Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Nucleic Acids Res ; 52(D1): D770-D776, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-37930838

RESUMO

Rhinovirus (RV), a prominent causative agent of both upper and lower respiratory diseases, ranks among the most prevalent human respiratory viruses. RV infections are associated with various illnesses, including colds, asthma exacerbations, croup and pneumonia, imposing significant and extended societal burdens. Characterized by a high mutation rate and genomic diversity, RV displays a diverse serological landscape, encompassing a total of 174 serotypes identified to date. Understanding RV genetic diversity is crucial for epidemiological surveillance and investigation of respiratory diseases. This study introduces a comprehensive and high-quality RV data resource, designated RVdb (http://rvdb.mgc.ac.cn), covering 26 909 currently identified RV strains, along with RV-related sequences, 3D protein structures and publications. Furthermore, this resource features a suite of web-based utilities optimized for easy browsing and searching, as well as automatic sequence annotation, multiple sequence alignment (MSA), phylogenetic tree construction, RVdb BLAST and a serotyping pipeline. Equipped with a user-friendly interface and integrated online bioinformatics tools, RVdb provides a convenient and powerful platform on which to analyse the genetic characteristics of RVs. Additionally, RVdb also supports the efforts of virologists and epidemiologists to monitor and trace both existing and emerging RV-related infectious conditions in a public health context.


Assuntos
Asma , Infecções por Enterovirus , Infecções por Picornaviridae , Rhinovirus , Humanos , Genômica , Filogenia , Infecções por Picornaviridae/genética , Rhinovirus/genética
2.
Nat Commun ; 14(1): 2329, 2023 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-37087523

RESUMO

Rhinoviruses and allergens, such as house dust mite are major agents responsible for asthma exacerbations. The influence of pre-existing airway inflammation on the infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is largely unknown. We analyse mechanisms of response to viral infection in experimental in vivo rhinovirus infection in healthy controls and patients with asthma, and in in vitro experiments with house dust mite, rhinovirus and SARS-CoV-2 in human primary airway epithelium. Here, we show that rhinovirus infection in patients with asthma leads to an excessive RIG-I inflammasome activation, which diminishes its accessibility for type I/III interferon responses, leading to their early functional impairment, delayed resolution, prolonged viral clearance and unresolved inflammation in vitro and in vivo. Pre-exposure to house dust mite augments this phenomenon by inflammasome priming and auxiliary inhibition of early type I/III interferon responses. Prior infection with rhinovirus followed by SARS-CoV-2 infection augments RIG-I inflammasome activation and epithelial inflammation. Timely inhibition of the epithelial RIG-I inflammasome may lead to more efficient viral clearance and lower the burden of rhinovirus and SARS-CoV-2 infections.


Assuntos
Fatores de Restrição Antivirais , Asma , COVID-19 , Proteína DEAD-box 58 , Inflamassomos , Rhinovirus , Humanos , Fatores de Restrição Antivirais/genética , Fatores de Restrição Antivirais/metabolismo , Asma/genética , Asma/imunologia , COVID-19/genética , COVID-19/imunologia , Proteína DEAD-box 58/metabolismo , Infecções por Enterovirus/genética , Infecções por Enterovirus/imunologia , Inflamassomos/genética , Inflamassomos/metabolismo , Inflamação , Interferon Tipo I , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/imunologia , Rhinovirus/metabolismo , Rhinovirus/patogenicidade , SARS-CoV-2
3.
Nat Commun ; 14(1): 1817, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-37002207

RESUMO

Human parechovirus (PeV-A) is an RNA virus that belongs to the family Picornaviridae and it is currently classified into 19 genotypes. PeV-As usually cause mild illness in children and adults. Among the genotypes, PeV-A3 can cause severe diseases in neonates and young infants, resulting in neurological sequelae and death. In this study, we identify the human myeloid-associated differentiation marker (MYADM) as an essential host factor for the entry of six PeV-As (PeV-A1 to PeV-A6), including PeV-A3. The infection of six PeV-As (PeV-A1 to PeV-A6) to human cells is abolished by knocking out the expression of MYADM. Hamster BHK-21 cells are resistant to PeV-A infection, but the expression of human MYADM in BHK-21 confers PeV-A infection and viral production. Furthermore, VP0 capsid protein of PeV-A3 interacts with one extracellular domain of human MYADM on the cell membrane of BHK-21. The identification of MYADM as an essential entry factor for PeV-As infection is expected to advance our understanding of the pathogenesis of PeV-As.


