Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.221
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 160(4): 619-630, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25679758

RESUMEN

A central paradigm within virology is that each viral particle largely behaves as an independent infectious unit. Here, we demonstrate that clusters of enteroviral particles are packaged within phosphatidylserine (PS) lipid-enriched vesicles that are non-lytically released from cells and provide greater infection efficiency than free single viral particles. We show that vesicular PS lipids are co-factors to the relevant enterovirus receptors in mediating subsequent infectivity and transmission, in particular to primary human macrophages. We demonstrate that clustered packaging of viral particles within vesicles enables multiple viral RNA genomes to be collectively transferred into single cells. This study reveals a novel mode of viral transmission, where enteroviral genomes are transmitted from cell-to-cell en bloc in membrane-bound PS vesicles instead of as single independent genomes. This has implications for facilitating genetic cooperativity among viral quasispecies as well as enhancing viral replication.


Asunto(s)
Vesículas Citoplasmáticas/virología , Infecciones por Enterovirus/transmisión , Enterovirus/fisiología , Macrófagos/virología , Vesículas Citoplasmáticas/química , Humanos , Macrófagos/citología , Fosfatidilserinas , Poliovirus/fisiología , ARN Viral/metabolismo , Rhinovirus/fisiología , Replicación Viral
2.
Immunity ; 53(6): 1258-1271.e5, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33296686

RESUMEN

CD4+ T cells reactive against SARS-CoV-2 can be found in unexposed individuals, and these are suggested to arise in response to common cold coronavirus (CCCoV) infection. Here, we utilized SARS-CoV-2-reactive CD4+ T cell enrichment to examine the antigen avidity and clonality of these cells, as well as the relative contribution of CCCoV cross-reactivity. SARS-CoV-2-reactive CD4+ memory T cells were present in virtually all unexposed individuals examined, displaying low functional avidity and multiple, highly variable cross-reactivities that were not restricted to CCCoVs. SARS-CoV-2-reactive CD4+ T cells from COVID-19 patients lacked cross-reactivity to CCCoVs, irrespective of strong memory T cell responses against CCCoV in all donors analyzed. In severe but not mild COVID-19, SARS-CoV-2-specific T cells displayed low functional avidity and clonality, despite increased frequencies. Our findings identify low-avidity CD4+ T cell responses as a hallmark of severe COVID-19 and argue against a protective role for CCCoV-reactive T cells in SARS-CoV-2 infection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , COVID-19/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Rhinovirus/inmunología , SARS-CoV-2/inmunología , Antígenos Virales/inmunología , Células Cultivadas , Reacciones Cruzadas , Progresión de la Enfermedad , Exposición a Riesgos Ambientales , Humanos , Memoria Inmunológica , Activación de Linfocitos , Unión Proteica , Índice de Severidad de la Enfermedad , Especificidad del Receptor de Antígeno de Linfocitos T
3.
Cell ; 148(6): 1293-307, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22424236

RESUMEN

Personalized medicine is expected to benefit from combining genomic information with regular monitoring of physiological states by multiple high-throughput methods. Here, we present an integrative personal omics profile (iPOP), an analysis that combines genomic, transcriptomic, proteomic, metabolomic, and autoantibody profiles from a single individual over a 14 month period. Our iPOP analysis revealed various medical risks, including type 2 diabetes. It also uncovered extensive, dynamic changes in diverse molecular components and biological pathways across healthy and diseased conditions. Extremely high-coverage genomic and transcriptomic data, which provide the basis of our iPOP, revealed extensive heteroallelic changes during healthy and diseased states and an unexpected RNA editing mechanism. This study demonstrates that longitudinal iPOP can be used to interpret healthy and diseased states by connecting genomic information with additional dynamic omics activity.


