Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 254
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 327(4): L557-L573, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39189801

RESUMO

Human coronavirus (HCoV)-NL63 causes respiratory tract infections in humans and uses angiotensin-converting enzyme 2 (ACE2) as a receptor. We sought to establish a mouse model of HCoV-NL63 and determine whether prior rhinovirus (RV)-A1B infection affected HCoV-NL63 replication. HCoV-NL63 was propagated in LLC-MK2 cells expressing human ACE2. RV-A1B was grown in HeLa-H1 cells. C57BL6/J or transgenic mice expressing human ACE2 were infected intranasally with sham LLC-MK2 cell supernatant or 1 × 105 tissue culture infectious dose (TCID50) units HCoV-NL63. Wild-type mice were infected with 1 × 106 plaque-forming units (PFU) RV-A1B. Lungs were assessed for vRNA, bronchoalveolar lavage (BAL) cells, histology, HCoV-NL63 nonstructural protein 3 (nsp3), and host gene expression by next-generation sequencing and qPCR. To evaluate sequential infections, mice were infected with RV-A1B followed by HCoV-NL63 infection 4 days later. We report that hACE2 mice infected with HCoV-NL63 showed evidence of replicative infection with increased levels of vRNA, BAL neutrophils and lymphocytes, peribronchial and perivascular infiltrates, and expression of nsp3. Viral replication peaked 3 days after infection and inflammation persisted 6 days after infection. HCoV-NL63-infected hACE2 mice showed increased mRNA expression of IFNs, IFN-stimulated proteins, and proinflammatory cytokines. Infection with RV-A1B 4 days before HCoV-NL63 significantly decreased both HCoV-NL63 vRNA levels and airway inflammation. Mice infected with RV-A1B prior to HCoV-NL63 showed increased expression of antiviral proteins compared with sham-treated mice. In conclusion, we established a mouse model of HCoV-NL63 replicative infection characterized by relatively persistent viral replication and inflammation. Prior infection with RV-A1B reduced HCoV-NL63 replication and airway inflammation, indicative of viral interference.NEW & NOTEWORTHY We describe a mouse model of human coronavirus (HCoV) infection. Infection of transgenic mice expressing human angiotensin-converting enzyme 2 (ACE2) with HCoV-NL63 produced a replicative infection with peribronchial inflammation and nonstructural protein 3 expression. Mice infected with RV-A1B 4 days before HCoV-NL63 showed decreased HCoV-NL63 replication and airway inflammation and increased expression of antiviral proteins compared with sham-treated mice. This research may shed light on human coronavirus infections, viral interference, and viral-induced asthma exacerbations.


Assuntos
Enzima de Conversão de Angiotensina 2 , Coronavirus Humano NL63 , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Infecções por Picornaviridae , Rhinovirus , Animais , Humanos , Camundongos , Coronavirus Humano NL63/fisiologia , Rhinovirus/fisiologia , Rhinovirus/patogenicidade , Infecções por Picornaviridae/virologia , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/patologia , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Replicação Viral , Infecções por Coronavirus/virologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/metabolismo , Camundongos Transgênicos , Pulmão/virologia , Pulmão/patologia , Pulmão/metabolismo , Células HeLa , Líquido da Lavagem Broncoalveolar/virologia , Enterovirus
2.
FASEB J ; 35(1): e21208, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33230881

RESUMO

Human Rhinovirus (HRV) is a major cause of common cold, bronchiolitis, and exacerbations of chronic pulmonary diseases such as asthma. CD8 T cell responses likely play an important role in the control of HRV infection but, surprisingly, HRV-specific CD8 T cell epitopes remain yet to be identified. Here, we approached the discovery and characterization of conserved HRV-specific CD8 T cell epitopes from species A (HRV A) and C (HRV C), the most frequent subtypes in the clinics of various pulmonary diseases. We found IFNγ-ELISPOT positive responses to 23 conserved HRV-specific peptides on peripheral blood mononuclear cells (PBMCs) from 14 HLA I typed subjects. Peptide-specific IFNγ production by CD8 T cells and binding to the relevant HLA I were confirmed for six HRV A-specific and three HRV C-specific CD8 T cell epitopes. In addition, we validated A*02:01-restricted epitopes by DimerX staining and found out that these peptides mediated cytotoxicity. All these A*02:01-restricted epitopes were 9-mers but, interestingly, we also identified and validated an unusually long 16-mer epitope peptide restricted by A*02:01, HRVC1791-1806 (GLEPLDLNTSAGFPYV). HRV-specific CD8 T cell epitopes describe here are expected to elicit CD8 T cell responses in up to 87% of the population and could be key for developing an HRV vaccine.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Enterovirus/imunologia , Epitopos de Linfócito T/imunologia , Antígeno HLA-A2/imunologia , Peptídeos/imunologia , Infecções por Picornaviridae/imunologia , Proteínas Virais/imunologia , Linfócitos T CD8-Positivos/patologia , Feminino , Humanos , Masculino , Infecções por Picornaviridae/patologia
3.
Inflamm Res ; 71(3): 357-368, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35107605