Assuntos
Parechovirus , Infecções por Picornaviridae , Picornaviridae , Adulto , Criança , Humanos , Lactente , Recém-Nascido , Genótipo , Parechovirus/genética , Infecções por Picornaviridae/genética
4.
PLoS Pathog ; 19(2): e1011132, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36745686

RESUMO

Cyclic GMP-AMP synthase (cGAS) plays a key role in the innate immune responses to both DNA and RNA virus infection. Here, we found that enterovirus 71 (EV-A71), Seneca Valley virus (SVV), and foot-and-mouth disease virus (FMDV) infection triggered mitochondria damage and mitochondrial DNA (mtDNA) release in vitro and vivo. These responses were mediated by picornavirus 2B proteins which induced mtDNA release during viral replication. SVV infection caused the opening of mitochondrial permeability transition pore (mPTP) and led to voltage-dependent anion channel 1 (VDAC1)- and BCL2 antagonist/killer 1 (Bak) and Bak/BCL2-associated X (Bax)-dependent mtDNA leakage into the cytoplasm, while EV-A71 and FMDV infection induced mPTP opening and resulted in VDAC1-dependent mtDNA release. The released mtDNA bound to cGAS and activated cGAS-mediated antiviral immune response. cGAS was essential for inhibiting EV-A71, SVV, and FMDV replication by regulation of IFN-ß production. cGAS deficiency contributed to higher mortality of EV-A71- or FMDV-infected mice. In addition, we found that SVV 2C protein was responsible for decreasing cGAS expression through the autophagy pathway. The 9th and 153rd amino acid sites in 2C were critical for induction of cGAS degradation. Furthermore, we also show that EV-A71, CA16, and EMCV 2C antagonize the cGAS-stimulator of interferon genes (STING) pathway through interaction with STING, and highly conserved amino acids Y155 and S156 were critical for this inhibitory effect. In conclusion, these data reveal novel mechanisms of picornaviruses to block the antiviral effect mediated by the cGAS-STING signaling pathway, which will provide insights for developing antiviral strategies against picornaviruses.


Assuntos
Vírus da Febre Aftosa , Infecções por Picornaviridae , Animais , Camundongos , Antivirais/metabolismo , DNA Mitocondrial/genética , Vírus da Febre Aftosa/genética , Imunidade Inata , Interferon beta/metabolismo , Mitocôndrias/metabolismo , Nucleotidiltransferases/metabolismo , Infecções por Picornaviridae/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
5.
Viruses ; 14(10)2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36298792

RESUMO

Human rhinoviruses (HRVs) are small non-enveloped RNA viruses that belong to the Enterovirus genus within the Picornaviridae family and are known for causing the common cold. Though symptoms are generally mild in healthy individuals, the economic burden associated with HRV infection is significant. A vaccine could prevent disease. The Vero-cell-based viral vaccine platform technology was considered for such vaccine development. Unfortunately, most HRV strains are unable to propagate on Vero cells due to a lack of the major receptor of HRV group A and B, intercellular adhesion molecule (ICAM1, also known as CD54). Therefore, stable human ICAM1 expressing Vero cell clones were generated by transfecting the ICAM1 gene in Vero cells and selecting clones that overexpressed ICAM1 on the cell surface. Cell banks were made and expression of ICAM1 was stable for at least 30 passages. The Vero_ICAM1 cells and parental Vero cells were infected with four HRV prototypes, B14, A16, B37 and A57. Replication of all four viruses was detected in Vero_ICAM1, but not in the parental Vero cells. Altogether, Vero cells expressing ICAM1 could efficiently propagate the tested HRV strains. Therefore, ICAM1-expressing cells could be a useful tool for the development and future production of polyvalent HRV vaccines or other viruses that use ICAM1 as a receptor.