Asunto(s)
Genoma Humano , Genómica , Medicina de Precisión , Diabetes Mellitus Tipo 2/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Metabolómica , Persona de Mediana Edad , Mutación , Proteómica , Virus Sincitiales Respiratorios/aislamiento & purificación , Rhinovirus/aislamiento & purificación
4.
Proc Natl Acad Sci U S A ; 121(21): e2402540121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38758698

RESUMEN

All respiratory viruses establish primary infections in the nasal epithelium, where efficient innate immune induction may prevent dissemination to the lower airway and thus minimize pathogenesis. Human coronaviruses (HCoVs) cause a range of pathologies, but the host and viral determinants of disease during common cold versus lethal HCoV infections are poorly understood. We model the initial site of infection using primary nasal epithelial cells cultured at an air-liquid interface (ALI). HCoV-229E, HCoV-NL63, and human rhinovirus-16 are common cold-associated viruses that exhibit unique features in this model: early induction of antiviral interferon (IFN) signaling, IFN-mediated viral clearance, and preferential replication at nasal airway temperature (33 °C) which confers muted host IFN responses. In contrast, lethal SARS-CoV-2 and MERS-CoV encode antagonist proteins that prevent IFN-mediated clearance in nasal cultures. Our study identifies features shared among common cold-associated viruses, highlighting nasal innate immune responses as predictive of infection outcomes and nasally directed IFNs as potential therapeutics.


Asunto(s)
Resfriado Común , Inmunidad Innata , Interferones , Mucosa Nasal , SARS-CoV-2 , Transducción de Señal , Humanos , Mucosa Nasal/virología , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Interferones/metabolismo , Interferones/inmunología , Resfriado Común/inmunología , Resfriado Común/virología , Transducción de Señal/inmunología , SARS-CoV-2/inmunología , Replicación Viral , Rhinovirus/inmunología , Coronavirus Humano 229E/inmunología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Células Epiteliales/virología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Coronavirus Humano NL63/inmunología
5.
EMBO Rep ; 25(3): 1156-1175, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38332148

RESUMEN

Human rhinovirus is the most frequently isolated virus during severe exacerbations of chronic respiratory diseases, like chronic obstructive pulmonary disease. In this disease, alveolar macrophages display significantly diminished phagocytic functions that could be associated with bacterial superinfections. However, how human rhinovirus affects the functions of macrophages is largely unknown. Macrophages treated with HRV16 demonstrate deficient bacteria-killing activity, impaired phagolysosome biogenesis, and altered intracellular compartments. Using RNA sequencing, we identify the small GTPase ARL5b to be upregulated by the virus in primary human macrophages. Importantly, depletion of ARL5b rescues bacterial clearance and localization of endosomal markers in macrophages upon HRV16 exposure. In permissive cells, depletion of ARL5b increases the secretion of HRV16 virions. Thus, we identify ARL5b as a novel regulator of intracellular trafficking dynamics and phagolysosomal biogenesis in macrophages and as a restriction factor of HRV16 in permissive cells.


Asunto(s)
Macrófagos , Rhinovirus , Humanos , Macrófagos/microbiología , Macrófagos Alveolares , Fagocitosis , Bacterias
6.
Nucleic Acids Res ; 52(D1): D770-D776, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-37930838

RESUMEN

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


Asunto(s)
Asma , Infecciones por Enterovirus , Infecciones por Picornaviridae , Rhinovirus , Humanos , Genómica , Filogenia , Infecciones por Picornaviridae/genética , Rhinovirus/genética
7.
J Virol ; 98(10): e0149924, 2024 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-39324790