RESUMO

OBJECTIVE: The maintenance of tight junction integrity contributes significantly to epithelial barrier function. If barrier function is destroyed, cell permeability increases and the movement of pathogens is promoted, further increasing the susceptibility to secondary infection. Here, we examined the protective effects of wogonin on rhinovirus (RV)-induced tight junction disruption. Additionally, we examined the signaling molecules responsible for anti-inflammatory activities in human nasal epithelial (HNE) cells. METHODS AND RESULTS: Primary HNE cells grown at an air-liquid interface and RPMI 2650 cells were infected apically with RV. Incubation with RV resulted in disruption of tight junction proteins (ZO-1, E-cadherin, claudin-1, and occludin) in the HNE cells. Cell viability of wogonin-treated HNE cells was measured using the MTT assay. Pretreatment with wogonin decreased RV-induced disruption of tight junctions in HNE cells. Furthermore, wogonin significantly decreased RV-induced phosphorylation of Akt/NF-κB and ERK1/2. Additionally, RV-induced generation of reactive oxygen species and RV-induced up-regulation of the production of inflammatory cytokines IL-8 and IL-6 were diminished by wogonin in HNE cells. CONCLUSION: Wogonin inhibits HRV-induced tight junction disruption via the suppression of inflammatory responses and phosphorylation of Akt/NF-κB and ERK1/2 in HNE cells. These finds will facilitate the development of novel therapeutic strategies.


Assuntos
Flavanonas , Proteína Quinase 3 Ativada por Mitógeno , NF-kappa B , Proteínas Proto-Oncogênicas c-akt , Rhinovirus , Junções Íntimas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Flavanonas/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Rhinovirus/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Junções Íntimas/patologia
4.
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
5.
Arch Virol ; 166(5): 1355-1370, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33709216

RESUMO

Porcine teschovirus (PTV) is a causative agent of reproductive disorders, encephalomyelitis, respiratory diseases, and diarrhea in swine, with a worldwide distribution. In this work, we identified PTV-associated nonsuppurative encephalitis as a potential cause of posterior paralysis in neonatal pigs in northeast China. Using indirect immunofluorescence assay, western blot, electron microscopy, and genome sequencing, we identified a neurotropic PTV strain, named CHN-NP1-2016, in the supernatants of pooled cerebrum and cerebellum samples from an affected piglet. Nucleotide sequence alignment revealed that the whole genome of CHN-NP1-2016 shared the highest sequence similarity (86.76% identity) with PTV 1 strain Talfan. A combination of phylogenetic and genetic divergence analysis was applied based on the deduced amino acid sequence of the P1 gene with a cutoff value of the genetic distance (0.102 ± 0.008) for defining PTV genotypes, and this showed that CHN-NP1-2016 is a variant of genotype 1. In total, 16 unique mutations and five mutant clusters were detected in the capsid proteins VP1 and VP2 of CHN-NP1-2016 when compared to other PTV1 isolates. Importantly, we detected three mutant clusters located in the exposed surface loops of the capsid protein, potentially indicating significant differences in major neutralization epitopes. Moreover, a potential recombination event in the P1 region of PTV CHN-NP1-2016 was detected. These findings provide valuable insights into the role of recombination in the evolution of teschoviruses. To our knowledge, this is the first case report of PTV-1-associated encephalitis in northeast China. Future investigations will narrow on the serology and pathogenicity of this novel isolate.


Assuntos
Encefalite Viral/veterinária , Infecções por Picornaviridae/veterinária , Doenças dos Suínos/virologia , Teschovirus/genética , Teschovirus/isolamento & purificação , Animais , Encéfalo/virologia , China/epidemiologia , Encefalite Viral/patologia , Encefalite Viral/virologia , Genoma Viral/genética , Genótipo , Mutação , Filogenia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , RNA Viral/genética , Recombinação Genética , Suínos , Teschovirus/classificação , Proteínas Virais/genética
6.
Arch Virol ; 166(11): 3105-3116, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34482448

RESUMO

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.