Assuntos
Molécula 1 de Adesão Intercelular , Infecções por Picornaviridae , Rhinovirus , Células Vero , Vacinas Virais , Animais , Humanos , Chlorocebus aethiops , Enterovirus/genética , Enterovirus/imunologia , Infecções por Enterovirus/genética , Infecções por Enterovirus/imunologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/imunologia , Rhinovirus/genética , Rhinovirus/imunologia , Células Vero/imunologia , Vacinas Virais/imunologia
6.
Virus Res ; 322: 198930, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36130655

RESUMO

BACKGROUND: Duck viral hepatitis (DVH) has a significant economic impact on duck industry, and duck hepatitis A virus genotype 3 (DHAV-3) is the most prevalent pathogen of DVH in Asian duck industry. The detailed study connecting differentially expressed genes (DEGs) and the differential resistance to DHAV-3 have not been accurately described, although a large numbers of DEGs have been identified by transcriptomic studies. RESULTS: Here, a resistant Pekin duck line (Z8R) and a susceptible Pekin duck line (Z8S) as models, high mortality and dramatically increased aspartate aminotransferase (AST), alanine aminotransferase (ALT) and the expression of immune-related genes of Z8S group were shown to be noticeable signs of cases caused by DHAV-3 infection. Compared with the control (Con) group, 1117 down-regulated DEGs and 612 up-regulated DEGs were found in the Z8S group and 37 down-regulated DEGs and 82 up-regulated DEGs were found in the Z8R group. Ultimately, the expression patterns of 10 DEGs were found to be diametrically opposite in Z8R and Z8S group. Functional analysis revealed that IFITM1 was associated with cell growth suppression, which was considered a key candidate gene. Results of flow cytometry showed that the conserved regions of IFITM1 (213-317 bp) could affected the cell cycle of duck embryo fibroblast (DEF) cells after infection with DHAV-3. Transcriptome and western blot analysis suggested that the CCND1, CCNE1 and CDK6 were significantly up-regulated in susceptible ducks by comparing with Con group. CONCLUSIONS: The hepatic injury and pathogenic outcomes caused by DHAV-3 infection were more severe in Z8S group compared to Z8R. Results of transcriptomics analysis and flow cytometry suggested that DHAV-3 infection can induce cell cycle changes that may be associated with IFITM1 expression level. These data will greatly enhance our understanding of the pathogenesis of DHAV-3 infection in ducklings and have implications for development of resistance breeding.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Patos , Genótipo , Vírus da Hepatite do Pato/genética , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/patologia , Transcriptoma
7.
Thorax ; 77(9): 929-932, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35790388

RESUMO

Despite good evidence of impaired innate antiviral responses in asthma, trials of inhaled interferon-ß given during exacerbations showed only modest benefits in moderate/severe asthma. Using human experimental rhinovirus infection, we observe robust in vivo induction of bronchial epithelial interferon response genes 4 days after virus inoculation in 25 subjects with asthma but not 11 control subjects. This signature correlated with virus loads and lower respiratory symptoms. Our data indicate that the in vivo innate antiviral response is dysregulated in asthma and open up the potential that prophylactic rather than therapeutic interferon therapy may have greater clinical benefit.


Assuntos
Asma , Imunidade Inata , Interferons , Infecções por Picornaviridae , Asma/imunologia , Asma/virologia , Células Epiteliais/imunologia , Humanos , Interferons/imunologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/imunologia , Rhinovirus
8.
Viruses ; 13(12)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34960671

RESUMO

Rhinoviruses (RV), like many other viruses, modulate programmed cell death to their own advantage. The viral protease, 3C has an integral role in the modulation, and we have shown that RVA-16 3C protease cleaves Receptor-interacting protein kinase-1 (RIPK1), a key host factor that modulates various cell death and cell survival pathways. In the current study, we have investigated whether this cleavage is conserved across selected RV strains. RIPK1 was cleaved in cells infected with strains representing diversity across phylogenetic groups (A and B) and receptor usage (major and minor groups). The cleavage was abrogated in the presence of the specific 3C protease inhibitor, Rupintrivir. Interestingly, there appears to be involvement of another protease (maybe 2A protease) in RIPK1 cleavage in strains belonging to genotype B. Our data show that 3C protease from diverse RV strains cleaves RIPK1, highlighting the importance of the cleavage to the RV lifecycle.


Assuntos
Proteases Virais 3C/metabolismo , Infecções por Picornaviridae/enzimologia , Rhinovirus/enzimologia , Proteases Virais 3C/genética , Antivirais/química , Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Isoxazóis/química , Isoxazóis/farmacologia , Fenilalanina/análogos & derivados , Fenilalanina/química , Fenilalanina/farmacologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/virologia , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Pirrolidinonas/química , Pirrolidinonas/farmacologia , Rhinovirus/química , Rhinovirus/efeitos dos fármacos , Rhinovirus/genética , Valina/análogos & derivados , Valina/química , Valina/farmacologia
9.
Front Immunol ; 12: 766740, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745142

RESUMO

Duck viral hepatitis (DVH) is an acute, highly lethal infectious disease of ducklings that causes huge losses in the duck industry. Duck hepatitis A virus genotype 3 (DHAV-3) has been one of the most prevalent DVH pathogen in the Asian duck industry in recent years. Here, we investigated the genetic basis of the resistance and susceptibility of ducks to DVH by comparing the genomes and transcriptomes of a resistant Pekin duck flock (Z8) and a susceptible Pekin duck flock (SZ7). Our comparative genomic and transcriptomic analyses suggested that NOD1 showed a strong signal of association with DVH susceptibility in ducks. Then, we found that NOD1 showed a significant expression difference between the livers of susceptible and resistant individuals after infection with DHAV-3, with higher expression in the SZ7 flock. Furthermore, suppression and overexpression experiments showed that the number of DHAV-3 genomic copies in primary duck hepatocytes was influenced by the expression level of NOD1. In addition, in situ RNAscope analysis showed that the localization of NOD1 and DHAV-3 in liver cells was consistent. Altogether, our data suggested that NOD1 was likely associated with DHAV-3 susceptibility in ducks, which provides a target for future investigations of the pathogenesis of DVH.


Assuntos
Proteínas Aviárias/genética , Patos/genética , Vírus da Hepatite do Pato/genética , Hepatite Viral Animal/genética , Proteína Adaptadora de Sinalização NOD1/genética , Infecções por Picornaviridae/genética , Doenças das Aves Domésticas/genética , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Proteínas Aviárias/metabolismo , Células Cultivadas , Patos/sangue , Patos/virologia , Feminino , Genótipo , Vírus da Hepatite do Pato/fisiologia , Hepatite Viral Animal/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Hibridização In Situ/métodos , Masculino , Proteína Adaptadora de Sinalização NOD1/metabolismo , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/virologia , Doenças das Aves Domésticas/virologia , RNA-Seq/métodos , Análise de Sobrevida , Transcriptoma/genética
10.
Pharmacol Res ; 174: 105956, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34700017

RESUMO

Asthma represents an inflammatory airway disease related to the induction of airway eosinophilia, mucus overproduction, and bronchial hyperresponsiveness. This study explored the effects of microRNA-423 (miR-423) on mitophagy and inflammation in asthmatic mice challenged with house dust mites (HDMs) and rhinovirus (RV). By searching for differentially expressed miRNAs in the GSE25230 microarray, miR-423 was identified as our target. Moreover, miR-423 was expressed at low levels in the lung tissues from patients with asthma, and agomiR-423 significantly inhibited RV-induced inflammatory injury and activation of inflammasome signaling in mouse lung tissues. Additionally, miR-423 downregulated the expression of IL-1ß/NLRP3/Caspase-1 inflammasome signaling by targeting phosphatase and tensin homolog-induced putative kinase 1 (PINK1). Furthermore, luciferase reporter experiments and ChIP-qPCR assays revealed that estrogen receptor 2 (ESR2) transcriptionally repressed miR-423 expression by coordinating with H3K9me2 modification of the miR-423 promoter histone. Overall, ESR2 synergized with the H3K9me2 modification of the miR-423 promoter histone to transcriptionally repress miR-423 expression and increase PINK1 expression in lung tissues, resulting in asthma exacerbation.


Assuntos
Asma/genética , Receptor beta de Estrogênio/genética , MicroRNAs , Proteínas Quinases/genética , Animais , Antígenos de Dermatophagoides , Asma/imunologia , Linhagem Celular , Citocinas/genética , Citocinas/imunologia , Feminino , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Pulmão/imunologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitofagia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/imunologia , Proteínas Quinases/imunologia , Rhinovirus , Transcrição Gênica
11.
Sci Rep ; 11(1): 17193, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34433834

RESUMO

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.


Assuntos
Simulação por Computador , Influenza Humana/virologia , Infecções por Picornaviridae/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Transcriptoma , Eliminação de Partículas Virais , Biomarcadores/metabolismo , Redes Reguladoras de Genes , Humanos , Influenza Humana/genética , Influenza Humana/metabolismo , Aprendizado de Máquina , Orthomyxoviridae/patogenicidade , Orthomyxoviridae/fisiologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/metabolismo , Infecções por Vírus Respiratório Sincicial/genética , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sinciciais Respiratórios/patogenicidade , Vírus Sinciciais Respiratórios/fisiologia , Rhinovirus/patogenicidade , Rhinovirus/fisiologia
12.
Am J Respir Cell Mol Biol ; 65(5): 544-554, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34181859

RESUMO

Human rhinovirus (RV) is a major risk factor for chronic obstructive pulmonary disease (COPD) and asthma exacerbations. The exploration of RV pathogenesis has been hampered by a lack of disease-relevant model systems. We performed a detailed characterization of host responses to RV infection in human lung tissue ex vivo and investigated whether these responses are disease relevant for patients with COPD and asthma. In addition, impact of the viral replication inhibitor rupintrivir was evaluated. Human precision-cut lung slices (PCLS) were infected with RV1B with or without rupintrivir. At Days 1 and 3 after infection, RV tissue localization, tissue viability, and viral load were determined. To characterize host responses to infection, mediator and whole genome analyses were performed. RV successfully replicated in PCLS airway epithelial cells and induced both antiviral and proinflammatory cytokines such as IFNα2a, CXCL10, CXCL11, IFN-γ, TNFα, and CCL5. Genomic analyses revealed that RV not only induced antiviral immune responses but also triggered changes in epithelial cell-associated pathways. Strikingly, the RV response in PCLS was reflective of gene expression changes described in patients with COPD and asthma. Although RV-induced host immune responses were abrogated by rupintrivir, RV-triggered epithelial processes were largely refractory to antiviral treatment. Detailed analysis of RV-infected human PCLS and comparison with gene signatures of patients with COPD and asthma revealed that the human RV PCLS model represents disease-relevant biological mechanisms that can be partially inhibited by a well-known antiviral compound and provide an outstanding opportunity to evaluate novel therapeutics.


Assuntos
Asma/genética , Interações Hospedeiro-Patógeno/genética , Pulmão/virologia , Infecções por Picornaviridae/genética , Doença Pulmonar Obstrutiva Crônica/genética , Idoso , Antivirais/farmacologia , Asma/patologia , Brônquios/patologia , Brônquios/fisiologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Isoxazóis/farmacologia , Pulmão/fisiologia , Masculino , Pessoa de Meia-Idade , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/patologia , Doença Pulmonar Obstrutiva Crônica/patologia , Pirrolidinonas/farmacologia , Rhinovirus/patogenicidade , Valina/análogos & derivados , Valina/farmacologia
13.
Sci Rep ; 11(1): 10948, 2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34040002

RESUMO

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.


Assuntos
Citocinas/biossíntese , Infecções por Picornaviridae/metabolismo , Rhinovirus , Coqueluche/metabolismo , Idade de Início , Antibacterianos/uso terapêutico , Coinfecção , Citocinas/genética , Progressão da Doença , Características da Família , Feminino , Regulação da Expressão Gênica , Humanos , Lactente , Recém-Nascido , Inflamação , Mediadores da Inflamação/sangue , Masculino , Nasofaringe/metabolismo , Nasofaringe/microbiologia , Nasofaringe/virologia , Infecções por Picornaviridae/genética , Fatores Socioeconômicos , Coqueluche/tratamento farmacológico , Coqueluche/genética
14.
Virus Res ; 300: 198408, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-33878402

RESUMO

Human rhinovirus (HRV) is one of the most common viruses, causing mild to severe respiratory tract infections in children and adults. Moreover, it can lead to patients' hospitalization. Nowadays, evaluation of gene expression alterations in host cells due to viral respiratory infections considered essential to understand the viral effects on cells. OBJECTIVE: In this study, we aimed to find important differentially expressed genes (DEGs) related to rhinitis and asthma exacerbation stimulated with Poly (I: C) and then to validate their expression in clinical samples of children how were less than 5 years old, hospitalized with severe acute respiratory infection (SARI) due to HRV infection in comparison with healthy cases. METHODS: Eight candidate genes involved in immunity, viral defense, inflammation, P53 pathway, and viral release processes were selected based on the analysis of a gene expression data set (GSE51392) and gene enrichment analysis. Then quantitative real-time PCR on cDNAs was performed for selected genes. The results were analyzed by Livak method and visualized by GraphPad prism software (8.4.3). RESULT: CXCL10, CMPK2, RSAD2, SERPINA3, TNFAIP6, CXCL14, IVNS1AB, and ZMAT3 were selected based on the enrichment and topological analysis of the constructed protein-protein interaction (PPI) network. Laboratory validation by real-time PCR showed CXCL10, CMPK2, RSAD2, SERPINA3, and TNFAIP6 (belonged to immunity, inflammatory responses and viral defense) were up-regulated, whereas CXCL14 (related to immunity) and IVNS1AB, ZMAT3 (associated to Influenza and P53 pathway) were down-regulated. CONCLUSION: Our results showed, that in children less than 5 years old affected by HRV and hospitalized with SARI, the inflammatory responses, antiviral defense, and type 1 interferon-signaling pathway have significantly affected by viral infection.


Assuntos
Infecções por Picornaviridae , Infecções Respiratórias , Pré-Escolar , Expressão Gênica , Humanos , Lactente , Infecções por Picornaviridae/genética , Infecções Respiratórias/genética , Infecções Respiratórias/virologia , Rhinovirus/genética , Proteína Supressora de Tumor p53/genética
15.
JCI Insight ; 6(7)2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33830082

RESUMO

Virus-induced respiratory tract infections are a major health burden in childhood, and available treatments are supportive rather than disease modifying. Rhinoviruses (RVs), the cause of approximately 80% of common colds, are detected in nearly half of all infants with bronchiolitis and the majority of children with an asthma exacerbation. Bronchiolitis in early life is a strong risk factor for the development of asthma. Here, we found that RV infection induced the expression of miRNA 122 (miR-122) in mouse lungs and in human airway epithelial cells. In vivo inhibition specifically in the lung reduced neutrophilic inflammation and CXCL2 expression, boosted innate IFN responses, and ameliorated airway hyperreactivity in the absence and in the presence of allergic lung inflammation. Inhibition of miR-122 in the lung increased the levels of suppressor of cytokine signaling 1 (SOCS1), which is an in vitro-validated target of miR-122. Importantly, gene silencing of SOCS1 in vivo completely reversed the protective effects of miR-122 inhibition on RV-induced lung disease. Higher miR-122 expression in nasopharyngeal aspirates was associated with a longer time on oxygen therapy and a higher rate of treatment failure in 87 infants hospitalized with moderately severe bronchiolitis. These results suggest that miR-122 promotes RV-induced lung disease via suppression of its target SOCS1 in vivo. Higher miR-122 expression was associated with worse clinical outcomes, highlighting the potential use of anti-miR-122 oligonucleotides, successfully trialed for treatment of hepatitis C, as potential therapeutics for RV-induced bronchiolitis and asthma exacerbations.


Assuntos
Bronquite/terapia , Pneumopatias/virologia , MicroRNAs/genética , Infecções por Picornaviridae/genética , Proteína 1 Supressora da Sinalização de Citocina/genética , Animais , Antagomirs/farmacologia , Bronquite/virologia , Quimiocina CXCL1/metabolismo , Quimiocina CXCL2/metabolismo , Feminino , Humanos , Lactente , Pneumopatias/genética , Pneumopatias/terapia , Masculino , Camundongos Endogâmicos BALB C , Nasofaringe/virologia , Infecções por Picornaviridae/tratamento farmacológico , Rhinovirus/fisiologia , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Falha de Tratamento , Replicação Viral
16.
APMIS ; 129(3): 143-151, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33230840

RESUMO

The effect of rhinovirus on airway epithelium is very well described. However, its influence on the vascular endothelium is unknown. The current study assesses the effect of rhinovirus HRV16 on the antiviral and inflammatory response in the human vascular endothelial cells (ECs). HRV16 increased IFN-ß, RANTES, and IP-10 mRNA expression and protein release. HRV16 copy number in ECs reached maximal value 10 h after incubation. Increase in virus copies was accompanied by the enhancement of Toll- and RIG-I-like receptors: TLR3, RIG-I, and MDA5. Additionally, HRV16 increased OAS-1 and PKR mRNA expression, enzymes responsible for virus degradation and inhibition of replication. ICAM-1 blockade decreased HRV16 copy number in ECs and inhibited IFN-ß, RANTES, IP-10, OAS1, PKR, TLR3, RIG-I, and MDA5 mRNA expression increase upon subsequent induction with HRV16. The vascular endothelium may be infected by human rhinovirus and generate antiviral and inflammatory innate response. Results of the study indicate the possible involvement of the vascular endothelium in the immunopathology of rhinoviral airway infections.


Assuntos
Endotélio Vascular/imunologia , Infecções por Picornaviridae/imunologia , Rhinovirus/imunologia , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/imunologia , Endotélio Vascular/virologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/virologia , Humanos , Interferon beta/genética , Interferon beta/imunologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/virologia , Receptores Imunológicos , Rhinovirus/genética , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia
17.
Anim Biotechnol ; 32(6): 676-682, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32180490

RESUMO

Specificity Protein 3 (SP3) is a newly identified regulator of tumor growth and invasiveness in humans. In this study, we identified and characterized the function of duck SP3 (duSP3). The full-length cDNA sequence of the duSP3 gene was cloned via rapid amplification of cDNA ends. It contained 2468 nucleotides, including a 111 base pair (bp) 5'-untranslated region (UTR), 215 bp 3'-UTR, and 2142 bp open reading frame (ORF), which encoded a 713 amino acid (AA) strongly conserved with Avian SP3. Tissue specificity analysis demonstrated that duSP3 was constitutively expressed in the eight tissues tested: liver, spleen, lung, heart, kidney, thymus, breast, and leg; and low expression levels were observed in all tissues, except the spleen and thymus. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis revealed that duSP3 expression rapidly increased in vitro after stimulation with both the hepatitis virus (DHV-1) and polyriboinosinic polyribocytidylic acid (poly(I:C)). However, the expression under these treatments varied in kidney and liver tissues; in the liver, duSP3 increased significantly at 36 h after the DHV-1 treatment and peaked at 72 h after poly(I:C) stimulation. These results suggested that SP3 may play a positive role in immune responses against viral infections in ducks.


Assuntos
Patos , Hepatite Viral Animal , Infecções por Picornaviridae , Fator de Transcrição Sp3/genética , Animais , Clonagem Molecular , DNA Complementar , Patos/genética , Patos/virologia , Vírus da Hepatite do Pato , Hepatite Viral Animal/genética , Hepatite Viral Animal/imunologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/veterinária
18.
Med Sci Monit ; 26: e928861, 2020 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-33315853

RESUMO

BACKGROUND Rhinovirus (RV) is the most common pathogen involved in asthma, and COVID-19, caused by SARS-COV-2, may be more severe in asthma patients. Here, we applied integrated bioinformatics to identify potential key genes and cytokine pathways after RV infection in asthma, and analyzed changes in angiotensin-converting enzyme 2 (ACE2), the cellular receptor of SARS-COV-2. MATERIAL AND METHODS The gene expression profile dataset GSE149273 was downloaded from NCBI-GEO, which included 90 samples of non-infected, RVA, and RVC. Differentially expressed genes (DEGs) were identified using t tests in the limma R package, and subsequently investigated by GO, KEGG, and DO analysis. Moreover, the expression of ACE2 and the proportion of immune cells were further analyzed to determine the effects of RV on cytokines. RESULTS A total of 555 DEGs of RVA and 421 of RVC were identified. There were 415 DEGs in RVA and RVC, of which 406 were upregulated and 9 were downregulated. The functional enrichment analysis showed that most DEGs were obviously enriched in cytokines, and were mainly enriched in "influenza" and "hepatitis C, chronic". In addition, the expression of ACE2 increased significantly and the proportion of immune cytokines significantly changed after RV infection. Our results suggest that RV can activate the cytokine pathway associated with COVID-19 by increasing ACE2. CONCLUSIONS The DEGs and related cytokine pathways after asthma RV infection identified using integrated bioinformatics in this study elucidate the potential link between RV and COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Asma/imunologia , COVID-19/imunologia , Citocinas/metabolismo , Infecções por Picornaviridae/imunologia , Mapas de Interação de Proteínas/genética , Asma/complicações , COVID-19/genética , COVID-19/virologia , Biologia Computacional , Conjuntos de Dados como Assunto , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Infecções por Picornaviridae/genética , Mapas de Interação de Proteínas/imunologia , Rhinovirus/imunologia , SARS-CoV-2/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
19.
Front Immunol ; 11: 568565, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33240261

RESUMO

Duck hepatitis A virus 3 (DHAV-3) is a wild endemic virus, which seriously endangers the duck industry in China. The present study aims to elucidate the mechanism of duck resistance to DHAV-3 infection. Both resistant and susceptible ducks were challenged with DHAV-3 in this experiment. The histopathological features and serum biochemical indices (ALT and AST) were analyzed to estimate liver injury status at 6, 12, 15, and 24 h post-infection (hpi). The dynamic transcriptomes of liver were analyzed to explain the molecular regulation mechanism in ducks against DHAV-3. The result showed that the liver injury in susceptible ducks was more serious than that in the resistant ducks throughout the four time points. A total of 2,127 differentially expressed genes (DEGs) were identified by comparing the transcriptome of the two populations. The expression levels of genes involved in innate immune response increased rapidly in susceptible ducks from 12 hpi. Similarly, the expression of genes involved in cytokine regulation also increased at the same time points, while the expression levels of these genes in resistant ducks remained similar between the various time points. KEGG enrichment analysis of the DEGs revealed that the genes involved in cytokine regulation and apoptosis were highly expressed in susceptible ducks than that in resistant ducks, suggesting that excessive cytokine storm and apoptosis may partially explain the mechanism of liver injury caused by DHAV-3 infection. Besides, we found that the FUT9 gene may contribute to resistance towards DHAV-3 in resistant ducklings. These findings will provide insight into duck resistance and susceptibility to DHAV-3 infection in the early phases, facilitate the development of a strategy for DHAV-3 prevention and treatment, and enhance genetic resistance via genetic selection in animal breeding.


Assuntos
Patos/genética , Vírus da Hepatite do Pato , Hepatite Viral Animal/genética , Infecções por Picornaviridae/genética , Doenças das Aves Domésticas/genética , Animais , Hepatite Viral Animal/patologia , Fígado/metabolismo , Fígado/patologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/veterinária , Doenças das Aves Domésticas/patologia , Transcriptoma
20.
PLoS Pathog ; 16(10): e1008970, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33104725

RESUMO

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.


Assuntos
Infecções por Picornaviridae/virologia , Picornaviridae/crescimento & desenvolvimento , RNA Polimerase Dependente de RNA/química , Enzimas de Conjugação de Ubiquitina/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Animais , Células Cultivadas , Cricetinae , Interações Hospedeiro-Patógeno , Picornaviridae/enzimologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/metabolismo , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Suínos , Enzimas de Conjugação de Ubiquitina/genética , Ubiquitinação , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...