RESUMEN

Human rhinovirus (HRV) infections are the leading cause of disease exacerbations in individuals with chronic pulmonary diseases, primarily due to impaired macrophage functions, resulting in defective bacterial elimination. We previously demonstrated that HRV16 impairs macrophages' functions in an ARL5b-dependent manner. In permissive cells, ARL5b acted as an HRV16 restriction factor and was repressed. Here, we delve into the dual regulation of ARL5b by HRV16 in both cell types. We analyzed the effect of HRV16 on primary human macrophages using neutralizing antibodies, specific inhibitors, siRNA, and chromatin immune precipitation. Our study reveals that, while the virus does not replicate in macrophages, it induces interferon and pro-inflammatory responses. We identify the ICAM-1-PKR-ATF2 signaling axis as crucial for ARL5b induction in macrophages, whereas only ICAM-1 plays a role in ARL5b repression in permissive cells. Furthermore, HRV16 triggers epigenetic reprogramming in both cell types at the ARL5b promoter. In macrophages, epigenetic changes are ATF2 dependent. In conclusion, our findings highlight previously unknown signaling pathways activated by HRV16 in macrophages. Targeting these pathways could offer novel strategies to improve outcomes for individuals with respiratory conditions. IMPORTANCE: Human rhinovirus (HRV) infections are the leading cause of disease exacerbations in individuals with chronic pulmonary conditions and are frequently associated with bacterial superinfections due to defective bacterial elimination by macrophages. We previously identified ARL5b-induction by HRV16 to be responsible for the impairment of bacteria elimination. In contrast, in permissive cells, ARL5b is repressed and acts as a restriction factor for HRV16. Here, we investigated the dual regulation of ARL5b by HRV16 in these cells. Our study reveals that the ICAM-1-PKR-ATF2 signaling axis is crucial for ARL5b induction in macrophages. In permissive cells, only ICAM-1 plays a role in ARL5b repression. Moreover, HRV16 triggered epigenetic reprogramming in macrophages. ARL5b promoter was repressed in an ATF2-dependent manner. Collectively, our findings reveal previously unknown signaling pathways activated by HRV16 in macrophages. Targeting these pathways provides novel strategies to target ARL5b expression specifically in macrophages and improve outcomes for individuals with respiratory pathologies.


Asunto(s)
Factor de Transcripción Activador 2 , Molécula 1 de Adhesión Intercelular , Macrófagos , Rhinovirus , Transducción de Señal , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Macrófagos/virología , Macrófagos/metabolismo , Rhinovirus/fisiología , Factor de Transcripción Activador 2/metabolismo , Factor de Transcripción Activador 2/genética , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/genética , Regiones Promotoras Genéticas , Epigénesis Genética
8.
Rev Med Virol ; 34(2): e2526, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38446531

RESUMEN

miRNAs are single-stranded ncRNAs that act as regulators of different human body processes. Several miRNAs have been noted to control the human immune and inflammatory response during severe acute respiratory infection syndrome (SARS-CoV-2) infection. Similarly, many miRNAs were upregulated and downregulated during different respiratory virus infections. Here, an attempt has been made to capture the regulatory role of miRNAs in the human immune and inflammatory response during the infection of SARS-CoV-2 and other respiratory viruses. Firstly, the role of miRNAs has been depicted in the human immune and inflammatory response during the infection of SARS-CoV-2. In this direction, several significant points have been discussed about SARS-CoV-2 infection, such as the role of miRNAs in human innate immune response; miRNAs and its regulation of granulocytes; the role of miRNAs in macrophage activation and polarisation; miRNAs and neutrophil extracellular trap formation; miRNA-related inflammatory response; and miRNAs association in adaptive immunity. Secondly, the miRNAs landscape has been depicted during human respiratory virus infections such as human coronavirus, respiratory syncytial virus, influenza virus, rhinovirus, and human metapneumovirus. The article will provide more understanding of the miRNA-controlled mechanism of the immune and inflammatory response during COVID-19, which will help more therapeutics discoveries to fight against the future pandemic.


Asunto(s)
COVID-19 , MicroARNs , Humanos , MicroARNs/genética , SARS-CoV-2/genética , COVID-19/genética , Virus Sincitiales Respiratorios , Rhinovirus
9.
J Allergy Clin Immunol ; 154(3): 571-579.e6, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38761997

RESUMEN

BACKGROUND: Rhinovirus (RV) infections trigger wheeze episodes in children. Thus, understanding of the lung inflammatory response to RV in children with wheeze is important. OBJECTIVES: This study sought to examine the associations of RV on bronchoalveolar lavage (BAL) granulocyte patterns and biomarkers of inflammation with age in children with treatment-refractory, recurrent wheeze (n = 616). METHODS: Children underwent BAL to examine viral nucleic acid sequences, bacterial cultures, granulocyte counts, and phlebotomy for both general and type-2 inflammatory markers. RESULTS: Despite the absence of cold symptoms, RV was the most common pathogen detected (30%), and when present, was accompanied by BAL granulocytosis in 75% of children. Compared to children with no BAL pathogens (n = 341), those with RV alone (n = 127) had greater (P < .05) isolated neutrophilia (43% vs 16%), mixed eosinophils and neutrophils (26% vs 11%), and less pauci-granulocytic (27% vs 61%) BAL. Children with RV alone furthermore had biomarkers of active infection with higher total blood neutrophils and serum C-reactive protein, but no differences in blood eosinophils or total IgE. With advancing age, the log odds of BAL RV alone were lower, 0.82 (5th-95th percentile CI: 0.76-0.88; P < .001), but higher, 1.58 (5th-95th percentile CI: 1.01-2.51; P = .04), with high-dose daily corticosteroid treatment. CONCLUSIONS: Children with severe recurrent wheeze often (22%) have a silent syndrome of lung RV infection with granulocytic bronchoalveolitis and elevated systemic markers of inflammation. The syndrome is less prevalent by school age and is not informed by markers of type-2 inflammation. The investigators speculate that dysregulated mucosal innate antiviral immunity is a responsible mechanism.


Asunto(s)
Infecciones por Picornaviridae , Ruidos Respiratorios , Rhinovirus , Humanos , Rhinovirus/inmunología , Masculino , Femenino , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/complicaciones , Niño , Preescolar , Líquido del Lavado Bronquioalveolar/virología , Líquido del Lavado Bronquioalveolar/inmunología , Biomarcadores , Síndrome , Recurrencia , Lactante , Granulocitos/inmunología , Adolescente
10.
J Allergy Clin Immunol ; 154(2): 308-315, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38494094

RESUMEN

BACKGROUND: Single nucleotide polymorphisms (SNPs) in genes on chromosome 17q12-q21 are associated with childhood-onset asthma and rhinovirus-induced wheeze. There are few mechanistic data linking chromosome 17q12-q21 to wheezing illness. OBJECTIVE: We investigated whether 17q12-q21 risk alleles were associated with impaired interferon responses to rhinovirus. METHODS: In a population-based birth cohort of European ancestry, we stimulated peripheral blood mononuclear cells with rhinovirus A1 (RV-A1) and rhinovirus A16 (RV-A16) and measured IFN and IFN-induced C-X-C motif chemokine ligand 10 (aka IP10) responses in supernatants. We investigated associations between virus-induced cytokines and 6 SNPs in 17q12-q21. Bayesian profile regression was applied to identify clusters of individuals with different immune response profiles and genetic variants. RESULTS: Five SNPs (in high linkage disequilibrium, r2 ≥ 0.8) were significantly associated with RV-A1-induced IFN-ß (rs9303277, P = .010; rs11557467, P = .012; rs2290400, P = .006; rs7216389, P = .008; rs8079416, P = .005). A reduction in RV-A1-induced IFN-ß was observed among individuals with asthma risk alleles. There were no significant associations for RV-A1-induced IFN-α or CXCL10, or for any RV-A16-induced IFN/CXCL10. Bayesian profile regression analysis identified 3 clusters that differed in IFN-ß induction to RV-A1 (low, medium, high). The typical genetic profile of the cluster associated with low RV-A1-induced IFN-ß responses was characterized by a very high probability of being homozygous for the asthma risk allele for all SNPs. Children with persistent wheeze were almost 3 times more likely to be in clusters with reduced/average RV-A1-induced IFN-ß responses than in the high immune response cluster. CONCLUSIONS: Polymorphisms on chromosome 17q12-q21 are associated with rhinovirus-induced IFN-ß, suggesting a novel mechanism-impaired IFN-ß induction-links 17q12-q21 risk alleles with asthma/wheeze.


Asunto(s)
Cromosomas Humanos Par 17 , Polimorfismo de Nucleótido Simple , Rhinovirus , Humanos , Cromosomas Humanos Par 17/genética , Masculino , Femenino , Asma/genética , Asma/inmunología , Interferones , Niño , Ruidos Respiratorios/genética , Ruidos Respiratorios/inmunología , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/genética , Predisposición Genética a la Enfermedad , Quimiocina CXCL10/genética , Leucocitos Mononucleares/inmunología , Preescolar
11.
J Infect Dis ; 229(2): 403-412, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-37486790

RESUMEN

BACKGROUND: Rhinovirus (RV) infections can progress from the upper (URT) to lower (LRT) respiratory tract in immunocompromised individuals, causing high rates of fatal pneumonia. Little is known about how RV evolves within hosts during infection. METHODS: We sequenced RV complete genomes from 12 hematopoietic cell transplant patients with infection for up to 190 days from both URT (nasal wash, NW) and LRT (bronchoalveolar lavage, BAL). Metagenomic and amplicon next-generation sequencing were used to track the emergence and evolution of intrahost single nucleotide variants (iSNVs). RESULTS: Identical RV intrahost populations in matched NW and BAL specimens indicated no genetic adaptation is required for RV to progress from URT to LRT. Coding iSNVs were 2.3-fold more prevalent in capsid over nonstructural genes. iSNVs modeled were significantly more likely to be found in capsid surface residues, but were not preferentially located in known RV-neutralizing antibody epitopes. Newly emergent, genotype-matched iSNV haplotypes from immunocompromised individuals in 2008-2010 could be detected in Seattle-area community RV sequences in 2020-2021. CONCLUSIONS: RV infections in immunocompromised hosts can progress from URT to LRT with no specific evolutionary requirement. Capsid proteins carry the highest variability and emergent mutations can be detected in other, including future, RV sequences.


Asunto(s)
Infecciones por Enterovirus , Trasplante de Células Madre Hematopoyéticas , Humanos , Proteínas de la Cápside/genética , Cápside , Rhinovirus/genética , Mutación
12.
J Infect Dis ; 229(2): 422-431, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-37531658

RESUMEN

BACKGROUND: The epidemiology of respiratory viral infections is complex. How infection with one respiratory virus affects risk of subsequent infection with the same or another respiratory virus is not well described. METHODS: From October 2019 to June 2021, enrolled households completed active surveillance for acute respiratory illness (ARI), and participants with ARI self-collected nasal swab specimens; after April 2020, participants with ARI or laboratory-confirmed severe acute respiratory syndrome coronavirus 2 and their household members self-collected nasal swab specimens. Specimens were tested using multiplex reverse-transcription polymerase chain reaction for respiratory viruses. A Cox regression model with a time-dependent covariate examined risk of subsequent detections following a specific primary viral detection. RESULTS: Rhinovirus was the most frequently detected pathogen in study specimens (406 [9.5%]). Among 51 participants with multiple viral detections, rhinovirus to seasonal coronavirus (8 [14.8%]) was the most common viral detection pairing. Relative to no primary detection, there was a 1.03-2.06-fold increase in risk of subsequent virus detection in the 90 days after primary detection; risk varied by primary virus: human parainfluenza virus, rhinovirus, and respiratory syncytial virus were statistically significant. CONCLUSIONS: Primary virus detection was associated with higher risk of subsequent virus detection within the first 90 days after primary detection.


Asunto(s)
Infecciones por Enterovirus , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio , Virosis , Virus , Humanos , Lactante , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/epidemiología , Washingtón/epidemiología , Virus/genética , Rhinovirus/genética
13.
Am J Physiol Lung Cell Mol Physiol ; 327(4): L557-L573, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39189801

RESUMEN

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.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Coronavirus Humano NL63 , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Infecciones por Picornaviridae , Rhinovirus , Animales , Humanos , Ratones , Coronavirus Humano NL63/fisiología , Rhinovirus/fisiología , Rhinovirus/patogenicidad , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/patología , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Replicación Viral , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/metabolismo , Ratones Transgénicos , Pulmón/virología , Pulmón/patología , Pulmón/metabolismo , Células HeLa , Líquido del Lavado Bronquioalveolar/virología , Enterovirus
14.
Antimicrob Agents Chemother ; 68(3): e0106423, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38349161

RESUMEN

Screening a library of 1,200 preselected kinase inhibitors for anti-human rhinovirus 2 (HRV-2) activity in HeLa cells identified a class of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKI) as effective virus blockers. These were based on the 4-anilinoquinazoline-7-oxypiperidine scaffold, with the most potent representative AZ5385 inhibiting the virus with EC50 of 0.35 µM. Several structurally related analogs confirmed activity in the low µM range, while interestingly, other TKIs targeting EGFR lacked anti-HRV-2 activity. To further probe this lack of association between antiviral activity and EGFR inhibition, we stained infected cells with antibodies specific for activated EGFR (Y1068) and did not observe a dependency on EGFR-TK activity. Instead, consecutive passages of HRV-2 in HeLa cells in the presence of a compound and subsequent nucleotide sequence analysis of resistant viral variants identified the S181T and T210A alterations in the major capsid VP1 protein, with both residues located in the vicinity of a known hydrophobic pocket on the viral capsid. Further characterization of the antiviral effects of AZ5385 showed a modest virus-inactivating (virucidal) activity, while anti-HRV-2 activity was still evident when the inhibitor was added as late as 10 h post infection. The RNA copy/infectivity ratio of HRV-2 propagated in AZ5385 presence was substantially higher than that of control HRV indicating that the compound preferentially targeted HRV progeny virions during their maturation in infected cells. Besides HRV, the compound showed anti-respiratory syncytial virus activity, which warrants its further studies as a candidate compound against viral respiratory infections.


Asunto(s)
Rhinovirus , Humanos , Rhinovirus/química , Rhinovirus/genética , Células HeLa , Proteínas de la Cápside , Antivirales/química , Receptores ErbB
15.
J Gen Virol ; 105(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38717926

RESUMEN

Background. Respiratory tract infections are among the most important causes of mortality and morbidity in children worldwide. The COVID-19 pandemic has affected the distribution of seasonal respiratory viruses as in all areas of life. In this study, we have aimed to evaluate the changes in the rates of seasonal respiratory viruses with the onset of the pandemic.Methods. This study included patients who were admitted to the Pediatrics Clinic of Eskisehir Osmangazi University Faculty of Medicine Hospital between December 2018 and February 2022 with respiratory tract infections and in whom pathogens were detected from nasopharyngeal swab samples analysed by multiplex PCR method.Results. A total of 833 respiratory tract pathogens were detected in 684 cases consisting of male (55.3 %), and female (44.7 %), patients with a total mean age of 42 months. Single pathogen was revealed in 550, and multiple pathogens in 134 cases. Intensive care was needed in 14 % of the cases. Most frequently influenza A/B, rhinovirus and respiratory syncytial virus (RSV) were detected during the pre-pandemic period, while rhinovirus, RSV, and adenovirus were observed during the lockdown period. In the post-lockdown period, the incidence rates of rhinovirus, RSV, human bocavirus (HboV) (12 %), influenza virus infections increased, and patients with RSV and bocavirus infections required intensive care hospitalization.Conclusion. It is thought that the COVID-9 pandemic lockdown measures may have an impact on the distribution of seasonal respiratory viruses, especially RSV and influenza. Current, prospective and large case series regarding the mechanism of action and dynamics are needed.


Asunto(s)
COVID-19 , Infecciones del Sistema Respiratorio , SARS-CoV-2 , Estaciones del Año , Humanos , Femenino , Masculino , COVID-19/epidemiología , COVID-19/virología , Preescolar , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Lactante , SARS-CoV-2/genética , SARS-CoV-2/aislamiento & purificación , Niño , Rhinovirus/aislamiento & purificación , Rhinovirus/genética , Nasofaringe/virología , Adolescente , Gripe Humana/epidemiología , Gripe Humana/virología , Pandemias , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/virología
16.
Small ; 20(6): e2304722, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37806749

RESUMEN

Infection of humans by many viruses is typically initiated by the internalization of a single virion in each of a few susceptible cells. Thus, the outcome of the infection process may depend on stochastic single-molecule events. A crucial process for viral infection, and thus a target for developing antiviral drugs, is the uncoating of the viral genome. Here a force spectroscopy procedure using an atomic force microscope is implemented to study uncoating for individual human rhinovirus particles. Application of an increasing mechanical force on a virion led to a high force-induced structural transition that facilitated extrusion of the viral RNA molecule without loss of capsid integrity. Application of force to virions that h ad previously extruded the RNA, or to RNA-free capsids, led to a lower force-induced event associated with capsid disruption. The kinetic parameters are determined for each reaction. The high-force event is a stochastic process governed by a moderate free energy barrier (≈20 kcal mol-1 ), which results in a heterogeneous population of structurally weakened virions in which different fractions of the RNA molecule are externalized. The effects of antiviral compounds or capsid mutation on the kinetics of this reaction reveal a correlation between the reaction rate and virus infectivity.


Asunto(s)
Proteínas de la Cápside , Rhinovirus , Humanos , Rhinovirus/genética , Cápside/química , ARN Viral/genética , Antivirales/farmacología , Virión
17.
J Med Virol ; 96(8): e29804, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39092809

RESUMEN

Although rhinoviruses play a major role in exacerbations of childhood asthma, the presence of rhinovirus (RV) RNA in plasma, referred to as viremia, has been investigated in a few studies. The aim of the study was to investigate the presence of rhinovirus viremia at the time of asthma exacerbation and to describe the molecular characteristics of rhinoviruses associated with viremia. We conducted an observational, prospective, multicenter study in eight pediatric hospitals (VIRASTHMA2). Preschool-aged recurrent wheezers (1-5 years) hospitalized for a severe exacerbation were included. Reverse-transcription polymerase chain reaction (RT-PCR) and molecular typing for RV/enteroviruses (EV) were performed on nasal swabs and plasma. Plasma specimens were available for 105 children with positive RT-PCR for RV/EV in respiratory specimens. Thirty-six (34.3%) had positive viremia. In plasma, 28 (82.4%) of the typable specimens were RV-C, five (14.7%) were EV-D68, and one was RV-A (2.9%). In all cases, the RV/EV type was identical in the plasma and respiratory specimens. In conclusion, RV/EV viremia is frequent in severe exacerbations of preschool recurrent wheezers, particularly in RV-C infections.


Asunto(s)
Asma , Infecciones por Picornaviridae , Rhinovirus , Viremia , Humanos , Viremia/virología , Preescolar , Rhinovirus/genética , Rhinovirus/aislamiento & purificación , Rhinovirus/clasificación , Asma/virología , Masculino , Femenino , Estudios Prospectivos , Infecciones por Picornaviridae/virología , Lactante , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Plasma/virología
18.
J Med Virol ; 96(9): e29833, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39233489

RESUMEN

Rhinoviruses (RVs) are a leading cause of acute respiratory infections (ARI) in children. The relationship between RV viral loads (VL), RV/viral-co-detections and disease severity, is incompletely understood. We studied children and adolescents ≤21 years with RV-ARI that were identified as inpatients or outpatients using a PCR panel from 2011-2013. RV VL were stratified according to cycle threshold (CT) values in high (≤25), intermediate (26-32) and low (>32). Adjusted analyses were performed to assess the role RV VL and RV/viral codetections on hospital admission, oxygen requirement, PICU care, and length of stay. Of 1,899 children with RV-ARI, 78% had chronic comorbidities and 24% RV/viral co-detections. Single RV vs RV/viral co-detections was associated with higher VL (24.74 vs 26.62 CT; p = 0.001) and older age (14.9 vs 9.5 months; p = 0.0001). Frequency of RV/viral co-detections were inversely proportional to RV loads: 32% with low; 28% with intermediate, and 19% with high VL, p = 0.0001. Underlying conditions were independently associated with all clinical outcomes, high VL with PICU care, and single RV-ARI with higher odds of hospitalization. In summary, single RV vs RV/viral co-detections were associated with higher VL and older age. Underlying diseases, rather than RV loads or RV/viral co-detections, consistently predicted worse clinical outcomes.


Asunto(s)
Coinfección , Comorbilidad , Infecciones por Picornaviridae , Infecciones del Sistema Respiratorio , Rhinovirus , Índice de Severidad de la Enfermedad , Carga Viral , Humanos , Rhinovirus/genética , Rhinovirus/aislamiento & purificación , Masculino , Femenino , Niño , Preescolar , Adolescente , Lactante , Infecciones por Picornaviridae/epidemiología , Infecciones por Picornaviridae/virología , Coinfección/virología , Coinfección/epidemiología , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/epidemiología , Hospitalización/estadística & datos numéricos , Adulto Joven
19.
J Med Virol ; 96(6): e29755, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38922896

RESUMEN

Throughout the COVID-19 pandemic, rhinovirus (RV) remained notable persistence, maintaining its presence while other seasonal respiratory viruses were largely suppressed by pandemic restrictions during national lockdowns. This research explores the epidemiological dynamics of RV infections among pediatric populations on Hainan Island, China, specifically focusing on the impact before and after the zero-COVID policy was lifted. From January 2021 to December 2023, 19 680 samples were collected from pediatric patients hospitalized with acute lower respiratory tract infections (ARTIs) at the Hainan Maternal and Child Health Hospital. The infection of RV was detected by tNGS. RV species and subtypes were identified in 32 RV-positive samples representing diverse time points by analyzing the VP4/VP2 partial regions. Among the 19 680 pediatric inpatients with ARTIs analyzed, 21.55% were found to be positive for RV infection, with notable peaks observed in April 2021 and November 2022. A gradual annual decline in RV infections was observed, alongside a seasonal pattern of higher prevalence during the colder months. The highest proportion of RV infections was observed in the 0-1-year age group. Phylogenetic analysis on 32 samples indicated a trend from RV-A to RV-C in 2022. This observation suggests potential evolving dynamics within the RV species although further studies are needed due to the limited sample size. The research emphasizes the necessity for ongoing surveillance and targeted management, particularly for populations highly susceptible to severe illnesses caused by RV infections.


Asunto(s)
COVID-19 , Variación Genética , Filogenia , Infecciones por Picornaviridae , Infecciones del Sistema Respiratorio , Rhinovirus , Humanos , Rhinovirus/genética , Rhinovirus/clasificación , Rhinovirus/aislamiento & purificación , China/epidemiología , Lactante , Preescolar , Infecciones por Picornaviridae/epidemiología , Infecciones por Picornaviridae/virología , Niño , Femenino , Masculino , COVID-19/epidemiología , COVID-19/virología , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/epidemiología , Recién Nacido , Estaciones del Año , Adolescente , Prevalencia , Niño Hospitalizado/estadística & datos numéricos , SARS-CoV-2/genética , Hospitalización/estadística & datos numéricos
20.
J Med Virol ; 96(8): e29890, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39188069

RESUMEN

Prolonged positive polymerase chain reaction (PCR) results, irrespective of the transmission risk, can lead to prolonged restrictions on daily activities and infection precaution interventions. Studies evaluating the duration of PCR positivity for multiple pathogens in a single patient cohort are scarce. This study aimed to evaluate and compare the durations of PCR positivity for multiple respiratory viruses among children and adolescents. This retrospective study was conducted between April 2018 and March 2024 using a multiplex PCR respiratory panel for symptomatic children and adolescents who had at least two tests within 90 days of study period, with the first PCR test positive. The rate and likelihood of persistent PCR positivity were evaluated for multiple respiratory viruses. For 1325 positive results, repeat tests were conducted within 90 days. The persistent PCR positivity rate at repeat testing decreased over time (60.6%, Days 1-15 and 21.7%, Days 76-90, after the first test). In multivariate logistic regression analysis, an increased likelihood of persistent PCR positivity was observed for rhinovirus/enterovirus and adenovirus, whereas decreased likelihood of persistent positivity was seen in influenza and seasonal coronaviruses, compared with parainfluenza viruses. Persistent PCR positivity is common for multiple respiratory viruses in symptomatic children.


Asunto(s)
Reacción en Cadena de la Polimerasa Multiplex , Infecciones del Sistema Respiratorio , Humanos , Reacción en Cadena de la Polimerasa Multiplex/métodos , Niño , Estudios Retrospectivos , Infecciones del Sistema Respiratorio/virología , Infecciones del Sistema Respiratorio/diagnóstico , Preescolar , Femenino , Masculino , Adolescente , Lactante , Virus/aislamiento & purificación , Virus/genética , Virus/clasificación , Virosis/diagnóstico , Virosis/virología , Factores de Tiempo , Rhinovirus/genética , Rhinovirus/aislamiento & purificación , Enterovirus/genética , Enterovirus/aislamiento & purificación , Enterovirus/clasificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA