, our results demonstrated that viral proteins VP1, VP3, and 3C contribute synergistically to activation of the AKT-AMPK-MAPK-MTOR signaling pathway for SVV-induced autophagy. These findings reveal systemically the finely tuned molecular mechanism of SVV-induced autophagy, thereby facilitating deeper insight into the development of potential control strategies against SVV infection.


Asunto(s)
Proteasas Virales 3C/metabolismo , Autofagia , Proteínas de la Cápside/metabolismo , Picornaviridae/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Línea Celular , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosforilación , Picornaviridae/metabolismo , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Proteína Sequestosoma-1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Replicación Viral
6.
Front Immunol ; 12: 753683, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899705

RESUMEN

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Asunto(s)
Efecto Citopatogénico Viral , Modelos Animales de Enfermedad , Encefalitis Viral/metabolismo , Parechovirus/patogenicidad , Infecciones por Picornaviridae/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Autofagia , Línea Celular Tumoral , Corteza Cerebral/virología , Chlorocebus aethiops , Citocinas/biosíntesis , Citocinas/genética , Encefalitis Viral/patología , Encefalitis Viral/virología , Glioblastoma/patología , Hipocampo/virología , Humanos , Inflamación , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Interferón Tipo I/farmacología , Interferones/biosíntesis , Interferones/genética , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/patología , Parechovirus/efectos de los fármacos , Parechovirus/fisiología , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Piroptosis , Células Vero , Replicación Viral/efectos de los fármacos , Interferón lambda
7.
Arch Virol ; 166(11): 3105-3116, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34482448

RESUMEN

Several outbreaks of duck hepatitis A virus type 1 (DHAV-1), which were characterized by yellow coloration and hemorrhage in pancreatic tissues, have occurred in China. The causative agent is called pancreatitis-associated DHAV-1. The mechanisms involved in pancreatitis-associated DHAV-1 infection are still unclear. Transcriptome analysis of duck pancreas infected with classical-type DHAV-1 and pancreatitis-associated DHAV-1 was carried out. Deep sequencing with Illumina-Solexa resulted in a total of 53.9 Gb of clean data from the cDNA library of the pancreas, and a total of 29,597 unigenes with an average length of 993.43 bp were generated by de novo sequence assembly. The expression levels of D-3-phosphoglycerate dehydrogenase, phosphoserine aminotransferase, and phosphoserine phosphatase, which are involved in glycine, serine, and threonine metabolism pathways, were significantly downregulated in ducks infected with pancreatitis-associated DHAV-1 compared with those infected with classical-type DHAV-1. These findings provide information regarding differences in expression levels of metabolism-associated genes between ducks infected with pancreatitis-associated DHAV-1 and those infected with classical-type DHAV-1, indicating that intensive metabolism disorders may contribute to the different phenotypes of DHAV-1-infection.


Asunto(s)
Virus de la Hepatitis del Pato/patogenicidad , Hepatitis Viral Animal/virología , Interacciones Huésped-Patógeno/genética , Infecciones por Picornaviridae/veterinaria , Enfermedades de las Aves de Corral/virología , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Patos/virología , Expresión Génica , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/patología , Páncreas/citología , Páncreas/patología , Páncreas/virología , Pancreatitis/patología , Pancreatitis/virología , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN
8.
Mol Cell Proteomics ; 20: 100147, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34530158

RESUMEN

Seneca Valley virus (SVV) or commonly known as senecavirus A, is one of the picornavirus that is associated with vesicular disease and neonatal mortality in swine herds. Our previous study found that SVV replicates extremely faster in porcine Instituto Biologico-Rim Suino-2 (IBRS-2) cells than that in porcine kidney-15 (PK-15) cells. However, the underlying mechanism remains unknown. In this study, we comprehensively compared the expression features between IBRS-2 cells and PK-15 cells in response to SVV infection by an unbiased high-throughput quantitative proteomic analysis. We found that the innate immune response-related pathways were efficiently activated in PK-15 cells but not in IBRS-2 cells during SVV infection. A large amount of interferon (IFN)-stimulated genes were induced in PK-15 cells. In contrast, no IFN-stimulated genes were induced in IBRS-2 cells. Besides, we determined similar results in the two cell lines infected by another porcine picornavirus foot-and-mouth disease virus. Further study demonstrated that the Janus kinase signal transducer and activator of transcription signaling pathway was functioning properly in both IBRS-2 and PK-15 cells. A systematic screening study revealed that the aberrant signal transduction from TANK-binding kinase 1 to IFN regulatory factor 3 in the retinoic acid-inducible gene I-like receptor signaling pathway in IBRS-2 cells was the fundamental cause of the different innate immune response manifestation and different viral replication rate in the two cell lines. Together, our findings determined the different features of IBRS-2 and PK-15 cell lines, which will help for clarification of the pathogenesis of SVV. Besides, identification of the underlying mechanisms will provide new targets and an insight for decreasing the viral clearance rate and probably improve the oncolytic effect by SVV in cancer cells.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Picornaviridae/fisiología , Receptores Inmunológicos/metabolismo , Animales , Línea Celular , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Transducción de Señal , Porcinos , Replicación Viral
9.
Sci Rep ; 11(1): 17193, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34433834

RESUMEN

This paper addresses the development of predictive models for distinguishing pre-symptomatic infections from uninfected individuals. Our machine learning experiments are conducted on publicly available challenge studies that collected whole-blood transcriptomics data from individuals infected with HRV, RSV, H1N1, and H3N2. We address the problem of identifying discriminatory biomarkers between controls and eventual shedders in the first 32 h post-infection. Our exploratory analysis shows that the most discriminatory biomarkers exhibit a strong dependence on time over the course of the human response to infection. We visualize the feature sets to provide evidence of the rapid evolution of the gene expression profiles. To quantify this observation, we partition the data in the first 32 h into four equal time windows of 8 h each and identify all discriminatory biomarkers using sparsity-promoting classifiers and Iterated Feature Removal. We then perform a comparative machine learning classification analysis using linear support vector machines, artificial neural networks and Centroid-Encoder. We present a range of experiments on different groupings of the diseases to demonstrate the robustness of the resulting models.


Asunto(s)
Simulación por Computador , Gripe Humana/virología , Infecciones por Picornaviridae/virología , Infecciones por Virus Sincitial Respiratorio/virología , Transcriptoma , Esparcimiento de Virus , Biomarcadores/metabolismo , Redes Reguladoras de Genes , Humanos , Gripe Humana/genética , Gripe Humana/metabolismo , Aprendizaje Automático , Orthomyxoviridae/patogenicidad , Orthomyxoviridae/fisiología , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/metabolismo , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitiales Respiratorios/patogenicidad , Virus Sincitiales Respiratorios/fisiología , Rhinovirus/patogenicidad , Rhinovirus/fisiología
10.
Viruses ; 13(7)2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201715

RESUMEN

Picornaviruses are positive-stranded RNA viruses. Even though replication and translation of their genome take place in the cytoplasm, these viruses evolved different strategies to disturb nucleocytoplasmic trafficking of host proteins and RNA. The major targets of picornavirus are the phenylalanine-glycine (FG)-nucleoporins, which form a mesh in the central channel of the nuclear pore complex through which protein cargos and karyopherins are actively transported in both directions. Interestingly, while enteroviruses use the proteolytic activity of their 2A protein to degrade FG-nucleoporins, cardioviruses act by triggering phosphorylation of these proteins by cellular kinases. By targeting the nuclear pore complex, picornaviruses recruit nuclear proteins to the cytoplasm, where they increase viral genome translation and replication; they affect nuclear translocation of cytoplasmic proteins such as transcription factors that induce innate immune responses and retain host mRNA in the nucleus thereby preventing cell emergency responses and likely making the ribosomal machinery available for translation of viral RNAs.


Asunto(s)
Núcleo Celular/metabolismo , Infecciones por Picornaviridae/metabolismo , Picornaviridae/metabolismo , Transporte Activo de Núcleo Celular , Humanos , Carioferinas/metabolismo , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Picornaviridae/clasificación , Infecciones por Picornaviridae/virología , Especificidad de la Especie , Proteínas Virales/metabolismo , Replicación Viral , Proteína de Unión al GTP ran/metabolismo
11.
Sci Rep ; 11(1): 10948, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34040002

RESUMEN

Mechanisms of interaction between Bordetella pertussis and other viral agents are yet to be fully explored. We studied the inflammatory cytokine expression patterns among children with both viral-bacterial infections. Nasopharyngeal aspirate (NPA) samples were taken from children, aged < 1 year, positive for Rhinovirus, Bordetella pertussis and for Rhinovirus and Bordetella pertussis. Forty cytokines were evaluated in NPA by using human cytokine protein arrays and a quantitative analysis was performed on significantly altered cytokines. Forty cytokines were evaluated in NPA by using human cytokine protein arrays and a quantitative analysis was performed on significantly altered cytokines. Our results show that co-infections display a different inflammatory pattern compared to single infections, suggesting that a chronic inflammation caused by one of the two pathogens could be the trigger for exacerbation in co-infections.


Asunto(s)
Citocinas/biosíntesis , Infecciones por Picornaviridae/metabolismo , Rhinovirus , Tos Ferina/metabolismo , Edad de Inicio , Antibacterianos/uso terapéutico , Coinfección , Citocinas/genética , Progresión de la Enfermedad , Composición Familiar , Femenino , Regulación de la Expresión Génica , Humanos , Lactante , Recién Nacido , Inflamación , Mediadores de Inflamación/sangre , Masculino , Nasofaringe/metabolismo , Nasofaringe/microbiología , Nasofaringe/virología , Infecciones por Picornaviridae/genética , Factores Socioeconómicos , Tos Ferina/tratamiento farmacológico , Tos Ferina/genética
12.
PLoS Pathog ; 16(10): e1008970, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33104725

RESUMEN

Senecavirus A (SVA), discovered in 2002, is an emerging pathogen of swine that has since been reported in numerous pork producing countries. To date, the mechanism of SVA replication remains poorly understood. In this study, utilizing iTRAQ analysis we found that UBE2L6, an E2 ubiquitin-conjugating enzyme, is up-regulated in SVA-infected BHK-21 cells, and that its overexpression promotes SVA replication. We determined that UBE2L6 interacts with, and ubiquitinates the RNA-dependent RNA polymerase of SVA, (the 3D protein) and this ubiquitination serves to inhibit the degradation of 3D. UBE2L6-mediated ubiquitination of 3D requires a cystine at residue 86 in UBE2L6, and lysines at residues 169 and 321 in 3D. Virus with mutations in 3D (rK169R and rK321R) exhibited significantly decreased replication compared to wild type SVA and the repaired viruses, rK169R(R) and rK321R(R). These data indicate that UBE2L6, the enzyme, targets the 3D polymerase, the substrate, during SVA infection to facilitate replication.


Asunto(s)
Infecciones por Picornaviridae/virología , Picornaviridae/crecimiento & desarrollo , ARN Polimerasa Dependiente del ARN/química , Enzimas Ubiquitina-Conjugadoras/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Animales , Células Cultivadas , Cricetinae , Interacciones Huésped-Patógeno , Picornaviridae/enzimología , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/metabolismo , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Porcinos , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitinación , Proteínas Virales/genética
13.
Int J Pharm ; 589: 119826, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32871219

RESUMEN

Viral infections represent 44% of newly emerging infections, and as is shown by the COVID-19 outbreak constitute a major risk to human health and wellbeing. Although there are many efficient antiviral agents, they still have drawbacks such as development of virus resistance and accumulation within off-target organs. Encapsulation of antiviral agents into nanoparticles (NPs) has been shown to improve bioavailability, control release, and reduce side effects. However, there is little quantitative understanding of how the uptake of NPs into virally infected cells compares to uninfected cells. In this work, the uptake of fluorescently labeled polymer NPs was investigated in several models of rhinovirus (RV) infected cells. Different multiplicities of RV infections (MOI) and timings of NPs uptake were also investigated. In some cases, RV infection resulted in a significant increase of NPs uptake, but this was not universally noted. For HeLa cells, RV-A16 and RV-A01 infection elevated NPs uptake upon increasing the incubation time, whereas at later timepoints (6 h) a reduced uptake was noted with RV-A01 infection (owing to decreased cell viability). Beas-2B cells exhibited more complex trends: decreases in NPs uptake (cf. uninfected cells) were observed at short incubation times following RV-A01 and RV-A16 infection. At later incubation times (4 h), we found a marked decrease of NPs uptake for RV-A01 infected cells but an increase in uptake with RV-A16 infected cells. Where increases in NPs uptake were found, they were very modest compared to results previously reported for a hepatitis C/ Huh7.5 cell line model. An increase in RV dose (MOI) was not associated with any notable change of NPs uptake. We argue that the diverse endocytic pathways among the different cell lines, together with changes in virus nature, size, and entry mechanism are responsible for these differences. These findings suggest that NPs entry into virally infected cells is a complex process, and further work is required to unravel the different factors which govern this. Undertaking this additional research will be crucial to develop potent nanomedicines for the delivery of antiviral agents.


Asunto(s)
Nanopartículas/administración & dosificación , Infecciones por Picornaviridae/metabolismo , Poliésteres/administración & dosificación , Rhinovirus , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN Viral , Endocitosis , Genoma Viral , Humanos , Rhinovirus/genética
15.
Int J Mol Sci ; 21(12)2020 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-32575635

RESUMEN

Porcine sapelovirus A (PSV) is a single stranded, positive-sense, non-enveloped RNA virus that causes enteritis, pneumonia, polioencephalomyelitis, and reproductive disorders in pigs. Research on PSV infection and interaction with host cells is unclear. In this study, we applied tandem mass tag proteomics analysis to investigate the differentially expressed proteins (DEPs) in PSV-infected pig kidney (PK)-15 cells and explored the interactions between PSV and host cells. Here we mapped 181 DEPs, including 59 up-regulated and 122 down-regulated DEPs. Among them, osteopontin (SPP1), induced protein with tetratricopeptide repeats 5 (IFIT5), ISG15 ubiquitin-like modifier (ISG15), vinculin (VCL), and syndecan-1 (SDC1) were verified significantly changed using RT-qPCR. Additionally, overexpression of SDC1 promoted PSV viral protein (VP)1 synthesis and virus titer, and silencing of SDC1 revealed the opposite results. Our findings show that SDC1 is a novel host protein and plays crucial roles in regulating PSV replication.


Asunto(s)
Infecciones por Picornaviridae/metabolismo , Picornaviridae/fisiología , Proteómica/métodos , Enfermedades de los Porcinos/virología , Sindecano-1/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica , Modelos Biológicos , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/veterinaria , Porcinos , Enfermedades de los Porcinos/genética , Enfermedades de los Porcinos/metabolismo , Sindecano-1/genética , Espectrometría de Masas en Tándem , Carga Viral , Proteínas Virales/metabolismo , Replicación Viral
16.
J Virol ; 94(16)2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32522851

RESUMEN

Picornaviruses have both asexual and sexual RNA replication mechanisms. Asexual RNA replication mechanisms involve one parental template, whereas sexual RNA replication mechanisms involve two or more parental templates. Because sexual RNA replication mechanisms counteract ribavirin-induced error catastrophe, we selected for ribavirin-resistant poliovirus to identify polymerase residues that facilitate sexual RNA replication mechanisms. We used serial passage in ribavirin, beginning with a variety of ribavirin-sensitive and ribavirin-resistant parental viruses. Ribavirin-sensitive virus contained an L420A polymerase mutation, while ribavirin-resistant virus contained a G64S polymerase mutation. A G64 codon mutation (G64Fix) was used to inhibit emergence of G64S-mediated ribavirin resistance. Revertants (L420) or pseudorevertants (L420V and L420I) were selected from all independent lineages of L420A, G64Fix L420A, and G64S L420A parental viruses. Ribavirin resistance G64S mutations were selected in two independent lineages, and novel ribavirin resistance mutations were selected in the polymerase in other lineages (M299I, M323I, M392V, and T353I). The structural orientation of M392, immediately adjacent to L420 and the polymerase primer grip region, led us to engineer additional polymerase mutations into poliovirus (M392A, M392L, M392V, K375R, and R376K). L420A revertants and pseudorevertants (L420V and L420I) restored efficient viral RNA recombination, confirming that ribavirin-induced error catastrophe coincides with defects in sexual RNA replication mechanisms. Viruses containing M392 mutations (M392A, M392L, and M392V) and primer grip mutations (K375R and R376K) exhibited divergent RNA recombination, ribavirin sensitivity, and biochemical phenotypes, consistent with changes in the fidelity of RNA synthesis. We conclude that an extended primer grip of the polymerase, including L420, M392, K375, and R376, contributes to the fidelity of RNA synthesis and to efficient sexual RNA replication mechanisms.IMPORTANCE Picornaviruses have both asexual and sexual RNA replication mechanisms. Sexual RNA replication shapes picornavirus species groups, contributes to the emergence of vaccine-derived polioviruses, and counteracts error catastrophe. Can viruses distinguish between homologous and nonhomologous partners during sexual RNA replication? We implicate an extended primer grip of the viral polymerase in sexual RNA replication mechanisms. By sensing RNA sequence complementarity near the active site, the extended primer grip of the polymerase has the potential to distinguish between homologous and nonhomologous RNA templates during sexual RNA replication.


Asunto(s)
Picornaviridae/genética , ARN Polimerasa Dependiente del ARN/genética , Replicación Viral/efectos de los fármacos , Sustitución de Aminoácidos/genética , Antivirales/farmacología , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Farmacorresistencia Viral/efectos de los fármacos , Células HeLa , Humanos , Mutación/efectos de los fármacos , Picornaviridae/metabolismo , Picornaviridae/patogenicidad , Infecciones por Picornaviridae/genética , Infecciones por Picornaviridae/metabolismo , Poliovirus/genética , ARN/genética , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Ribavirina/farmacología , Replicación Viral/genética
17.
J Breath Res ; 14(4): 041001, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32531777

RESUMEN

The COVID-19 pandemic has highlighted the importance of rapid, cost effective, accurate, and non-invasive testing for viral infections. Volatile compounds (VCs) have been suggested for several decades as fulfilling these criteria. However currently very little work has been done in trying to diagnose viral infections using VCs. Much of the work carried out to date involves the differentiation of bacterial and viral sources of infection and often the detection of bacterial and viral co-infection. However, this has usually been done in vitro and very little work has involved the use of human participants. Viruses hijack the host cell metabolism and do not produce their own metabolites so identifying virus specific VCs is at best a challenging task. However, there are proteins and lipids that are potential candidates as markers of viral infection. The current understanding is that host cell glycolysis is upregulated under viral infection to increase the available energy for viral replication. There is some evidence that viral infection leads to the increase of production of fatty acids, alkanes, and alkanes related products. For instance, 2,3-butandione, aldehydes, 2,8-dimethyl-undecane and n-propyl acetate have all been correlated with viral infection. Currently, the literature points to markers of oxidative stress (e.g. nitric oxide, aldehydes etc) being the most useful in the determination of viral infection. The issue, however, is that there are also many other conditions that can lead to oxidative stress markers being produced. In this review a range of (mainly mass spectrometric) methods are discussed for viral detection in breath, including breath condensate. Currently MALDI-ToF-MS is likely to be the preferred method for the identification of viral strains and variants of those strains, however it is limited by its need for the viral strains to have been sequenced and logged in a database.


Asunto(s)
Pruebas Respiratorias/métodos , Virosis/diagnóstico , Aldehídos/metabolismo , Animales , Betacoronavirus , Biomarcadores/metabolismo , COVID-19 , Prueba de COVID-19 , Técnicas de Laboratorio Clínico , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/metabolismo , Cromatografía de Gases y Espectrometría de Masas , Hepatitis B/diagnóstico , Hepatitis B/metabolismo , Humanos , Gripe Humana/diagnóstico , Gripe Humana/metabolismo , Espectrometría de Masas , Óxido Nítrico/metabolismo , Infecciones por Orthomyxoviridae/diagnóstico , Infecciones por Orthomyxoviridae/metabolismo , Estrés Oxidativo , Pandemias , Infecciones por Picornaviridae/diagnóstico , Infecciones por Picornaviridae/metabolismo , Neumonía Viral/diagnóstico , Neumonía Viral/metabolismo , Infecciones por Rotavirus/diagnóstico , Infecciones por Rotavirus/metabolismo , SARS-CoV-2 , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Porcinos , Virosis/metabolismo , Virus
18.
Gene ; 748: 144710, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32339622

RESUMEN

Earlier works identified the second generation (Z8R2) of a resistant Pekin duck line to duck hepatitis A virus genotype 3 (DHAV-3), which displays significantly strong resistance than that of the second generation (Z8S2) of a susceptible Pekin duck line. To understand the genetic mechanisms that determine the different resistance/susceptibility of Z8R2 and Z8S2 to DHAV-3, transcriptome analysis on livers of infected Pekin ducklings was performed to screen differentially expressed genes (DEGs). We found that DHAV-3 infection has a great effect on metabolism of Z8S2 at the transcription level. Using a newly created fourth generation of the resistant Pekin duck line (Z8R4) and an unselected Pekin duck flock (Z7) as models, hypoglycemia and dramatically increased aspartate aminotransferase and alanine aminotransferase were shown to be noticeable signs of fatal cases caused by DHAV-3 infection. These findings, together with expression analysis and verification of DEGs, support the view that DHAV-3 infection results in glucose metabolic abnormalities in susceptible individuals and that there are significant differences in expression patterns of glucose metabolism-related DEGs between susceptible and resistant individuals. Notably, cytokines displayed a negative correlation with glucose synthesis in terms of expression in susceptible individuals following DHAV-3 infection. Mechanism analyses suggests that cytokines will activate PI3K-AKT pathway and/or JAK-STAT pathway by up-regulated expression of JAK2, and thereby causes down-regulated expression of G6PC and/or ACAT1. Cytokines can also cause down-regulated expression of HPGDS by JAK2. The present work contributes to the understanding of pathogenesis of DHAV-3 infection and the resistance breeding project against DHAV-3.


Asunto(s)
Patos/virología , Glucosa/metabolismo , Virus de la Hepatitis del Pato/genética , Infecciones por Picornaviridae/metabolismo , Animales , Genotipo , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología
20.
Front Immunol ; 11: 85, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117246

RESUMEN

Human rhinoviruses (HRV) are the most common cause of viral respiratory tract infections. While normally mild and self-limiting in healthy adults, HRV infections are associated with bronchiolitis in infants, pneumonia in immunocompromised patients, and exacerbations of asthma and COPD. The human cathelicidin LL-37 is a host defense peptide (HDP) with broad immunomodulatory and antimicrobial activities that has direct antiviral effects against HRV. However, LL-37 is known to be susceptible to the enzymatic activity of peptidyl arginine deiminases (PAD), and exposure of the peptide to these enzymes results in the conversion of positively charged arginines to neutral citrullines (citrullination). Here, we demonstrate that citrullination of LL-37 reduced its direct antiviral activity against HRV. Furthermore, while the anti-rhinovirus activity of LL-37 results in dampened epithelial cell inflammatory responses, citrullination of the peptide, and a loss in antiviral activity, ameliorates this effect. This study also demonstrates that HRV infection upregulates PAD2 protein expression, and increases levels of protein citrullination, including histone H3, in human bronchial epithelial cells. Increased PADI gene expression and HDP citrullination during infection may represent a novel viral evasion mechanism, likely applicable to a wide range of pathogens, and should therefore be considered in the design of therapeutic peptide derivatives.


Asunto(s)
Catelicidinas/metabolismo , Citrulinación , Fragmentos de Péptidos/metabolismo , Infecciones por Picornaviridae/metabolismo , Rhinovirus , Bronquios , Catelicidinas/inmunología , Línea Celular , Citocinas/metabolismo , Células Epiteliales , Humanos , Fragmentos de Péptidos/inmunología , Infecciones por Picornaviridae/inmunología , Poli I-C/metabolismo , Arginina Deiminasa Proteína-Tipo 2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA