Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.311
Filtrar
1.
Nat Commun ; 15(1): 3469, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38658526

RESUMO

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.


Assuntos
Parechovirus , Infecções por Picornaviridae , Internalização do Vírus , Parechovirus/genética , Parechovirus/metabolismo , Humanos , Infecções por Picornaviridae/virologia , Infecções por Picornaviridae/metabolismo , Sistemas CRISPR-Cas , Ligação Proteica , Receptores Virais/metabolismo , Receptores Virais/genética , Linhagem Celular , Células HEK293 , Organoides/virologia , Organoides/metabolismo
2.
Infect Genet Evol ; 120: 105585, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38508364

RESUMO

In this study, a picornavirus and a nidovirus were identified from a single available nasopharyngeal swab (NPS) sample of a freshly deceased sheep, as the only vertebrate viruses found with viral metagenomics and next-generation sequencing methods. The sample was originated from a mixed feedlot farm in Hungary where sheep and cattle were held together but in separate stalls. Most of the sheep had respiratory signs (coughing and increased respiratory effort) at the time of sampling. Other NPS were not, but additional enteric samples were collected from sheep (n = 27) and cattle (n = 11) of the same farm at that time. The complete/nearly complete genomes of the identified viruses were determined using RT-PCR and Nanopore (MinION-Flonge) / Dye-terminator sequencing techniques. The results of detailed genomic and phylogenetic analyses indicate that the identified picornavirus most likely belongs to a type 4 genotype of species Bovine rhinitis B virus (BRBV-4, OR885914) of genus Aphthovirus, family Picornaviridae while the ovine nidovirus (OvNV, OR885915) - as a novel variant - could belong to the recently created Bovine nidovirus 1 (BoNV) species of genus Bostovirus, family Tobaniviridae. None of the identified viruses were detectable in the enteric samples using RT-PCR and generic screening primer pairs. Both viruses are well-known respiratory pathogens of cattle, but their presence was not demonstrated before in other animals, like sheep. Furthermore, neither BRBV-4 nor BoNVs were investigated in European cattle and/or sheep flocks, therefore it cannot be determined whether the presence of these viruses in sheep was a result of a single host species switch/spillover event or these viruses are circulating in not just cattle but sheep populations as well. Further studies required to investigate the spread of these viruses in Hungarian and European sheep and cattle populations and to identify their pathogenic potential in sheep.


Assuntos
Filogenia , Infecções por Picornaviridae , Picornaviridae , Doenças dos Ovinos , Animais , Hungria , Picornaviridae/genética , Picornaviridae/isolamento & purificação , Picornaviridae/classificação , Ovinos , Doenças dos Ovinos/virologia , Bovinos , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/virologia , Coinfecção/virologia , Coinfecção/veterinária , Genoma Viral , Nidovirales/genética , Nidovirales/isolamento & purificação , Nidovirales/classificação , Infecções por Nidovirales/veterinária , Infecções por Nidovirales/virologia
3.
J Virol ; 97(8): e0060423, 2023 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-37555661

RESUMO

Viruses have evolved diverse strategies to evade the host innate immune response and promote infection. The retinoic acid-inducible gene I (RIG-I)-like receptors RIG-I and MDA5 are antiviral factors that sense viral RNA and trigger downstream signal via mitochondrial antiviral-signaling protein (MAVS) to activate type I interferon expression. 14-3-3ε is a key component of the RIG-I translocon complex that interacts with MAVS at the mitochondrial membrane; however, the exact role of 14-3-3ε in this pathway is not well understood. In this study, we demonstrate that 14-3-3ε is a direct substrate of both the poliovirus and coxsackievirus B3 (CVB3) 3C proteases (3Cpro) and that it is cleaved at Q236↓G237, resulting in the generation of N- and C-terminal fragments of 27.0 and 2.1 kDa, respectively. While the exogenous expression of wild-type 14-3-3ε enhances IFNB mRNA production during poly(I:C) stimulation, expression of the truncated N-terminal fragment does not. The N-terminal 14-3-3ε fragment does not interact with RIG-I in co-immunoprecipitation assays, nor can it facilitate RIG-I translocation to the mitochondria. Probing the intrinsically disordered C-terminal region identifies key residues responsible for the interaction between 14-3-3ε and RIG-I. Finally, overexpression of the N-terminal fragment promotes CVB3 infection in mammalian cells. The strategic enterovirus 3Cpro-mediated cleavage of 14-3-3ε antagonizes RIG-I signaling by disrupting critical interactions within the RIG-I translocon complex, thus contributing to evasion of the host antiviral response. IMPORTANCE Host antiviral factors work to sense virus infection through various mechanisms, including a complex signaling pathway known as the retinoic acid-inducible gene I (RIG-I)-like receptor pathway. This pathway drives the production of antiviral molecules known as interferons, which are necessary to establish an antiviral state in the cellular environment. Key to this antiviral signaling pathway is the small chaperone protein 14-3-3ε, which facilitates the delivery of a viral sensor protein, RIG-I, to the mitochondria. In this study, we show that the enteroviral 3C protease cleaves 14-3-3ε during infection, rendering it incapable of facilitating this antiviral response. We also find that the resulting N-terminal cleavage fragment dampens RIG-I signaling and promotes virus infection. Our findings reveal a novel viral strategy that restricts the antiviral host response and provides insights into the mechanisms underlying 14-3-3ε function in RIG-I antiviral signaling.


Assuntos
Infecções por Picornaviridae , Picornaviridae , Animais , Cisteína Endopeptidases/metabolismo , Proteína DEAD-box 58/metabolismo , Imunidade Inata , Mamíferos , Peptídeo Hidrolases/metabolismo , Picornaviridae/metabolismo , Transdução de Sinais , Tretinoína , Proteínas Virais/metabolismo , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/virologia , Proteases Virais 3C
5.
Front Immunol ; 13: 792716, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35173718

RESUMO

Prematurity and bronchopulmonary dysplasia (BPD) increase the risk of asthma later in life. Supplemental oxygen therapy is a risk factor for chronic respiratory symptoms in infants with BPD. Hyperoxia induces cell injury and release of damage-associated molecular patterns (DAMPs). Cytoskeletal filamentous actin (F-actin) is a DAMP which binds Clec9a, a C-type lectin selectively expressed on CD103+ dendritic cells (DCs). Co-stimulation of Clec9a and TLR3 induces maximal proinflammatory responses. We have shown that neonatal hyperoxia (a model of BPD) increases lung IL-12+Clec9a+CD103+ DCs, pro-inflammatory responses and airway hyperreactivity following rhinovirus (RV) infection. CD103+ DCs and Clec9a are required for these responses. Hyperoxia increases F-actin levels in bronchoalveolar lavage fluid (BALF). We hypothesized that the F-actin severing protein gelsolin attenuates neonatal hyperoxia-induced Clec9a+CD103+ DC-dependent pro-inflammatory responses to RV and preserves alveolarization. We exposed neonatal mice to hyperoxia and treated them with gelsolin intranasally. Subsequently we inoculated the mice with RV intranasally. Alternatively, we inoculated normoxic neonatal mice with BALF from hyperoxia-exposed mice (hyperoxic BALF), RV and gelsolin. We analyzed lung gene expression two days after RV infection. For in vitro studies, lung CD11c+ cells were isolated from C57BL/6J or Clec9agfp-/- mice and incubated with hyperoxic BALF and RV. Cells were analyzed by flow cytometry. In neonatal mice, gelsolin blocked hyperoxia-induced Il12p40, TNF-α and IFN-γ mRNA and protein expression in response to RV infection. Similar effects were observed when gelsolin was co-administered with hyperoxic BALF and RV. Gelsolin decreased F-actin levels in hyperoxic BALF in vitro and inhibited hyperoxia-induced D103lo DC expansion and inflammation in vivo. Gelsolin also attenuated hyperoxia-induced hypoalveolarization. Further, incubation of lung CD11c+ cells from WT and Clec9agfp-/- mice with hyperoxic BALF and RV, showed Clec9a is required for maximal hyperoxic BALF and RV induced IL-12 expression in CD103+ DCs. Finally, in tracheal aspirates from mechanically ventilated human preterm infants the F-actin to gelsolin ratio positively correlates with FiO2, and gelsolin levels decrease during the first two weeks of mechanical ventilation. Collectively, our findings demonstrate a promising role for gelsolin, administered by inhalation into the airway to treat RV-induced exacerbations of BPD and prevent chronic lung disease.


Assuntos
Displasia Broncopulmonar/tratamento farmacológico , Gelsolina/administração & dosagem , Hiperóxia/fisiopatologia , Lectinas Tipo C/metabolismo , Infecções por Picornaviridae/tratamento farmacológico , Receptores Imunológicos/metabolismo , Administração por Inalação , Animais , Animais Recém-Nascidos/metabolismo , Antígenos CD/metabolismo , Displasia Broncopulmonar/virologia , Feminino , Humanos , Recém-Nascido , Cadeias alfa de Integrinas/metabolismo , Interleucina-12/metabolismo , Lectinas Tipo C/genética , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxigenoterapia/efeitos adversos , Infecções por Picornaviridae/virologia , Receptores Imunológicos/genética , Testes de Função Respiratória , Rhinovirus/isolamento & purificação
6.
J Gen Virol ; 103(2)2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35138239

RESUMO

A novel picornavirus was isolated from the faeces of a diarrhoeic cow using MA-104 cells at the third blind passage. This virus, named Den1/2021/JPN, was completely sequenced using total RNA from the cell culture supernatant by deep sequencing. The genome of Den1/2021/JPN had a standard picornavirus genome organisation with conserved picornaviral motifs. The 5' untranslated region harboured a type-II internal ribosomal entry site. Den1/2021/JPN was most closely related to a bovine parechovirus (Bo_ParV) named cow/2018/4, which has been recently identified in publicly available databases. Phylogenetic analyses and pairwise sequence comparison revealed that Den1/2021/JPN and Bo_ParV cow/2018/4 clustered with parechoviruses and were most closely related to Parechovirus E identified in birds of prey, exhibiting nucleotide sequence similarity of 64.2-64.5 %, 58.6-59.7 % and 66.3-66.4 % in the polyprotein, P1 and 2C+3 CD coding regions, respectively. This study presents the first report on the isolation of Bo_ParV. Den1/2021/JPN and Bo_ParV cow/2018/4, which are candidates for a novel species in the genus Parechovirus.


Assuntos
Fezes/virologia , Genoma Viral , Parechovirus/isolamento & purificação , Infecções por Picornaviridae , RNA Viral , Animais , Bovinos , Japão , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/virologia
7.
Viruses ; 14(2)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35215935

RESUMO

Porcine sapelovirus (PSV) is an important emerging pathogen associated with a wide variety of diseases in swine, including acute diarrhoea, respiratory distress, skin lesions, severe neurological disorders, and reproductive failure. Although PSV is widespread, serological assays for field-based epidemiological studies are not yet available. Here, four PSV strains were recovered from diarrheic piglets, and electron microscopy revealed virus particles with a diameter of ~32 nm. Analysis of the entire genome sequence revealed that the genomes of PSV isolates ranged 7569-7572 nucleotides in length. Phylogenetic analysis showed that the isolated viruses were classified together with strains from China. Additionally, monoclonal antibodies for the recombinant PSV-VP1 protein were developed to specifically detect PSV infection in cells, and we demonstrated that isolated PSVs could only replicate in cells of porcine origin. Using recombinant PSV-VP1 protein as the coating antigen, we developed an indirect ELISA for the first time for the detection of PSV antibodies in serum. A total of 516 swine serum samples were tested, and PSV positive rate was 79.3%. The virus isolates, monoclonal antibodies and indirect ELISA developed would be useful for further understanding the pathophysiology of PSV, developing new diagnostic assays, and investigating the epidemiology of the PSV.


Assuntos
Infecções por Picornaviridae/veterinária , Picornaviridae/genética , Picornaviridae/isolamento & purificação , Doenças dos Suínos/virologia , Animais , Anticorpos Antivirais/sangue , Sequência de Bases , China , Fezes/virologia , Variação Genética , Genoma Viral , Filogenia , Picornaviridae/classificação , Picornaviridae/fisiologia , Infecções por Picornaviridae/sangue , Infecções por Picornaviridae/virologia , Suínos , Doenças dos Suínos/sangue , Replicação Viral , Sequenciamento Completo do Genoma
9.
Pediatr Infect Dis J ; 41(3): e95-e101, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35001055

RESUMO

BACKGROUND: The clinical impact of common human coronavirus (cHCoV) remains unclear. We studied the clinical manifestations of pediatric cHCoV infections and the possible modifying effects of codetected human rhinovirus (RV) and respiratory syncytial virus (RSV). METHODS: We used data from an 11-year-long prospective study of hospitalized children with community-acquired respiratory tract infections. Nasopharyngeal aspirates were analyzed with real-time polymerase chain reaction assay for cHCoV OC43, NL63, HKU1 and 229E, and 15 other respiratory viruses. We assessed disease severity based on the clinical factors hospitalization length, oxygen requirement, other respiratory support and supplementary fluids. RESULTS: cHCoV was detected in 341 (8%) of 4312 children. Among 104 children with single cHCoV detections, 58 (56%) had lower respiratory tract infection (LRTI) and 20 (19%) developed severe disease. The proportion with severe disease was lower among single cHCoV detections compared with single RSV detections (338 of 870; 39%), but similar to single RV detections (136 of 987; 14%). Compared with single cHCoV, codetected cHCoV-RSV was more often associated with LRTI (86 of 89; 97%) and severe disease (adjusted odds ratio, 3.3; 95% confidence interval: 1.6-6.7). LRTI was more frequent in codetected cHCoV-RV (52 of 68; 76%) than single cHCoV, but the risk of severe disease was lower (adjusted odds ratios, 0.3; 95% confidence interval: 0.1-1.0). CONCLUSIONS: cHCoV was associated with severe LRTI in hospitalized children. Viral codetections were present in two-thirds. Codetections of cHCoV-RV were associated with lower proportions of severe disease, suggesting a modifying effect of RV on HCoV.


Assuntos
Coinfecção/virologia , Infecções por Coronavirus/virologia , Infecções por Picornaviridae/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Adolescente , Criança , Criança Hospitalizada , Pré-Escolar , Coinfecção/epidemiologia , Coinfecção/terapia , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/terapia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Noruega/epidemiologia , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/terapia , Estudos Prospectivos , Infecções por Vírus Respiratório Sincicial/epidemiologia , Infecções por Vírus Respiratório Sincicial/terapia
10.
Viruses ; 14(1)2022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35062345

RESUMO

Rhinoviruses (RVs) have been reported as one of the main viral causes for severe respiratory illnesses that may require hospitalization, competing with the burden of other respiratory viruses such as influenza and RSV in terms of severity, economic cost, and resource utilization. With three species and 169 subtypes, RV presents the greatest diversity within the Enterovirus genus, and despite the efforts of the research community to identify clinically relevant subtypes to target therapeutic strategies, the role of species and subtype in the clinical outcomes of RV infection remains unclear. This review aims to collect and organize data relevant to RV illness in order to find patterns and links with species and/or subtype, with a specific focus on species and subtype diversity in clinical studies typing of respiratory samples.


Assuntos
Infecções por Picornaviridae/virologia , Rhinovirus/classificação , Rhinovirus/genética , Asma/etiologia , Coinfecção/virologia , Enterovirus , Infecções por Enterovirus/virologia , Técnicas de Genotipagem , Hospitalização , Humanos , Infecções Respiratórias/virologia , Sorotipagem
11.
Mol Immunol ; 142: 11-21, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34959069

RESUMO

Senecavirus A (SVA) is an oncolytic virus, which can propagate in human tumor cells and has been used as an oncolytic virotherapy candidate in humans. Besides, SVA circulates in pigs and causes vesicles and coalescing erosions on the snouts and coronary bands in infected pigs and results in neonatal morbidity. SVA has evolved the ability to suppress host innate immune response to benefit viral replication. SVA 3Cpro and 2C protein inhibit the production of host type I interferon (IFN) by degradation of several components of innate immune pathway. In this study, for the first time, we determined that SVA 2B antagonized host innate immune response in both human and porcine cells. SVA 2B protein degraded mitochondrial antiviral-signaling protein (MAVS), a key host molecule in the innate immune pathway, and a colocalization and interaction between 2B and MAVS was observed in the context of viral infection. Further study showed that the 1-48 and 100-128 regions of 2B were essential for inhibition of type I IFN expression. In addition, we determined that 2B degraded MAVS depending on caspase-9 and caspase-3. In conclusion, our results revealed a novel strategy for SVA 2B protein to antagonize host innate immune response, which will help for clarification of the pathogenesis of SVA and provide an insight for oncolytic virotherapy of SVA.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Imunidade Inata/imunologia , Interferon Tipo I/biossíntese , Picornaviridae/metabolismo , Proteínas não Estruturais Virais/imunologia , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferon Tipo I/imunologia , Terapia Viral Oncolítica/métodos , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Suínos , Proteínas não Estruturais Virais/genética , Proteínas Viroporinas/genética , Proteínas Viroporinas/imunologia , Liberação de Vírus/fisiologia , Replicação Viral/fisiologia
12.
J Virol ; 96(2): e0155021, 2022 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-34757844

RESUMO

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.


Assuntos
Proteases Virais 3C/metabolismo , Autofagia , Proteínas do Capsídeo/metabolismo , Picornaviridae/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Picornaviridae/metabolismo , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/virologia , Proteína Sequestossoma-1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Replicação Viral
13.
J Med Virol ; 94(2): 461-468, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34415627

RESUMO

The burden of pneumonia, especially that caused by respiratory viruses, is markedly high in the pediatric age group. This study aimed to assess viral agents causing severe pneumonia among mechanically ventilated patients. Nonbronchoscopic bronchoalveolar lavage was performed for pediatric patients having severe pneumonia indicated for mechanical ventilation to be tested with a multiplex PCR immediate diagnosis of their etiologic pathogen. Among the 75 patients recruited, viral agents were detected in 73.4% of cases. Rhinovirus and respiratory syncytial virus (RSV) were the most common viruses detected in 32.1% and 29.5%, respectively. The rate of viral infection showed a clear increased incidence in the winter season. The mortality rate among viral-associated severe pneumonia reached 56.36%. Odds of mortality increased threefolds in presence of comorbid conditions and 10-folds with congenital heart disease. The study demonstrated the neglected importance of rhinovirus besides RSV in causing severe critical pneumonia in the pediatric age.


Assuntos
Infecções por Picornaviridae/virologia , Pneumonia Viral/virologia , Respiração Artificial , Infecções por Vírus Respiratório Sincicial/virologia , Vírus/isolamento & purificação , Adolescente , Líquido da Lavagem Broncoalveolar/virologia , Criança , Pré-Escolar , Estudos Transversais , Egito , Feminino , Cardiopatias Congênitas/complicações , Humanos , Lactente , Modelos Logísticos , Masculino , Reação em Cadeia da Polimerase Multiplex , Infecções por Picornaviridae/diagnóstico , Pneumonia Viral/mortalidade , Infecções por Vírus Respiratório Sincicial/diagnóstico , Vírus Sincicial Respiratório Humano/genética , Sistema Respiratório , Rhinovirus/genética , Estações do Ano , Vírus/genética
14.
Nutrients ; 13(12)2021 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-34959949

RESUMO

Acute respiratory infections are an important health concern. Traditionally, polysaccharide-enriched extracts from plants, containing immunomodulatory rhamnogalacturonan-I (RG-1), were used prophylactically. We established the effects of dietary supplementation with carrot-derived RG-I (cRG-I, 0-0.3-1.5 g/day) in 177 healthy individuals (18-65 years) on symptoms following infection with rhinovirus strain 16 (RV16). Primary outcomes were changes in severity and duration of symptoms, and viral load in nasal lavage. Secondary outcomes were changes in innate immune and anti-viral responses, reflected by CXCL10 and CXCL8 levels and cell differentials in nasal lavage. In a nested cohort, exploratory transcriptome analysis was conducted on nasal epithelium. Intake of cRG-I was safe, well-tolerated and accelerated local cellular and humoral innate immune responses induced by RV16 infection, with the strongest effects at 1.5 g/d. At 0.3 g/d, a faster interferon-induced response, induction of the key anti-viral gene EIF2AK2, faster viral clearance, and reduced symptom severity (-20%) and duration (-25%) were observed. Anti-viral responses, viral clearance and symptom scores at 1.5 g/d were in between those of 0 and 0.3 g/d, suggesting a negative feedback loop preventing excessive interferon responses. Dietary intake of cRG-I accelerated innate immune and antiviral responses, and reduced symptoms of an acute respiratory viral infection.


Assuntos
Antivirais , Quimiocina CXCL10/metabolismo , Daucus carota/química , Suplementos Nutricionais , Imunidade Inata/efeitos dos fármacos , Interleucina-8/metabolismo , Pectinas/farmacologia , Pectinas/uso terapêutico , Fitoterapia , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/virologia , Rhinovirus , Adolescente , Adulto , Idoso , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Lavagem Nasal , Gravidade do Paciente , Pectinas/isolamento & purificação , Infecções por Picornaviridae/prevenção & controle , Resultado do Tratamento , Adulto Jovem
15.
Front Immunol ; 12: 753683, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899705

RESUMO

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.


Assuntos
Efeito Citopatogênico Viral , Modelos Animais de Doenças , Encefalite Viral/metabolismo , Parechovirus/patogenicidade , Infecções por Picornaviridae/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Autofagia , Linhagem Celular Tumoral , Córtex Cerebral/virologia , Chlorocebus aethiops , Citocinas/biossíntese , Citocinas/genética , Encefalite Viral/patologia , Encefalite Viral/virologia , Glioblastoma/patologia , Hipocampo/virologia , Humanos , Inflamação , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Interferon Tipo I/farmacologia , Interferons/biossíntese , Interferons/genética , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/patologia , Parechovirus/efeitos dos fármacos , Parechovirus/fisiologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Piroptose , Células Vero , Replicação Viral/efeitos dos fármacos , Interferon lambda
16.
Sci Rep ; 11(1): 23741, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34887467

RESUMO

The mechanisms explaining excess morbidity and mortality in respiratory infections among males are poorly understood. Innate immune responses are critical in protection against respiratory virus infections. We hypothesised that innate immune responses to respiratory viruses may be deficient in males. We stimulated peripheral blood mononuclear cells from 345 participants at age 16 years in a population-based birth cohort with three live respiratory viruses (rhinoviruses A16 and A1, and respiratory syncytial virus) and two viral mimics (R848 and CpG-A, to mimic responses to SARS-CoV-2) and investigated sex differences in interferon (IFN) responses. IFN-α responses to all viruses and stimuli were 1.34-2.06-fold lower in males than females (P = 0.018 - < 0.001). IFN-ß, IFN-γ and IFN-induced chemokines were also deficient in males across all stimuli/viruses. Healthcare records revealed 12.1% of males and 6.6% of females were hospitalized with respiratory infections in infancy (P = 0.017). In conclusion, impaired innate anti-viral immunity in males likely results in high male morbidity and mortality from respiratory virus infections.


Assuntos
Imidazóis/imunologia , Imunidade Inata , Oligodesoxirribonucleotídeos/imunologia , Infecções por Picornaviridae/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sincicial Respiratório Humano/imunologia , Rhinovirus/imunologia , Adolescente , Coorte de Nascimento , Estudos de Coortes , Feminino , Humanos , Interferons/imunologia , Interferons/metabolismo , Leucócitos Mononucleares/imunologia , Masculino , Infecções por Picornaviridae/mortalidade , Infecções por Picornaviridae/virologia , Infecções por Vírus Respiratório Sincicial/mortalidade , Infecções por Vírus Respiratório Sincicial/virologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/mortalidade , Infecções Respiratórias/virologia , SARS-CoV-2 , Fatores Sexuais
17.
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
18.
Viruses ; 13(12)2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34960681

RESUMO

The use of high-throughput sequencing has facilitated virus discovery in wild animals and helped determine their potential threat to humans and other animals. We report the complete genome sequence of a novel picornavirus identified by next-generation sequencing in faeces from Australian fallow deer. Genomic analysis revealed that this virus possesses a typical picornavirus-like genomic organisation of 7554 nt with a single open reading frame (ORF) encoding a polyprotein of 2225 amino acids. Based on the amino acid identity comparison and phylogenetic analysis of the P1, 2C, 3CD, and VP1 regions, this novel picornavirus was closely related to but distinct from known bopiviruses detected to date. This finding suggests that deer/bopivirus could belong to a novel species within the genus Bopivirus, tentatively designated as "Bopivirus C". Epidemiological investigation of 91 deer (71 fallow, 14 sambar and 6 red deer) and 23 cattle faecal samples showed that six fallow deer and one red deer (overall prevalence 7.7%, 95% confidence interval [CI] 3.8-15.0%) tested positive, but deer/bopivirus was undetectable in sambar deer and cattle. In addition, phylogenetic and sequence analyses indicate that the same genotype is circulating in south-eastern Australia. To our knowledge, this study reports for the first time a deer-origin bopivirus and the presence of a member of genus Bopivirus in Australia. Further epidemiological and molecular studies are needed to investigate the geographic distribution and pathogenic potential of this novel Bopivirus species in other domestic and wild animal species.


Assuntos
Animais Selvagens/virologia , Cervos/virologia , Infecções por Picornaviridae/veterinária , Picornaviridae/classificação , Picornaviridae/genética , Animais , Austrália/epidemiologia , Bovinos , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/virologia , Fezes/virologia , Genoma Viral , Sequenciamento de Nucleotídeos em Larga Escala , Filogenia , Picornaviridae/isolamento & purificação , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/virologia , Prevalência , RNA Viral/genética
19.
Viruses ; 13(12)2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34960797

RESUMO

BACKGROUND: We aimed to compare the clinical severity in patients who were coinfected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and rhinovirus or monoinfected with a single one of these viruses. METHODS: The study period ranged from 1 March 2020 to 28 February 2021 (one year). SARS-CoV-2 and other respiratory viruses were identified by real-time reverse-transcription-PCR as part of the routine work at Marseille University hospitals. Bacterial and fungal infections were detected by standard methods. Clinical data were retrospectively collected from medical files. This study was approved by the ethical committee of our institute. RESULTS: A total of 6034/15,157 (40%) tested patients were positive for at least one respiratory virus. Ninety-three (4.3%) SARS-CoV-2-infected patients were coinfected with another respiratory virus, with rhinovirus being the most frequent (62/93, 67%). Patients coinfected with SARS-CoV-2 and rhinovirus were significantly more likely to report a cough than those with SARS-CoV-2 monoinfection (62% vs. 31%; p = 0.0008). In addition, they were also significantly more likely to report dyspnea than patients with rhinovirus monoinfection (45% vs. 36%; p = 0.02). They were also more likely to be transferred to an intensive care unit and to die than patients with rhinovirus monoinfection (16% vs. 5% and 7% vs. 2%, respectively) but these differences were not statistically significant. CONCLUSIONS: A close surveillance and investigation of the co-incidence and interactions of SARS-CoV-2 and other respiratory viruses is needed. The possible higher risk of increased clinical severity in SARS-CoV-2-positive patients coinfected with rhinovirus warrants further large scale studies.


Assuntos
COVID-19/epidemiologia , Coinfecção/epidemiologia , Coinfecção/virologia , Infecções por Picornaviridae/epidemiologia , Adolescente , Adulto , Idoso , COVID-19/diagnóstico , Criança , Coinfecção/diagnóstico , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Infecções por Picornaviridae/diagnóstico , Infecções por Picornaviridae/virologia , Reação em Cadeia da Polimerase em Tempo Real , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Estudos Retrospectivos , Rhinovirus , SARS-CoV-2 , Índice de Gravidade de Doença , Adulto Jovem
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...