Assuntos
Vírus da Hepatite do Pato/patogenicidade , Hepatite Viral Animal/virologia , Interações Hospedeiro-Patógeno/genética , Infecções por Picornaviridae/veterinária , Doenças das Aves Domésticas/virologia , Aminoácidos/genética , Aminoácidos/metabolismo , Animais , Patos/virologia , Expressão Gênica , Hepatite Viral Animal/genética , Hepatite Viral Animal/metabolismo , Hepatite Viral Animal/patologia , Pâncreas/citologia , Pâncreas/patologia , Pâncreas/virologia , Pancreatite/patologia , Pancreatite/virologia , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/patologia , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA
7.
Pediatr Dev Pathol ; 24(5): 460-466, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754905

RESUMO

The Neuropathology of Human Parechovirus (HPeV) is not widely described due to the relatively recent discovery of the virus combined with a limited number of autopsy case reports. We report the case of an infant boy born at 38 weeks who, six days after birth, presented with fever and severe neurological dysfunction. Human Parechovirus Type 3 (HPeV3) RNA was detected in his cerebrospinal fluid (CSF) and blood. He died five days after his initial presentation. Neuropathologic examination demonstrated multicystic encephalomalacia (ME). This case report confirms that white matter pathology is dominant in HPeV3 infection. A unique feature, of HPeV encephalomalacia is absence of CSF pleocytosis and minimal inflammation in the meninges. The findings permit comment on the pathogenesis of brain injury by this virus.


Assuntos
Encefalomalacia/patologia , Encefalomalacia/virologia , Parechovirus , Infecções por Picornaviridae/patologia , Encefalomalacia/diagnóstico , Evolução Fatal , Humanos , Recém-Nascido , Masculino , Parechovirus/isolamento & purificação , Infecções por Picornaviridae/diagnóstico
8.
Proc Natl Acad Sci U S A ; 115(30): E7158-E7165, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-29987044

RESUMO

Rhinoviruses (RVs) are responsible for the majority of upper airway infections; despite their high prevalence and the resulting economic burden, effective treatment is lacking. We report here that RV induces metabolic alterations in host cells, which offer an efficient target for antiviral intervention. We show that RV-infected cells rapidly up-regulate glucose uptake in a PI3K-dependent manner. In parallel, infected cells enhance the expression of the PI3K-regulated glucose transporter GLUT1. In-depth metabolomic analysis of RV-infected cells revealed a critical role of glucose mobilization from extracellular and intracellular pools via glycogenolysis for viral replication. Infection resulted in a highly anabolic state, including enhanced nucleotide synthesis and lipogenesis. Consistently, we observed that glucose deprivation from medium and via glycolysis inhibition by 2-deoxyglucose (2-DG) potently impairs viral replication. Metabolomic analysis showed that 2-DG specifically reverts the RV-induced anabolic reprogramming. In addition, treatment with 2-DG inhibited RV infection and inflammation in a murine model. Thus, we demonstrate that the specific metabolic fingerprint of RV infection can be used to identify new targets for therapeutic intervention.


Assuntos
Infecções por Picornaviridae/metabolismo , Rhinovirus/fisiologia , Replicação Viral/fisiologia , Animais , Desoxiglucose/farmacologia , Feminino , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Lipogênese/efeitos dos fármacos , Lipogênese/genética , Camundongos , Nucleotídeos/biossíntese , Nucleotídeos/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/patologia , Replicação Viral/efeitos dos fármacos
9.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209576

RESUMO

Neurotropic viruses target the brain and contribute to neurologic diseases. Caspase recruitment domain containing family member 9 (CARD9) controls protective immunity in a variety of infectious disorders. To investigate the effect of CARD9 in neurotropic virus infection, CARD9-/- and corresponding C57BL/6 wild-type control mice were infected with Theiler's murine encephalomyelitis virus (TMEV). Brain tissue was analyzed by histology, immunohistochemistry and molecular analyses, and spleens by flow cytometry. To determine the impact of CARD9 deficiency on T cell responses in vitro, antigen presentation assays were utilized. Genetic ablation of CARD9 enhanced early pro-inflammatory cytokine responses and accelerated infiltration of T and B cells in the brain, together with a transient increase in TMEV-infected cells in the hippocampus. CARD9-/- mice showed an increased loss of neuronal nuclear protein+ mature neurons and doublecortin+ neuronal precursor cells and an increase in ß-amyloid precursor protein+ damaged axons in the hippocampus. No effect of CARD9 deficiency was found on the initiation of CD8+ T cell responses by flow cytometry and co-culture experiments using virus-exposed dendritic cells or microglia-enriched glial cell mixtures, respectively. The present study indicates that CARD9 is dispensable for the initiation of early antiviral responses and TMEV elimination but may contribute to the modulation of neuroinflammation, thereby reducing hippocampal injury following neurotropic virus infection.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/deficiência , Suscetibilidade a Doenças , Encefalite Viral/etiologia , Hipocampo/virologia , Infecções por Picornaviridae/etiologia , Picornaviridae/fisiologia , Animais , Biomarcadores , Modelos Animais de Doenças , Encefalite Viral/patologia , Predisposição Genética para Doença , Hipocampo/metabolismo , Hipocampo/patologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imuno-Histoquímica , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Infecções por Picornaviridae/patologia , Carga Viral
10.
Emerg Infect Dis ; 26(1): 176-178, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31855534

RESUMO

We detected bovine kobuvirus (BKV) in calves with diarrhea in the United States. The strain identified is related genetically to BKVs detected in other countries. Histopathologic findings also confirmed viral infection in 2 BKV cases. Our data show BKV is a potential causative agent for diarrhea in calves.


Assuntos
Doenças dos Bovinos/virologia , Diarreia/veterinária , Kobuvirus , Infecções por Picornaviridae/veterinária , Animais , Animais Recém-Nascidos/virologia , Bovinos , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/patologia , Diarreia/epidemiologia , Diarreia/virologia , Kobuvirus/genética , Filogenia , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Análise de Sequência de DNA , Estados Unidos/epidemiologia
11.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31434730

RESUMO

Senecavirus A (SVA) is a picornavirus that causes acute vesicular disease (VD), that is clinically indistinguishable from foot-and-mouth disease (FMD), in pigs. Notably, SVA RNA has been detected in lymphoid tissues of infected animals several weeks following resolution of the clinical disease, suggesting that the virus may persist in select host tissues. Here, we investigated the occurrence of persistent SVA infection and the contribution of stressors (transportation, immunosuppression, or parturition) to acute disease and recrudescence from persistent SVA infection. Our results show that transportation stress leads to a slight increase in disease severity following infection. During persistence, transportation, immunosuppression, and parturition stressors did not lead to overt/recrudescent clinical disease, but intermittent viremia and virus shedding were detected up to day 60 postinfection (p.i.) in all treatment groups following stress stimulation. Notably, real-time PCR and in situ hybridization (ISH) assays confirmed that the tonsil harbors SVA RNA during the persistent phase of infection. Immunofluorescence assays (IFA) specific for double-stranded RNA (dsRNA) demonstrated the presence of double-stranded viral RNA in tonsillar cells. Most importantly, infectious SVA was isolated from the tonsil of two animals on day 60 p.i., confirming the occurrence of carrier animals following SVA infection. These findings were supported by the fact that contact piglets (11/44) born to persistently infected sows were infected by SVA, demonstrating successful transmission of the virus from carrier sows to contact piglets. Results here confirm the establishment of persistent infection by SVA and demonstrate successful transmission of the virus from persistently infected animals.IMPORTANCE Persistent viral infections have significant implications for disease control strategies. Previous studies demonstrated the persistence of SVA RNA in the tonsil of experimentally or naturally infected animals long after resolution of the clinical disease. Here, we showed that SVA establishes persistent infection in SVA-infected animals, with the tonsil serving as one of the sites of virus persistence. Importantly, persistently infected carrier animals shedding SVA in oral and nasal secretions or feces can serve as sources of infection to other susceptible animals, as evidenced by successful transmission of SVA from persistently infected sows to contact piglets. These findings unveil an important aspect of SVA infection biology, suggesting that persistently infected pigs may function as reservoirs for SVA.


Assuntos
Portador Sadio/veterinária , Transmissão Vertical de Doenças Infecciosas/veterinária , Infecções por Picornaviridae/veterinária , Picornaviridae/patogenicidade , Doenças dos Suínos/transmissão , Animais , Portador Sadio/patologia , Portador Sadio/transmissão , Portador Sadio/virologia , Doença Crônica , Feminino , Tonsila Palatina/virologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/transmissão , Infecções por Picornaviridae/virologia , Recidiva , Estresse Fisiológico , Suínos , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia , Viremia/patologia , Viremia/transmissão , Viremia/veterinária , Viremia/virologia , Eliminação de Partículas Virais
12.
J Neurovirol ; 26(3): 449-451, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32026339

RESUMO

Human enteroviruses (EV) are the most common cause of viral meningitis in children. Human parechoviruses (HPeV) are increasingly being recognized as a cause of central nervous system (CNS) infections and sepsis-like disease in children. Both viruses belong to Picornaviridae family. The clinical picture in EV and HPeV infections is usually nonspecific. Therefore, molecular detection of both viruses is needed for etiological diagnosis. In this case report, we describe and discuss clinical and laboratory findings of two consecutive episodes of viral meningitis caused by EV and HPeV, respectively, occurring in the first month of a newborn's life.


Assuntos
Enterovirus Humano B/genética , Meningite Viral/diagnóstico , Parechovirus/genética , Infecções por Picornaviridae/diagnóstico , RNA Viral/genética , Sepse/diagnóstico , Enterovirus Humano B/classificação , Enterovirus Humano B/isolamento & purificação , Enterovirus Humano B/patogenicidade , Feminino , Humanos , Recém-Nascido , Meningite Viral/patologia , Meningite Viral/virologia , Parechovirus/classificação , Parechovirus/isolamento & purificação , Parechovirus/patogenicidade , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Recidiva , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sepse/patologia , Sepse/virologia , Análise de Sequência de DNA
13.
J Allergy Clin Immunol ; 143(1): 114-125.e4, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29698627

RESUMO

BACKGROUND: The innate immune system senses viral infection through pattern recognition receptors (PRRs), leading to type I interferon production. The role of type I interferon and PPRs in rhinovirus-induced asthma exacerbations in vivo are uncertain. OBJECTIVES: We sought to compare bronchial mucosal type I interferon and PRR expression at baseline and after rhinovirus infection in atopic asthmatic patients and control subjects. METHODS: Immunohistochemistry was used to detect expression of IFN-α, IFN-ß, and the PRRs: Toll-like receptor 3, melanoma differentiation-associated gene 5, and retinoic acid-inducible protein I in bronchial biopsy specimens from 10 atopic asthmatic patients and 15 nonasthmatic nonatopic control subjects at baseline and on day 4 and 6 weeks after rhinovirus infection. RESULTS: We observed IFN-α/ß deficiency in the bronchial epithelium at 3 time points in asthmatic patients in vivo. Lower epithelial IFN-α/ß expression was related to greater viral load, worse airway symptoms, airway hyperresponsiveness, and reductions in lung function during rhinovirus infection. We found lower frequencies of bronchial subepithelial monocytes/macrophages expressing IFN-α/ß in asthmatic patients during infection. Interferon deficiency at baseline was not accompanied by deficient PRR expression in asthmatic patients. Both epithelial and subepithelial PRR expression were induced during rhinovirus infection. Rhinovirus infection-increased numbers of subepithelial interferon/PRR-expressing inflammatory cells were related to greater viral load, airway hyperresponsiveness, and reductions in lung function. CONCLUSIONS: Bronchial epithelial IFN-α/ß expression and numbers of subepithelial IFN-α/ß-expressing monocytes/macrophages during infection were both deficient in asthmatic patients. Lower epithelial IFN-α/ß expression was associated with adverse clinical outcomes after rhinovirus infection in vivo. Increases in numbers of subepithelial cells expressing interferon/PRRs during infection were also related to greater viral load/illness severity.


Assuntos
Asma/imunologia , Proteína DEAD-box 58/imunologia , Regulação da Expressão Gênica/imunologia , Helicase IFIH1 Induzida por Interferon/biossíntese , Interferon-alfa/imunologia , Interferon beta/imunologia , Infecções por Picornaviridae/imunologia , Rhinovirus/imunologia , Receptor 3 Toll-Like/imunologia , Adulto , Asma/metabolismo , Asma/patologia , Biópsia , Brônquios/imunologia , Brônquios/metabolismo , Brônquios/patologia , Proteína DEAD-box 58/biossíntese , Feminino , Humanos , Helicase IFIH1 Induzida por Interferon/imunologia , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Masculino , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/patologia , Receptores Imunológicos , Rhinovirus/metabolismo , Índice de Gravidade de Doença , Receptor 3 Toll-Like/biossíntese
14.
Clin Microbiol Rev ; 31(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29142080

RESUMO

Human parechovirus (HPeV) is increasingly being recognized as a potentially severe viral infection in neonates and young infants. HPeV belongs to the family Picornaviridae and is currently divided into 19 genotypes. HPeV-1 is the most prevalent genotype and most commonly causes gastrointestinal and respiratory disease. HPeV-3 is clinically the most important genotype due to its association with severe disease in younger infants, which may partly be explained by its distinct virological properties. In young infants, the typical clinical presentation includes fever, severe irritability, and rash, often leading to descriptions of "hot, red, angry babies." Infants with severe central nervous system (CNS) infections are at an increased risk of long-term sequelae. Considering the importance of HPeV as a cause of severe viral infections in young infants, we recommend that molecular diagnostic techniques for early detection be included in the standard practice for the investigation of sepsis-like illnesses and CNS infections in this age group.


Assuntos
Parechovirus , Infecções por Picornaviridae/diagnóstico , Infecções por Picornaviridae/virologia , Sepse/diagnóstico , Sepse/virologia , Humanos , Técnicas de Diagnóstico Molecular/tendências , Parechovirus/genética , Infecções por Picornaviridae/patologia
15.
Am J Physiol Lung Cell Mol Physiol ; 317(1): L57-L70, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30908938

RESUMO

Asthma exacerbations are often caused by rhinovirus (RV). We and others have shown that Toll-like receptor 2 (TLR2), a membrane surface receptor that recognizes bacterial lipopeptides and lipoteichoic acid, is required and sufficient for RV-induced proinflammatory responses in vitro and in vivo. We hypothesized that viral protein-4 (VP4), an internal capsid protein that is myristoylated upon viral replication and externalized upon viral binding, is a ligand for TLR2. Recombinant VP4 and myristoylated VP4 (MyrVP4) were purified by Ni-affinity chromatography. MyrVP4 was also purified from RV-A1B-infected HeLa cells by urea solubilization and anti-VP4 affinity chromatography. Finally, synthetic MyrVP4 was produced by chemical peptide synthesis. MyrVP4-TLR2 interactions were assessed by confocal fluorescence microscopy, fluorescence resonance energy transfer (FRET), and monitoring VP4-induced cytokine mRNA expression in the presence of anti-TLR2 and anti-VP4. MyrVP4 and TLR2 colocalized in TLR2-expressing HEK-293 cells, mouse bone marrow-derived macrophages, human bronchoalveolar macrophages, and human airway epithelial cells. Colocalization was absent in TLR2-null HEK-293 cells and blocked by anti-TLR2 and anti-VP4. Cy3-labeled MyrVP4 and Cy5-labeled anti-TLR2 showed an average fractional FRET efficiency of 0.24 ± 0.05, and Cy5-labeled anti-TLR2 increased and unlabeled MyrVP4 decreased FRET efficiency. MyrVP4-induced chemokine mRNA expression was higher than that elicited by VP4 alone and was attenuated by anti-TLR2 and anti-VP4. Cytokine expression was similarly increased by MyrVP4 purified from RV-infected HeLa cells and synthetic MyrVP4. We conclude that, during RV infection, MyrVP4 and TLR2 interact to generate a proinflammatory response.


Assuntos
Asma/genética , Proteínas do Capsídeo/genética , Eosinofilia/genética , Infecções por Picornaviridae/genética , Processamento de Proteína Pós-Traducional , Receptor 2 Toll-Like/genética , Proteínas Virais/genética , Adolescente , Sequência de Aminoácidos , Animais , Asma/imunologia , Asma/patologia , Asma/virologia , Proteínas do Capsídeo/imunologia , Criança , Eosinofilia/imunologia , Eosinofilia/patologia , Eosinofilia/virologia , Células Epiteliais/imunologia , Células Epiteliais/virologia , Feminino , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácidos Mirísticos/imunologia , Ácidos Mirísticos/metabolismo , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Ligação Proteica , Rhinovirus/imunologia , Rhinovirus/patogenicidade , Transdução de Sinais , Receptor 2 Toll-Like/imunologia , Proteínas Virais/imunologia , Replicação Viral
16.
J Virol ; 92(8)2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29367253

RESUMO

Positive-strand RNA viruses, including picornaviruses, utilize cellular machinery for genome replication. Previously, we reported that each of the 2B, 2BC, 2C, 3A, and 3AB proteins of Aichi virus (AiV), a picornavirus, forms a complex with the Golgi apparatus protein ACBD3 and phosphatidylinositol 4-kinase IIIß (PI4KB) at viral RNA replication sites (replication organelles [ROs]), enhancing PI4KB-dependent phosphatidylinositol 4-phosphate (PI4P) production. Here, we demonstrate AiV hijacking of the cellular cholesterol transport system involving oxysterol-binding protein (OSBP), a PI4P-binding cholesterol transfer protein. AiV RNA replication was inhibited by silencing cellular proteins known to be components of this pathway, OSBP, the ER membrane proteins VAPA and VAPB (VAP-A/B), the PI4P-phosphatase SAC1, and PI-transfer protein ß. OSBP, VAP-A/B, and SAC1 were present at RNA replication sites. We also found various previously unknown interactions among the AiV proteins (2B, 2BC, 2C, 3A, and 3AB), ACBD3, OSBP, VAP-A/B, and SAC1, and the interactions were suggested to be involved in recruiting the component proteins to AiV ROs. Importantly, the OSBP-2B interaction enabled PI4P-independent recruitment of OSBP to AiV ROs, indicating preferential recruitment of OSBP among PI4P-binding proteins. Protein-protein interaction-based OSBP recruitment has not been reported for other picornaviruses. Cholesterol was accumulated at AiV ROs, and inhibition of OSBP-mediated cholesterol transfer impaired cholesterol accumulation and AiV RNA replication. Electron microscopy showed that AiV-induced vesicle-like structures were close to ER membranes. Altogether, we conclude that AiV directly recruits the cholesterol transport machinery through protein-protein interactions, resulting in formation of membrane contact sites between the ER and AiV ROs and cholesterol supply to the ROs.IMPORTANCE Positive-strand RNA viruses utilize host pathways to modulate the lipid composition of viral RNA replication sites for replication. Previously, we demonstrated that Aichi virus (AiV), a picornavirus, forms a complex comprising certain proteins of AiV, the Golgi apparatus protein ACBD3, and the lipid kinase PI4KB to synthesize PI4P lipid at the sites for AiV RNA replication. Here, we confirmed cholesterol accumulation at the AiV RNA replication sites, which are established by hijacking the host cholesterol transfer machinery mediated by a PI4P-binding cholesterol transfer protein, OSBP. We showed that the component proteins of the machinery, OSBP, VAP, SAC1, and PITPNB, are all essential host factors for AiV replication. Importantly, the machinery is directly recruited to the RNA replication sites through previously unknown interactions of VAP/OSBP/SAC1 with the AiV proteins and with ACBD3. Consequently, we propose a specific strategy employed by AiV to efficiently accumulate cholesterol at the RNA replication sites via protein-protein interactions.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Colesterol/metabolismo , Kobuvirus/fisiologia , Proteínas de Membrana/metabolismo , Modelos Biológicos , Infecções por Picornaviridae/metabolismo , RNA Viral/metabolismo , Receptores de Esteroides/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Colesterol/genética , Humanos , Proteínas de Membrana/genética , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/patologia , RNA Viral/genética , Receptores de Esteroides/genética , Proteínas de Transporte Vesicular/genética , Proteínas Virais/genética
17.
Clin Exp Allergy ; 49(12): 1587-1597, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31400236

RESUMO

BACKGROUND: The temporal in vivo response of epithelial cells to a viral challenge and its association with viral clearance and clinical outcomes has been largely unexplored in asthma. OBJECTIVE: To determine gene expression profiles over time in nasal epithelial cells (NECs) challenged in vivo with rhinovirus-16 (RV16) and compare to nasal symptoms and viral clearance. METHODS: Patients with stable mild to moderate asthma (n = 20) were challenged intranasally with RV16. Nasal brush samples for RNA sequencing were taken 7 days prior to infection and 3, 6 and 14 days post-infection, and blood samples 4 days prior to infection and day 6 post-infection. Viral load was measured in nasal lavage fluid at day 3, 6 and 14. RESULTS: Top differentially (>2.5-fold increase) expressed gene sets in NECs post-RV16 at days 3 and 6, compared with baseline, were interferon alpha and gamma response genes. Patients clearing the virus within 6 days (early resolvers) had a significantly increased interferon response at day 6, whereas those having cleared the virus by day 14 (late resolvers) had significantly increased responses at day 3, 6 and 14. Interestingly, patients not having cleared the virus by day 14 (non-resolvers) had no enhanced interferon responses at any of these days. The daily Cold Symptom Scores (CSS) peaked at days 3 to 5 and correlated positively with interferon response genes at day 3 (R = 0.48), but not at other time-points. Interferon response genes were also enhanced in blood at day 6 after RV16 challenge. CONCLUSION AND CLINICAL RELEVANCE: This study shows that viral load and clearance varies markedly over time in mild to moderate asthma patients exposed to a fixed RV16 dose. The host's nasal interferon response to RV16 at day 3 is associated with upper respiratory tract symptoms. The temporal interferon response in nasal epithelium associates with viral clearance in the nasal compartment.


Assuntos
Asma , Brônquios , Interferon-alfa/imunologia , Interferon gama/imunologia , Mucosa Nasal , Infecções por Picornaviridae , Rhinovirus/imunologia , Adulto , Asma/imunologia , Asma/patologia , Asma/virologia , Brônquios/imunologia , Brônquios/patologia , Brônquios/virologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/imunologia , Mucosa Nasal/patologia , Mucosa Nasal/virologia , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/patologia
18.
Microb Pathog ; 136: 103695, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31449854

RESUMO

Seneca Valley virus (SVV) has been identified as the causative agent of SVV-associated vesicular disease (SAVD). To investigate the pathogenicity of two newly isolated SVV strains (GD-S5/2018 and GD04/2017) in China, experimental infections of pigs were performed. In pig experiments, both SVV strains successfully infected all animals, evidenced by presence of virus shedding and robust protective antibody responses. SVV GD-S5/2018 infection resulted in characteristic clinical signs, and ulcerative lesions on the tongue and gums. However, SVV GD04/2017 did not cause any clinical symptoms except depression in pigs during the experiment. Taken together, these results demonstrate that SVV GD-S5/2018 is a virulent strain for pigs, whereas SVV GD04/2017 is nearly avirulent. The established animal models for SVV infection will be utilized to dissect the immunity and pathogenesis, and develop vaccines and antivirals.


Assuntos
Infecções por Picornaviridae/veterinária , Picornaviridae/patogenicidade , Doença Vesicular Suína/patologia , Doença Vesicular Suína/virologia , Animais , China , Picornaviridae/isolamento & purificação , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Suínos , Virulência
19.
Microb Pathog ; 127: 320-325, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30529427

RESUMO

The aim of the present study was to pathological and molecular investigation of porcine sapelovirus (PSV) in naturally infected Indian pigs of various age groups. Eight samples (16%) out of 49 necropsied animals were positive for PSV on the basis of pathological and molecular investigation. Major lesions of PSV positive cases were thickening and clouding of meninges, congestion in brain, severe to moderate congestion in lungs along with froathy exudates in trachea, thickening of intestinal mucosa, especially mucosal folds of ileum. Microscopic lesions of PSV positive cases in CNS were perivascular cuffing, neuronophagia and focal gliosis. In lungs, interstitial pneumonia was noticed in all cases, and intestinal lesions comprised of sloughing of villi epithelium, moderate to severe congestion of blood vessels and infiltration of mononuclear cells mainly plasma cells in both large and small intestine. RT-PCR results of total cases examined for PSV were targeted for PSV 3D Polymerase, 5'UTR region and VP1 gene respectively. Genetic characterization was done on the basis of viral capsid protein 1 (VP1) gene of PSV. The sequencing and phylogenetic analysis of amplified VP1 gene product showed maximum identity 85-90% with South Korean, KJ821021.1 and Indian, KY053835.1 strain of PSV. Further explorative surveillance and epidemiological studies are suggested to find out the real impact of this economically important disease affecting pigs population of India.


Assuntos
Infecções por Picornaviridae/veterinária , Picornaviridae/isolamento & purificação , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia , Estruturas Animais/patologia , Estruturas Animais/virologia , Animais , Histocitoquímica , Índia , Filogenia , Picornaviridae/classificação , Picornaviridae/genética , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Suínos
20.
Clin Sci (Lond) ; 133(8): 983-996, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-30952808

RESUMO

Rhinovirus (RV), which is associated with acute exacerbations, also causes persistent lung inflammation in patients with chronic obstructive pulmonary disease (COPD), but the underlying mechanisms are not well-known. Recently, we demonstrated that RV causes persistent lung inflammation with accumulation of a subset of macrophages (CD11b+/CD11c+), and CD8+ T cells, and progression of emphysema. In the present study, we examined the mechanisms underlying the RV-induced persistent inflammation and progression of emphysema in mice with COPD phenotype. Our results demonstrate that at 14 days post-RV infection, in addition to sustained increase in CCL3, CXCL-10 and IFN-γ expression as previously observed, levels of interleukin-33 (IL-33), a ligand for ST2 receptor, and matrix metalloproteinase (MMP)12 are also elevated in mice with COPD phenotype, but not in normal mice. Further, MMP12 was primarily expressed in CD11b+/CD11c+ macrophages. Neutralization of ST2, reduced the expression of CXCL-10 and IFN-γ and attenuated accumulation of CD11b+/CD11c+ macrophages, neutrophils and CD8+ T cells in COPD mice. Neutralization of IFN-γ, or ST2 attenuated MMP12 expression and prevented progression of emphysema in these mice. Taken together, our results indicate that RV may stimulate expression of CXCL-10 and IFN-γ via activation of ST2/IL-33 signaling axis, which in turn promote accumulation of CD11b+/CD11c+ macrophages and CD8+ T cells. Furthermore, RV-induced IFN-γ stimulates MMP12 expression particularly in CD11b+/CD11c+ macrophages, which may degrade alveolar walls thus leading to progression of emphysema in these mice. In conclusion, our data suggest an important role for ST2/IL-33 signaling axis in RV-induced pathological changes in COPD mice.


Assuntos
Proteína 1 Semelhante a Receptor de Interleucina-1/imunologia , Interleucina-33/imunologia , Infecções por Picornaviridae/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Rhinovirus/fisiologia , Animais , Quimiocina CCL3/genética , Quimiocina CCL3/imunologia , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Interferon gama/genética , Interferon gama/imunologia , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Interleucina-33/genética , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Doença Pulmonar Obstrutiva Crônica/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA