Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
PLoS One ; 18(11): e0294173, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37963152

RESUMEN

Paramyxoviruses are negative-sense, single-stranded RNA viruses that are associated with numerous diseases in humans and animals. J paramyxovirus (JPV) was first isolated from moribund mice (Mus musculus) with hemorrhagic lung lesions in Australia in 1972. In 2016, JPV was classified into the newly established genus Jeilongvirus. Novel jeilongviruses are being discovered worldwide in wildlife populations. However, the effects of jeilongvirus infection on host gene expression remains uncharacterized. To address this, cellular RNA from JPV-infected mouse fibroblasts was collected at 2, 4, 8, 12, 16, 24, and 48 hours post-infection (hpi) and were sequenced using single-end 75 base pairs (SE75) sequencing chemistry on an Illumina NextSeq platform. Differentially expressed genes (DEGs) between the virus-infected replicates and mock replicates at each timepoint were identified using the Tophat2-Cufflinks-Cuffdiff protocol. At 2 hpi, 11 DEGs were identified in JPV-infected cells, while 1,837 DEGs were detected at 48 hpi. A GO analysis determined that the genes at the earlier timepoints were involved in interferon responses, while there was a shift towards genes that are involved in antigen processing and presentation processes at the later timepoints. At 48 hpi, a KEGG analysis revealed that many of the DEGs detected were involved in pathways that are important for immune responses. qRT-PCR verified that Rtp4, Ifit3, Mx2, and Stat2 were all upregulated during JPV infection, while G0s2 was downregulated. After JPV infection, the expression of inflammatory and antiviral factors in mouse fibroblasts changes significantly. This study provides crucial insight into the different arms of host immunity that mediate Jeilongvirus infection. Understanding the pathogenic mechanisms of Jeilongvirus will lead to better strategies for the prevention and control of potential diseases that may arise from this group of viruses.


Asunto(s)
Infecciones por Paramyxoviridae , Paramyxovirinae , Humanos , Animales , Ratones , Paramyxovirinae/genética , Paramyxoviridae/genética , Infecciones por Paramyxoviridae/genética , Expresión Génica , Australia , Perfilación de la Expresión Génica , Regulación de la Expresión Génica
2.
Mol Biol Rep ; 50(2): 1109-1116, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36399244

RESUMEN

BACKGROUND: Human metapneumovirus (HMPV) belongs to the family Pneumoviridae. It is one of the emerging respiratory viruses causing both upper and lower respiratory tract illnesses. HMPV has two genotypes: A and B. These genotypes are classified into lineage A1, A2, B1 and B2. Lineage-A2 is further classified as A2a, A2b and A2c. Similarly, B2 is classified as B2a and B2b. Studies have shown the circulation of A2b, B1 and B2 lineages in India. However, a limited amount of data is available on the current circulating genotypes of HMPV in India. METHODS: Throat swab samples positive for HMPV by real-time RT- PCR, archived at Manipal Institute of Virology as a part of a hospital-based acute febrile illness surveillance study, was used from April 2016 to August 2018 by purposive sampling method. We performed the conventional reverse transcriptase-polymerase chain reaction for twenty samples targeting the G gene and then subjected them to sequencing. Phylogenetic analysis was done using MEGA X software by the Maximum Likelihood method. RESULTS: All the twenty sequences belonged to the A2c subgroup. Phylogenetic analysis showed that strains from the study have genetic relation with circulating strains in Japan, China and Croatia. Seven out of the twenty sequences showed 180-nucleotide duplication and eleven sequences showed 111-nucleotide duplication. Two sequences did not show any duplications. CONCLUSION: In the current study, we report that A2c is the sub-lineage in India from April 2016 to August 2018. This study is the first retrospective study reporting the circulation of the A2c sub-lineage among adults in India with 180- and 111-nucleotide duplications in the G gene of human metapneumovirus.


Asunto(s)
Metapneumovirus , Infecciones por Paramyxoviridae , Infecciones del Sistema Respiratorio , Adulto , Humanos , Lactante , Metapneumovirus/genética , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/genética , Estudios Retrospectivos , Filogenia , Duplicación de Gen , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/epidemiología , Genotipo , India , Nucleótidos
3.
Sci Rep ; 12(1): 2790, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35181674

RESUMEN

Lower respiratory illness is one of the leading causes of death among children in low- and high-income countries. Human metapneumovirus (hMPV) is a key contributor to respiratory illnesses commonly reported among children and causes serious clinical complications ranging from mild respiratory infections to severe lower respiratory tract anomalies mainly in the form of bronchiolitis and pneumonia. However, due to the lack of a national surveillance system, the clinical significance of hMPV remains obscure in the Pakistani population. This study was conducted to screen throat swabs samples collected from 127 children reported with respiratory symptoms at a tertiary care hospital in Islamabad. Out of 127, 21 (16.5%) samples were positive for hMPV with its genotype distribution as A2a (10%), A2b (20%), B1 (10%), and B2 (60%). Phylogenetic analysis showed that the hMPV viruses were closely related to those reported from neighboring countries including India and China. This work will contribute to a better understanding of this virus, its diagnosis, and the handling of patients in clinical setups. Further studies at a large-scale are warranted for a better understanding of the disease burden and epidemiology of hMPV in Pakistan.


Asunto(s)
Metapneumovirus/aislamiento & purificación , Infecciones por Paramyxoviridae/diagnóstico , Infecciones del Sistema Respiratorio/diagnóstico , Preescolar , Femenino , Genotipo , Humanos , Lactante , Masculino , Metapneumovirus/genética , Metapneumovirus/patogenicidad , Epidemiología Molecular , Pakistán/epidemiología , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Filogenia , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/virología
4.
Mol Biol Rep ; 48(12): 7697-7702, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34665397

RESUMEN

BACKGROUND: Human metapneumovirus (hMPV) is one of the important pathogens in infant respiratory tract infection. However, the molecular epidemiology of hMPV among children < 14 years of age hospitalized with severe acute respiratory infection (SARI) is unclear. We investigated the hMPV infection status and genotypes of children hospitalized with SARI from January 2016 to December 2020 in Huzhou, China. METHODS: A nasopharyngeal flocked swab, nasal wash, or nasopharyngeal swab/or opharyngeal swab combination sample was collected from children with SARI in Huzhou from January 2016 to December 2020. Quantitative reverse transcription-polymerase chain reaction was performed to detect hMPV RNA. The hMPV F gene was amplified and sequenced, followed by analysis using MEGA software (ver. 7.0). Epidemiological data were analyzed using Microsoft Excel 2010 and SPSS (ver. 22.0) software. RESULTS: A total of 1133 children with SARI were recruited from 2016 to 2020. Among them, 56 (4.94%) were positive for hMPV-RNA. Children < 5 years of age accounted for 85.71% of the positive cases. The hMPV incidence was high in spring and winter, especially in December and January to March. Phylogenetic analysis of the F-gene sequences of 28 hMPV strains showed that the A1, B1, and B2 genotypes were prevalent in Huzhou, and the dominant hMPV genotype varied according to surveillance year. CONCLUSIONS: HMPV is an important respiratory pathogen in children in Huzhou, with a high incidence in winter and spring in children < 5 years of age. In this study, genotypes A1, B1, and B2 were the most prevalent.


Asunto(s)
Metapneumovirus/genética , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/genética , Secuencia de Bases/genética , Niño , Preescolar , China/epidemiología , Femenino , Genotipo , Hospitalización/tendencias , Humanos , Lactante , Masculino , Metapneumovirus/clasificación , Metapneumovirus/patogenicidad , Epidemiología Molecular/métodos , Tipificación Molecular/métodos , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Filogenia , Infecciones del Sistema Respiratorio/metabolismo , Análisis de Secuencia de ADN/métodos
5.
PLoS One ; 15(12): e0244093, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33370354

RESUMEN

Human Metapneumovirus (hMPV) is responsible for acute respiratory infections in humans, with clinical and epidemiological relevance in pediatric, elderly, and immunocompromised populations. These features are largely unknown in Córdoba, Argentina and in adults in general. Hence, our goal was to broadly characterize hMPV infection in patients of all ages hospitalized with acute respiratory infections in Córdoba, Argentina, including epidemiology, clinical features and genetic diversity. Nasopharyngeal secretions were obtained from 795 patients during 2011-2013, 621 patients were 0-25 years old and 174 were 26-85 years old. HMPV was assayed by RT-PCR and other respiratory viruses by indirect immunofluorescence. Local strains were identified by sequence analysis. Human Metapneumovirus was detected in 20.3% (161/795) patients, 13.1% as single infections and 7.2% in co-infections, more frequently with Respiratory Syncytial Virus. HMPV circulated during late winter and spring in all age patients, but mainly in children under 4 years old in 71.4% (115/161) and adults between 26 and 59 years old in 12.4% (20/161). The most prevalent diagnosis was mild acute respiratory infection in 59.6% (96/161) and bronchiolitis in 9.3% (15/161). Local strains were clustered within A2 subtype; they presented 73-100% identities among them, showing a high degree of homology compared to isolations from neighboring countries. We demonstrate that hMPV circulated among all age patients with respiratory infection during 2011-2013 in Córdoba, contributing to the understanding of this virus, its diagnosis and patient handling in local health-care centers.


Asunto(s)
Genotipo , Metapneumovirus/genética , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/genética , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Argentina/epidemiología , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Infecciones por Paramyxoviridae/virología , Infecciones del Sistema Respiratorio/virología
6.
Viruses ; 12(12)2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33276587

RESUMEN

The paramyxo- and pneumovirus family includes a wide range of viruses that can cause respiratory and/or systemic infections in humans and animals. The significant disease burden of these viruses is further exacerbated by the limited therapeutics that are currently available. Host cellular proteins that can antagonize or limit virus replication are therefore a promising area of research to identify candidate molecules with the potential for host-targeted therapies. Host proteins known as host cell restriction factors are constitutively expressed and/or induced in response to virus infection and include proteins from interferon-stimulated genes (ISGs). Many ISG proteins have been identified but relatively few have been characterized in detail and most studies have focused on studying their antiviral activities against particular viruses, such as influenza A viruses and human immunodeficiency virus (HIV)-1. This review summarizes current literature regarding host cell restriction factors against paramyxo- and pneumoviruses, on which there is more limited data. Alongside discussion of known restriction factors, this review also considers viral countermeasures in overcoming host restriction, the strengths and limitations in different experimental approaches in studies reported to date, and the challenges in reconciling differences between in vitro and in vivo data. Furthermore, this review provides an outlook regarding the landscape of emerging technologies and tools available to study host cell restriction factors, as well as the suitability of these proteins as targets for broad-spectrum antiviral therapeutics.


Asunto(s)
Interacciones Huésped-Patógeno , Infecciones por Paramyxoviridae/virología , Paramyxovirinae/fisiología , Infecciones por Pneumovirus/virología , Pneumovirus/fisiología , Animales , Biomarcadores , Regulación Viral de la Expresión Génica , Especificidad del Huésped , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/metabolismo , Infecciones por Pneumovirus/genética , Infecciones por Pneumovirus/metabolismo , Tropismo Viral , Replicación Viral
7.
Viruses ; 12(7)2020 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-32635475

RESUMEN

The host tropism of viral infection is determined by a variety of factors, from cell surface receptors to innate immune signaling. Many viruses encode proteins that interfere with host innate immune recognition in order to promote infection. STAT2 is divergent between species and therefore has a role in species restriction of some viruses. To understand the role of STAT2 in human metapneumovirus (HMPV) infection of human and murine tissues, we first infected STAT2-/- mice and found that HMPV could be serially passaged in STAT2-/-, but not WT, mice. We then used in vitro methods to show that HMPV inhibits expression of both STAT1 and STAT2 in human and primate cells, but not in mouse cells. Transfection of the murine form of STAT2 into STAT2-deficient human cells conferred resistance to STAT2 inhibition. Finally, we sought to understand the in vivo role of STAT2 by infecting hSTAT2 knock-in mice with HMPV, and found that mice had increased weight loss, inhibition of type I interferon signaling, and a Th2-polarized cytokine profile compared to WT mice. These results indicate that STAT2 is a target of HMPV in human infection, while the murine version of STAT2 restricts tropism of HMPV for murine cells and tissue.


Asunto(s)
Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/inmunología , Factor de Transcripción STAT2/inmunología , Animales , Femenino , Especificidad del Huésped , Humanos , Inmunidad Innata , Interferones/genética , Interferones/inmunología , Masculino , Metapneumovirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Factor de Transcripción STAT2/genética , Células Th2
8.
Viruses ; 12(6)2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32545470

RESUMEN

The human respiratory syncytial virus (hRSV) and human Metapneumovirus (hMPV) are two of the leading etiological agents of acute lower respiratory tract infections, which constitute the main cause of mortality in infants. However, there are currently approved vaccines for neither hRSV nor hMPV. Moreover, despite the similarity between the pathology caused by both viruses, the immune response elicited by the host is different in each case. In this review, we discuss how dendritic cells, alveolar macrophages, neutrophils, eosinophils, natural killer cells, innate lymphoid cells, and the complement system regulate both pathogenesis and the resolution of hRSV and hMPV infections. The roles that these cells play during infections by either of these viruses will help us to better understand the illnesses they cause. We also discuss several controversial findings, relative to some of these innate immune components. To better understand the inflammation in the lungs, the role of the respiratory epithelium in the recruitment of innate immune cells is briefly discussed. Finally, we review the main prophylactic strategies and current vaccine candidates against both hRSV and hMPV.


Asunto(s)
Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Metapneumovirus/genética , Neutrófilos/inmunología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética
9.
Viruses ; 12(3)2020 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-32245206

RESUMEN

Recent high-throughput sequencing revealed that only 2% of the transcribed human genome codes for proteins, while the majority of transcriptional products are non-coding RNAs (ncRNAs). Herein, we review the current knowledge regarding ncRNAs, both host- and virus-derived, and their role in respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) infections. RSV is known as the most common cause of lower respiratory tract infection (LRTI) in children, while hMPV is also a significant contributor to LRTI in the pediatrics population. Although RSV and hMPV are close members, belonging to the Pneumoviridae family, they induce distinct changes in the ncRNA profile. Several types of host ncRNAs, including long ncRNA (lncRNA), microRNAs (miRNAs), and transfer RNA (tRNA)-derived RNA fragments (tRFs), are involved as playing roles in RSV and/or hMPV infection. Given the importance of ncRNAs in regulating the expression and functions of genes and proteins, comprehensively understanding the roles of ncRNAs in RSV/hMPV infection could shed light upon the disease mechanisms of RSV and hMPV, potentially providing insights into the development of prevention strategies and antiviral therapy. The presence of viral-derived RNAs and the potential of using ncRNAs as diagnostic biomarkers are also discussed in this review.


Asunto(s)
Regulación de la Expresión Génica , Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , ARN no Traducido/genética , Infecciones por Virus Sincitial Respiratorio/genética , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/fisiología , Animales , Biomarcadores , Resistencia a la Enfermedad/genética , Descubrimiento de Drogas , Interacciones Huésped-Patógeno/genética , Humanos , MicroARNs
10.
Sci Rep ; 10(1): 2852, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32071381

RESUMEN

Human metapneumovirus (hMPV) is an important pathogen that causes upper and lower respiratory tract infections in children worldwide. hMPV has two major genotypes, hMPV-A and hMPV-B. Epidemiological studies have shown that the two hMPV genotypes alternate in predominance worldwide in recent years. Co-circulation of the two genotypes of hMPV was usually observed and there is no study about the interaction between them, such as competitive replication, which maybe the possible mechanisms for alternating prevalence of subtypes. Our present study have used two different genotypes of hMPV (genotype A: NL/1/00; B: NL/1/99) in different proportions in animal model (BALB/c mice) and cell model (Vero-E6) separately. The result showed that the competitive growth does exist in BALB/c mice, genotype B had a strong competitive advantage. However, genotype B did not cause more severe disease than non-predominant (genotype A) or mixed strains in the study, which were evaluated by the body weight, airway hyperresponsiveness and lung pathology of mouse. In cell model, competitive growth and the two genotypes alternately prevalence were observed. In summary, we confirmed that there was a competitive replication between hMPV genotype A and B, and no difference in disease severity caused by the two subtypes. This study shows a new insight to understand the alternation of hMPV genotype prevalence through genotype competition and provide experimental evidence for disease control and vaccine design.


Asunto(s)
Metapneumovirus/genética , Epidemiología Molecular , Infecciones por Paramyxoviridae/genética , Filogenia , Animales , Genotipo , Humanos , Metapneumovirus/patogenicidad , Ratones , Infecciones por Paramyxoviridae/patología , Infecciones por Paramyxoviridae/virología , ARN Viral/genética , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Análisis de Secuencia de ADN
11.
Nat Microbiol ; 5(4): 584-598, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32015498

RESUMEN

Internal N6-methyladenosine (m6A) modification is one of the most common and abundant modifications of RNA. However, the biological roles of viral RNA m6A remain elusive. Here, using human metapneumovirus (HMPV) as a model, we demonstrate that m6A serves as a molecular marker for innate immune discrimination of self from non-self RNAs. We show that HMPV RNAs are m6A methylated and that viral m6A methylation promotes HMPV replication and gene expression. Inactivating m6A addition sites with synonymous mutations or demethylase resulted in m6A-deficient recombinant HMPVs and virion RNAs that induced increased expression of type I interferon, which was dependent on the cytoplasmic RNA sensor RIG-I, and not on melanoma differentiation-associated protein 5 (MDA5). Mechanistically, m6A-deficient virion RNA induces higher expression of RIG-I, binds more efficiently to RIG-I and facilitates the conformational change of RIG-I, leading to enhanced interferon expression. Furthermore, m6A-deficient recombinant HMPVs triggered increased interferon in vivo and were attenuated in cotton rats but retained high immunogenicity. Collectively, our results highlight that (1) viruses acquire m6A in their RNA as a means of mimicking cellular RNA to avoid detection by innate immunity and (2) viral RNA m6A can serve as a target to attenuate HMPV for vaccine purposes.


Asunto(s)
Adenosina/análogos & derivados , Proteína 58 DEAD Box/genética , Evasión Inmune/genética , Interferón beta/genética , Metapneumovirus/inmunología , ARN Viral/genética , Células A549 , Adenosina/inmunología , Adenosina/metabolismo , Animales , Chlorocebus aethiops , Proteína 58 DEAD Box/inmunología , Regulación de la Expresión Génica , Genoma Viral/inmunología , Células HeLa , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/inmunología , Interferón beta/inmunología , Metapneumovirus/genética , Metapneumovirus/crecimiento & desarrollo , FN-kappa B/genética , FN-kappa B/inmunología , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/virología , ARN Viral/inmunología , Receptores Inmunológicos , Sigmodontinae , Transducción de Señal , Células THP-1 , Células Vero , Virión/genética , Virión/crecimiento & desarrollo , Virión/inmunología
12.
BMC Genomics ; 21(1): 5, 2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-31898474

RESUMEN

BACKGROUND: Human metapneumovirus (HMPV) is an important cause of acute respiratory illness in young children. Whole genome sequencing enables better identification of transmission events and outbreaks, which is not always possible with sub-genomic sequences. RESULTS: We report a 2-reaction amplicon-based next generation sequencing method to determine the complete genome sequences of five HMPV strains, representing three subgroups (A2, B1 and B2), directly from clinical samples. In addition to reporting five novel HMPV genomes from Africa we examined genetic diversity and sequence patterns of publicly available HMPV genomes. We found that the overall nucleotide sequence identity was 71.3 and 80% for HMPV group A and B, respectively, the diversity between HMPV groups was greater at amino acid level for SH and G surface protein genes, and multiple subgroups co-circulated in various countries. Comparison of sequences between HMPV groups revealed variability in G protein length (219 to 241 amino acids) due to changes in the stop codon position. Genome-wide phylogenetic analysis showed congruence with the individual gene sequence sets except for F and M2 genes. CONCLUSION: This is the first genomic characterization of HMPV genomes from African patients.


Asunto(s)
Genoma Viral/genética , Metapneumovirus/genética , Infecciones por Paramyxoviridae/genética , Secuenciación Completa del Genoma , Secuencia de Aminoácidos , Genotipo , Humanos , Kenia/epidemiología , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/epidemiología , Infecciones por Paramyxoviridae/virología , Filogenia , Proteínas Virales/genética , Zambia/epidemiología
13.
Virology ; 531: 183-191, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30927711

RESUMEN

Human metapneumovirus (hMPV) is an important cause of acute lower respiratory tract infections in infants, elderly and immunocompromised individuals. Ingenuity pathway analysis of microarrays data showed that 20% of genes affected by hMPV infection of airway epithelial cells (AECs) were related to metabolism. We found that levels of the glycolytic pathway enzymes hexokinase 2, pyruvate kinase M2, and lactate dehydrogenase A were significantly upregulated in normal human AECs upon hMPV infection, as well as levels of enzymes belonging to the hexosamine biosynthetic and glycosylation pathways. On the other hand, expression of the majority of the enzymes belonging to the tricarboxylic acid cycle was significantly diminished. Inhibition of hexokinase 2 and of the glycosylating enzyme O-linked N-acetylglucosamine transferase led to a significant reduction in hMPV titer, indicating that metabolic changes induced by hMPV infection play a major role during the virus life cycle, and could be explored as potential antiviral targets.


Asunto(s)
Células Epiteliales/metabolismo , Metapneumovirus/fisiología , Infecciones por Paramyxoviridae/metabolismo , Mucosa Respiratoria/metabolismo , Línea Celular , Células Epiteliales/virología , Glucólisis , Hexosaminas/biosíntesis , Humanos , Redes y Vías Metabólicas , Metapneumovirus/genética , Fosforilación Oxidativa , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/fisiopatología , Infecciones por Paramyxoviridae/virología , Mucosa Respiratoria/virología , Replicación Viral
14.
PLoS Pathog ; 15(2): e1007561, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30742688

RESUMEN

Paramyxoviruses can establish persistent infections both in vitro and in vivo, some of which lead to chronic disease. However, little is known about the molecular events that contribute to the establishment of persistent infections by RNA viruses. Using parainfluenza virus type 5 (PIV5) as a model we show that phosphorylation of the P protein, which is a key component of the viral RNA polymerase complex, determines whether or not viral transcription and replication becomes repressed at late times after infection. If the virus becomes repressed, persistence is established, but if not, the infected cells die. We found that single amino acid changes at various positions within the P protein switched the infection phenotype from lytic to persistent. Lytic variants replicated to higher titres in mice than persistent variants and caused greater infiltration of immune cells into infected lungs but were cleared more rapidly. We propose that during the acute phases of viral infection in vivo, lytic variants of PIV5 will be selected but, as the adaptive immune response develops, variants in which viral replication can be repressed will be selected, leading to the establishment of prolonged, persistent infections. We suggest that similar selection processes may operate for other RNA viruses.


Asunto(s)
Infecciones por Paramyxoviridae/genética , Paramyxoviridae/genética , Fosfoproteínas/genética , Proteínas Virales/genética , Células A549 , Sustitución de Aminoácidos/genética , Animales , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Virus de la Parainfluenza 5/genética , Virus de la Parainfluenza 5/patogenicidad , Paramyxoviridae/patogenicidad , Infecciones por Paramyxoviridae/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Fosforilación , ARN Viral , Proteínas Virales/metabolismo , Proteínas Virales/fisiología , Replicación Viral
15.
Virus Genes ; 55(2): 191-197, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30632017

RESUMEN

Using viral metagenomics, we characterized the mammalian virome of nasal swabs from 57 dogs with unexplained signs of respiratory infection showing mostly negative results using the IDEXX Canine Respiratory Disease RealPCR™ Panel. We identified canine parainfluenza virus 5, canine respiratory coronavirus, carnivore bocaparvovirus 3, canine circovirus and canine papillomavirus 9. Novel canine taupapillomaviruses (CPV21-23) were also identified in 3 dogs and their complete genome sequenced showing L1 nucleotide identity ranging from 68.4 to 70.3% to their closest taupapillomavirus relative. Taupapillomavirus were the only mammalian viral nucleic acids detected in two affected dogs, while a third dog was coinfected with low levels of canine parainfluenza 5. A role for these taupapillomavirues in canine respiratory disease remains to be determined.


Asunto(s)
Coronavirus Canino/genética , Metagenómica , Infecciones por Paramyxoviridae/virología , Infecciones del Sistema Respiratorio/virología , Animales , Coinfección/genética , Coinfección/veterinaria , Coinfección/virología , Coronavirus Canino/aislamiento & purificación , Coronavirus Canino/patogenicidad , Enfermedades de los Perros/genética , Enfermedades de los Perros/virología , Perros , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/veterinaria , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/veterinaria
16.
FEBS Lett ; 592(14): 2444-2457, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29931672

RESUMEN

Parainfluenza virus infection is a common respiratory illness in children. Although lncRNAs are novel regulators of virus-induced innate immunity, a systemic attempt to characterize the differential expression of lncRNAs upon parainfluenza virus infection is lacking. In this report, we identify 207 lncRNAs and 166 mRNAs differentially expressed in SeV-infected HEK293T cells by microarray. The functional annotation analysis reveals that differentially regulated transcripts are predominantly involved in the host antiviral response pathway. The lncRNAs with the potential to regulate SeV-induced antiviral response are identified by building the lncRNA-mRNA coexpression network. Furthermore, silencing lncRNA ENST00000565297 results in reduced type I IFN signaling upon SeV infection. These catalogs may facilitate future analysis of the functions of lncRNAs in innate immunity and related diseases.


Asunto(s)
Inmunidad Innata/genética , Infecciones por Paramyxoviridae/genética , ARN Largo no Codificante/fisiología , Niño , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Paramyxoviridae/inmunología , ARN Largo no Codificante/genética , Infecciones por Respirovirus/genética , Infecciones por Respirovirus/inmunología , Virus Sendai/inmunología , Virus Sendai/patogenicidad , Transcriptoma
17.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29321315

RESUMEN

Paramyxovirus V proteins are known antagonists of the RIG-I-like receptor (RLR)-mediated interferon induction pathway, interacting with and inhibiting the RLR MDA5. We report interactions between the Nipah virus V protein and both RIG-I regulatory protein TRIM25 and RIG-I. We also observed interactions between these host proteins and the V proteins of measles virus, Sendai virus, and parainfluenza virus. These interactions are mediated by the conserved C-terminal domain of the V protein, which binds to the tandem caspase activation and recruitment domains (CARDs) of RIG-I (the region of TRIM25 ubiquitination) and to the SPRY domain of TRIM25, which mediates TRIM25 interaction with the RIG-I CARDs. Furthermore, we show that V interaction with TRIM25 and RIG-I prevents TRIM25-mediated ubiquitination of RIG-I and disrupts downstream RIG-I signaling to the mitochondrial antiviral signaling protein. This is a novel mechanism for innate immune inhibition by paramyxovirus V proteins, distinct from other known V protein functions such as MDA5 and STAT1 antagonism.IMPORTANCE The host RIG-I signaling pathway is a key early obstacle to paramyxovirus infection, as it results in rapid induction of an antiviral response. This study shows that paramyxovirus V proteins interact with and inhibit the activation of RIG-I, thereby interrupting the antiviral signaling pathway and facilitating virus replication.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Infecciones por Paramyxoviridae/metabolismo , Paramyxoviridae/fisiología , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Células A549 , Animales , Proteína 58 DEAD Box/genética , Perros , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Infecciones por Paramyxoviridae/genética , Receptores Inmunológicos , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética , Proteínas Virales/genética
18.
Sci Rep ; 8(1): 883, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-29343779

RESUMEN

Thymic stromal lymphopoietin (TSLP) is associated with several allergic diseases including asthma. Two isoforms of TSLP exist in humans, a long form (lfTSLP) and a short form (sfTSLP), displaying distinct immunological functions. Recently, TSLP was found to be upregulated in human airway cells upon human metapneumovirus (hMPV) infection, yet it remains unclear if the two isoforms are regulated differently during hMPV infection. Importantly, the molecular mechanisms underlying hMPV-mediated TSLP induction remain undescribed. In this study, we characterized the expression and regulation of TSLP in hMPV-infected human airway cells. We demonstrated that hMPV strongly induced the expression of pro-inflammatory lfTSLP in human airway epithelial cells and lung fibroblasts. Further, knockdown of pattern recognition receptors retinoic acid-inducible gene I (RIG-I) or Toll-like receptor 3 (TLR3), as well as downstream signal transducers, abrogated hMPV-mediated lfTSLP induction. Importantly, silencing of TANK-binding kinase 1 (TBK1) also impaired hMPV-mediated lfTSLP induction, which could be attributed to compromised NF-κB activation. Overall, these results suggest that TBK1 may be instrumental for hMPV-mediated activation of NF-κB downstream RIG-I and TLR3, leading to a specific induction of lfTSLP in hMPV-infected human airway cells.


Asunto(s)
Citocinas/genética , Regulación de la Expresión Génica/genética , Inflamación/genética , Metapneumovirus/patogenicidad , Infecciones por Paramyxoviridae/genética , Transducción de Señal/genética , Células A549 , Asma/genética , Asma/virología , Línea Celular Tumoral , Proteína 58 DEAD Box/genética , Células Epiteliales/virología , Humanos , Hipersensibilidad/genética , Hipersensibilidad/virología , Inflamación/virología , FN-kappa B/genética , Infecciones por Paramyxoviridae/virología , Proteínas Serina-Treonina Quinasas/genética , Receptor Toll-Like 3/genética , Linfopoyetina del Estroma Tímico
19.
Autophagy ; 13(10): 1709-1721, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28949785

RESUMEN

An increasing number of studies have demonstrated that macroautophagy/autophagy plays an important role in the infectious processes of diverse pathogens. However, it remains unknown whether autophagy is induced in avian metapneumovirus (aMPV)-infected host cells, and, if so, how this occurs. Here, we report that aMPV subgroup C (aMPV/C) induces autophagy in cultured cells. We demonstrated this relationship by detecting classical autophagic features, including the formation of autophagsomes, the presence of GFP-LC3 puncta and the conversation of LC3-I into LC3-II. Also, we used pharmacological regulators and siRNAs targeting ATG7 or LC3 to examine the role of autophagy in aMPV/C replication. The results showed that autophagy is required for efficient replication of aMPV/C. Moreover, infection with aMPV/C promotes autophagosome maturation and induces a complete autophagic process. Finally, the ATF6 pathway, of which one component is the unfolded protein response (UPR), becomes activated in aMPV/C-infected cells. Knockdown of ATF6 inhibited aMPV/C-induced autophagy and viral replication. Collectively, these results not only show that autophagy promotes aMPV/C replication in the cultured cells, but also reveal that the molecular mechanisms underlying aMPV/C-induced autophagy depends on regulation of the ER stress-related UPR pathway.


Asunto(s)
Factor de Transcripción Activador 6/fisiología , Autofagia , Metapneumovirus/fisiología , Respuesta de Proteína Desplegada , Factor de Transcripción Activador 6/genética , Animales , Autofagia/genética , Enfermedades de las Aves/genética , Enfermedades de las Aves/virología , Células Cultivadas , Embrión de Pollo , Chlorocebus aethiops , Técnicas de Silenciamiento del Gen , Infecciones por Paramyxoviridae/genética , Infecciones por Paramyxoviridae/virología , Transducción de Señal/genética , Respuesta de Proteína Desplegada/genética , Células Vero , Replicación Viral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...