Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 362
Filter
1.
Nat Commun ; 14(1): 2329, 2023 04 22.
Article in English | MEDLINE | ID: mdl-37087523

ABSTRACT

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


Subject(s)
Antiviral Restriction Factors , Asthma , COVID-19 , DEAD Box Protein 58 , Inflammasomes , Rhinovirus , Humans , Antiviral Restriction Factors/genetics , Antiviral Restriction Factors/metabolism , Asthma/genetics , Asthma/immunology , COVID-19/genetics , COVID-19/immunology , DEAD Box Protein 58/metabolism , Enterovirus Infections/genetics , Enterovirus Infections/immunology , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation , Interferon Type I , Picornaviridae Infections/genetics , Picornaviridae Infections/immunology , Rhinovirus/metabolism , Rhinovirus/pathogenicity , SARS-CoV-2
2.
Int J Mol Sci ; 24(4)2023 Feb 11.
Article in English | MEDLINE | ID: mdl-36835041

ABSTRACT

Deficiencies in epithelial barrier integrity are involved in the pathogenesis of chronic rhinosinusitis (CRS). This study aimed to investigate the role of ephrinA1/ephA2 signaling on sinonasal epithelial permeability and rhinovirus-induced epithelial permeability. This role in the process of epithelial permeability was evaluated by stimulating ephA2 with ephrinA1 and inactivating ephA2 with ephA2 siRNA or inhibitor in cells exposed to rhinovirus infection. EphrinA1 treatment increased epithelial permeability, which was associated with decreased expression of ZO-1, ZO-2, and occludin. These effects of ephrinA1 were attenuated by blocking the action of ephA2 with ephA2 siRNA or inhibitor. Furthermore, rhinovirus infection upregulated the expression levels of ephrinA1 and ephA2, increasing epithelial permeability, which was suppressed in ephA2-deficient cells. These results suggest a novel role of ephrinA1/ephA2 signaling in epithelial barrier integrity in the sinonasal epithelium, suggesting their participation in rhinovirus-induced epithelial dysfunction.


Subject(s)
Cell Membrane Permeability , Epithelial Cells , Receptor, EphA1 , Receptor, EphA2 , Humans , Cell Membrane Permeability/genetics , Cell Membrane Permeability/physiology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Picornaviridae Infections/metabolism , Receptor, EphA2/metabolism , Rhinovirus/pathogenicity , RNA, Double-Stranded , RNA, Small Interfering/metabolism , Signal Transduction/physiology
3.
Respir Physiol Neurobiol ; 298: 103846, 2022 04.
Article in English | MEDLINE | ID: mdl-35063696

ABSTRACT

A significant proportion of chronic obstructive pulmonary disease exacerbations are strongly associated with rhinovirus infection (HRV). In this study, we combined long-term cigarette smoke exposure with HRV infection in a mouse model. Our aim was to better understand the effects of HRV infection on such exacerbations, using a realistic method for generating a COPD-like phenotype. After 12-weeks of cigarette smoke exposure, adult female BALB/c mice were infected with HRV-1A and three days later we assessed a range of outcomes including lung volume and function, collected lung tissue for measurement of viral titre, bronchoalveolar lavage for assessment of pulmonary inflammation and levels of key mediators, and fixed lungs for stereological structural analyses. Cigarette smoke exposure alone significantly increased total cells and macrophages, and reduced MIP-2 in bronchoalveolar lavage. HRV-1A infection alone increased neutrophilic inflammation, IP-10 and total protein in lavage and also increased specific airway resistance measured at functional residual capacity. Cigarette smoke and HRV-1A together impacted various lung structural parameters including increasing stereological lung volume. Our results show that long-term cigarette smoke exposure and HRV-1A infection both individually impact respiratory outcomes and combine to alter aspects of lung structure in a mouse model, thus providing insight into the development of future mechanistic studies and appropriate interventions in human disease.


Subject(s)
Cigarette Smoking/adverse effects , Inhalation Exposure/adverse effects , Picornaviridae Infections/complications , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/pathology , Rhinovirus/pathogenicity , Symptom Flare Up , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism
4.
PLoS One ; 16(11): e0259859, 2021.
Article in English | MEDLINE | ID: mdl-34807924

ABSTRACT

Rhinoviruses (RV) are a major cause of Severe Acute Respiratory Infection (SARI) in children, with high genotypic diversity in different regions. However, RV type diversity remains unknown in several regions of the world. In this study, the genetic variability of the frequently circulating RV types in Northern Tunisia was investigated, using phylogenetic and phylogeographic analyses with a specific focus on the most frequent RV types: RV-A101 and RV-C45. This study concerned 13 RV types frequently circulating in Northern Tunisia. They were obtained from respiratory samples collected in 271 pediatric SARI cases, between September 2015 and November 2017. A total of 37 RV VP4-VP2 sequences, selected among a total of 49 generated sequences, was compared to 359 sequences from different regions of the world. Evolutionary analysis of RV-A101 and RV-C45 showed high genetic relationship between different Tunisian strains and Malaysian strains. RV-A101 and C45 progenitor viruses' dates were estimated in 1981 and 1995, respectively. Since the early 2000s, the two types had a wide spread throughout the world. Phylogenetic analyses of other frequently circulating strains showed significant homology of Tunisian strains from the same epidemic period, in contrast with earlier strains. The genetic relatedness of RV-A101 and RV-C45 might result from an introduction of viruses from different clades followed by local dissemination rather than a local persistence of an endemic clades along seasons. International traffic may play a key role in the spread of RV-A101, RV-C45, and other RVs.


Subject(s)
Rhinovirus/classification , Rhinovirus/genetics , Severe Acute Respiratory Syndrome/epidemiology , Biological Evolution , Capsid Proteins/genetics , Child , Child, Preschool , Epidemics , Evolution, Molecular , Female , Genetic Variation/genetics , Genotype , Humans , Infant , Phylogeny , Phylogeography/methods , Pneumonia , Rhinovirus/pathogenicity , Severe Acute Respiratory Syndrome/virology , Tunisia/epidemiology
5.
Front Immunol ; 12: 731846, 2021.
Article in English | MEDLINE | ID: mdl-34691038

ABSTRACT

The majority of asthma exacerbations in children are caused by Rhinovirus (RV), a positive sense single stranded RNA virus of the Picornavirus family. The host has developed virus defense mechanisms that are mediated by the upregulation of interferon-activated signaling. However, the virus evades the immune system by inducing immunosuppressive cytokines and surface molecules like programmed cell death protein 1 (PD-1) and its ligand (PD-L1) on immunocompetent cells. Initially, RV infects epithelial cells, which constitute a physiologic mucosal barrier. Upon virus entrance, the host cell immediately recognizes viral components like dsRNA, ssRNA, viral glycoproteins or CpG-DNA by host pattern recognition receptors (PRRs). Activation of toll like receptors (TLR) 3, 7 and 8 within the endosome and through MDA-5 and RIG-I in the cytosol leads to the production of interferon (IFN) type I and other antiviral agents. Every cell type expresses IFNAR1/IFNAR2 receptors thus allowing a generalized antiviral activity of IFN type I resulting in the inhibition of viral replication in infected cells and preventing viral spread to non-infected cells. Among immune evasion mechanisms of the virus, there is downregulation of IFN type I and its receptor as well as induction of the immunosuppressive cytokine TGF-ß. TGF-ß promotes viral replication and is associated with induction of the immunosuppression signature markers LAP3, IDO and PD-L1. This article reviews the recent advances on the regulation of interferon type I expression in association with RV infection in asthmatics and the immunosuppression induced by the virus.


Subject(s)
Asthma/virology , Common Cold/virology , Immune Evasion , Lung/virology , Rhinovirus/immunology , Adaptive Immunity , Animals , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , Common Cold/immunology , Common Cold/metabolism , Common Cold/physiopathology , Cytokines/metabolism , Disease Progression , Host-Pathogen Interactions , Humans , Immunity, Innate , Immunocompromised Host , Lung/immunology , Lung/metabolism , Lung/physiopathology , Rhinovirus/pathogenicity , Signal Transduction
6.
Physiol Rep ; 9(18): e14994, 2021 09.
Article in English | MEDLINE | ID: mdl-34542243

ABSTRACT

Nasal epithelial cells (NECs) are among the first cells to be exposed to air pollutants and respiratory viruses. Although it is known that air pollution exposure and rhinovirus infections increase the risk for asthma development independently, it is unclear how these risk factors interact on a cellular level. Therefore, we aimed to investigate how exposure to diesel particulate matter (DPM) modifies the response of primary NECs to rhinovirus (RV) infection in vitro. Exposure of re-differentiated, primary NECs (49 healthy children [0-7 years], 12 adults) to DPM modified the mRNA expression of viral cell-surface receptors, pattern recognition receptors, and pro-inflammatory response (also protein levels). After exposure to DPM, we additionally infected the NECs with RV-1b and RV-16. Viral loads (assessed by titration assays) were significantly higher in DPM-exposed compared with non-exposed NECs. Exposure to DPM prior to RV infection resulted in a significant upregulation of pro-inflammatory cytokines (mRNA and protein level) and ß-defensins mRNA, and significant downregulation of pattern recognition receptors mRNA and CXCL10 (mRNA and protein levels). There was no difference between all outcomes of NECs from children and adults. We can conclude that exposure to DPM prior to RV infection increases viral loads by downregulation of viral defense receptors and upregulation of pro-inflammatory cytokines. Our findings indicate a strong interaction between air pollution and the antiviral response to RV infection in NECs. We provide mechanistic evidence that exposure to air pollution increases susceptibility to RV infection.


Subject(s)
Air Pollutants/toxicity , Nasal Mucosa/drug effects , Particulate Matter/toxicity , Picornaviridae Infections/immunology , Vehicle Emissions/toxicity , Adult , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Child , Child, Preschool , Humans , Infant , Nasal Mucosa/immunology , Nasal Mucosa/virology , Rhinovirus/pathogenicity
7.
Sci Rep ; 11(1): 17193, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433834

ABSTRACT

This paper addresses the development of predictive models for distinguishing pre-symptomatic infections from uninfected individuals. Our machine learning experiments are conducted on publicly available challenge studies that collected whole-blood transcriptomics data from individuals infected with HRV, RSV, H1N1, and H3N2. We address the problem of identifying discriminatory biomarkers between controls and eventual shedders in the first 32 h post-infection. Our exploratory analysis shows that the most discriminatory biomarkers exhibit a strong dependence on time over the course of the human response to infection. We visualize the feature sets to provide evidence of the rapid evolution of the gene expression profiles. To quantify this observation, we partition the data in the first 32 h into four equal time windows of 8 h each and identify all discriminatory biomarkers using sparsity-promoting classifiers and Iterated Feature Removal. We then perform a comparative machine learning classification analysis using linear support vector machines, artificial neural networks and Centroid-Encoder. We present a range of experiments on different groupings of the diseases to demonstrate the robustness of the resulting models.


Subject(s)
Computer Simulation , Influenza, Human/virology , Picornaviridae Infections/virology , Respiratory Syncytial Virus Infections/virology , Transcriptome , Virus Shedding , Biomarkers/metabolism , Gene Regulatory Networks , Humans , Influenza, Human/genetics , Influenza, Human/metabolism , Machine Learning , Orthomyxoviridae/pathogenicity , Orthomyxoviridae/physiology , Picornaviridae Infections/genetics , Picornaviridae Infections/metabolism , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Viruses/pathogenicity , Respiratory Syncytial Viruses/physiology , Rhinovirus/pathogenicity , Rhinovirus/physiology
8.
Cell Mol Life Sci ; 78(21-22): 6735-6744, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34459952

ABSTRACT

Kallikrein-related peptidases (KLKs) or kallikreins have been linked to diverse (patho) physiological processes, such as the epidermal desquamation and inflammation, seminal clot liquefaction, neurodegeneration, and cancer. Recent mounting evidence suggests that KLKs also represent important regulators of viral infections. It is well-established that certain enveloped viruses, including influenza and coronaviruses, require proteolytic processing of their hemagglutinin or spike proteins, respectively, to infect host cells. Similarly, the capsid protein of the non-enveloped papillomavirus L1 should be proteolytically cleaved for viral uncoating. Consequently, extracellular or membrane-bound proteases of the host cells are instrumental for viral infections and represent potential targets for drug development. Here, we summarize how extracellular proteolysis mediated by the kallikreins is implicated in the process of influenza (and potentially coronavirus and papillomavirus) entry into host cells. Besides direct proteolytic activation of viruses, KLK5 and 12 promote viral entry indirectly through proteolytic cascade events, like the activation of thrombolytic enzymes that also can process hemagglutinin, while additional functions of KLKs in infection cannot be excluded. In the light of recent evidence, KLKs represent potential host targets for the development of new antivirals. Humanized animal models to validate their key functions in viral infections will be valuable.


Subject(s)
COVID-19/enzymology , COVID-19/virology , Host Microbial Interactions/physiology , Kallikreins/metabolism , SARS-CoV-2 , Virus Diseases/enzymology , Animals , Asthma/etiology , Coronavirus/genetics , Coronavirus/pathogenicity , Coronavirus/physiology , Host Microbial Interactions/genetics , Humans , Orthomyxoviridae/genetics , Orthomyxoviridae/pathogenicity , Orthomyxoviridae/physiology , Papillomavirus Infections/enzymology , Papillomavirus Infections/virology , Picornaviridae Infections/complications , Picornaviridae Infections/enzymology , Picornaviridae Infections/virology , Protein Processing, Post-Translational , Proteolysis , Rhinovirus/pathogenicity , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Varicella Zoster Virus Infection/enzymology , Varicella Zoster Virus Infection/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Diseases/virology , Virus Internalization
9.
Viruses ; 13(7)2021 06 27.
Article in English | MEDLINE | ID: mdl-34198998

ABSTRACT

Rhinovirus (RV) is commonly detected in asymptomatic children; hence, its pathogenicity during childhood pneumonia remains controversial. We evaluated RV epidemiology in HIV-uninfected children hospitalized with clinical pneumonia and among community controls. PERCH was a case-control study that enrolled children (1-59 months) hospitalized with severe and very severe pneumonia per World Health Organization clinical criteria and age-frequency-matched community controls in seven countries. Nasopharyngeal/oropharyngeal swabs were collected for all participants, combined, and tested for RV and 18 other respiratory viruses using the Fast Track multiplex real-time PCR assay. RV detection was more common among cases (24%) than controls (21%) (aOR = 1.5, 95%CI:1.3-1.6). This association was driven by the children aged 12-59 months, where 28% of cases vs. 18% of controls were RV-positive (aOR = 2.1, 95%CI:1.8-2.5). Wheezing was 1.8-fold (aOR 95%CI:1.4-2.2) more prevalent among pneumonia cases who were RV-positive vs. RV-negative. Of the RV-positive cases, 13% had a higher probability (>75%) that RV was the cause of their pneumonia based on the PERCH integrated etiology analysis; 99% of these cases occurred in children over 12 months in Bangladesh. RV was commonly identified in both cases and controls and was significantly associated with severe pneumonia status among children over 12 months of age, particularly those in Bangladesh. RV-positive pneumonia was associated with wheezing.


Subject(s)
Nasopharynx/virology , Picornaviridae Infections/epidemiology , Pneumonia, Viral/epidemiology , Rhinovirus/pathogenicity , Africa/epidemiology , Asia/epidemiology , Asian People/statistics & numerical data , Case-Control Studies , Child, Preschool , Female , Humans , Infant , Male , Picornaviridae Infections/ethnology , Pneumonia, Viral/etiology , Respiratory Sounds/etiology
10.
Pediatr Infect Dis J ; 40(10): 873-879, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34321447

ABSTRACT

BACKGROUND: Acute wheezing is one of the most common hospital presentations for young children. Respiratory syncytial virus (RSV) and rhinovirus (RV) species A, B and the more recently described species C are implicated in the majority of these presentations. However, the relative importance and age-specificities of these viruses have not been defined. Hence, this study aimed to establish these relationships in a large cohort of prospectively recruited hospitalized children. METHODS: The study cohort was 390 children 0-16 years of age presenting with acute wheezing to a children's emergency department, 96.4% being admitted. A nonwheezing control population of 190 was also recruited. Nasal samples were analyzed for viruses. RESULTS: For the first 6 months of life, RSV was the dominant virus associated with wheezing (P < 0.001). From 6 months to 2 years, RSV, RV-A and RV-C were all common but none predominated. From 2 to 6 years, RV-C was the dominant virus detected (50-60% of cases), 2-3 times more common than RV-A and RSV, RSV decreasing to be absent from 4 to 7 years. RV-B was rare at all ages. RV-C was no longer dominant in children more than 10 years of age. Overall, RV-C was associated with lower mean oxygen saturation than any other virus (P < 0.001). Controls had no clear age distribution of viruses. CONCLUSION: This study establishes a clear profile of age specificity of virus infections causing moderate to severe wheezing in children: RSV as the dominant cause in the first 6 months and RV-C in preschool-age children.


Subject(s)
Hospitalization/statistics & numerical data , Respiratory Sounds/etiology , Respiratory Syncytial Virus, Human/pathogenicity , Rhinovirus/pathogenicity , Acute Disease , Adolescent , Age Factors , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Male , Nose/virology , Oxygen Saturation , Picornaviridae Infections/complications , Picornaviridae Infections/virology , Prospective Studies , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/virology
11.
mSphere ; 6(3): e0047921, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34160242

ABSTRACT

Coinfection by heterologous viruses in the respiratory tract is common and can alter disease severity compared to infection by individual virus strains. We previously found that inoculation of mice with rhinovirus (RV) 2 days before inoculation with a lethal dose of influenza A virus [A/Puerto Rico/8/34 (H1N1) (PR8)] provides complete protection against mortality. Here, we extended that finding to a second lethal respiratory virus, pneumonia virus of mice (PVM), and analyzed potential mechanisms of RV-induced protection. RV completely prevented mortality and weight loss associated with PVM infection. Major changes in host gene expression upon PVM infection were delayed compared to PR8. RV induced earlier recruitment of inflammatory cells, which were reduced at later times in RV-inoculated mice. Findings common to both virus pairs included the upregulated expression of mucin-associated genes and dampening of inflammation-related genes in mice that were inoculated with RV before lethal virus infection. However, type I interferon (IFN) signaling was required for RV-mediated protection against PR8 but not PVM. IFN signaling had minor effects on PR8 replication and contributed to controlling neutrophilic inflammation and hemorrhagic lung pathology in RV/PR8-infected mice. These findings, combined with differences in virus replication levels and disease severity, suggest that the suppression of inflammation in RV/PVM-infected mice may be due to early, IFN-independent suppression of viral replication, while that in RV/PR8-infected mice may be due to IFN-dependent modulation of immune responses. Thus, a mild upper respiratory viral infection can reduce the severity of a subsequent severe viral infection in the lungs through virus-dependent mechanisms. IMPORTANCE Respiratory viruses from diverse families cocirculate in human populations and are frequently detected within the same host. Although clinical studies suggest that infection by multiple different respiratory viruses may alter disease severity, animal models in which we can control the doses, timing, and strains of coinfecting viruses are critical to understanding how coinfection affects disease severity. Here, we compared gene expression and immune cell recruitment between two pairs of viruses (RV/PR8 and RV/PVM) inoculated sequentially in mice, both of which result in reduced severity compared to lethal infection by PR8 or PVM alone. Reduced disease severity was associated with suppression of inflammatory responses in the lungs. However, differences in disease kinetics and host and viral gene expression suggest that protection by coinfection with RV may be due to distinct molecular mechanisms. Indeed, we found that antiviral cytokine signaling was required for RV-mediated protection against lethal infection by PR8 but not PVM.


Subject(s)
Coinfection/immunology , Host-Pathogen Interactions , Interferon Type I/immunology , Picornaviridae Infections/immunology , Rhinovirus/immunology , Rhinovirus/pathogenicity , Animals , Coinfection/virology , Female , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Influenza A virus/immunology , Influenza A virus/pathogenicity , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Murine pneumonia virus/immunology , Murine pneumonia virus/pathogenicity , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Pneumovirus Infections/immunology , Pneumovirus Infections/prevention & control , Severity of Illness Index , Transcriptome , Virus Replication
12.
Am J Respir Cell Mol Biol ; 65(5): 544-554, 2021 11.
Article in English | MEDLINE | ID: mdl-34181859

ABSTRACT

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


Subject(s)
Asthma/genetics , Host-Pathogen Interactions/genetics , Lung/virology , Picornaviridae Infections/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Aged , Antiviral Agents/pharmacology , Asthma/pathology , Bronchi/pathology , Bronchi/physiology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Gene Expression Profiling , Genome, Human , Humans , Isoxazoles/pharmacology , Lung/physiology , Male , Middle Aged , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Picornaviridae Infections/drug therapy , Picornaviridae Infections/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Pyrrolidinones/pharmacology , Rhinovirus/pathogenicity , Valine/analogs & derivatives , Valine/pharmacology
13.
Virus Genes ; 57(3): 276-279, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33988838

ABSTRACT

Rhinoviruses (RVs) are the most common etiological agent implicated in respiratory infections among infants and children. There are currently no approved antivirals and vaccine for use against the virus; hence, the need for information on the genotypes of rhinovirus from developing countries of the world with high burden of the infection. This study determined the genotypes of rhinovirus circulating among children in selected cities in Nigeria. Nasopharyngeal and oropharyngeal samples were carefully collected from children showing signs of respiratory infection in two communities in South-west Nigeria. Polymerase Chain Reaction was used to amplify the hypervariable part of the 5'- non-coding region, the entire viral protein gene 4 and the 5' terminus of the VP2 gene of RV. Nucleotide BLAST and phylogenetic analyses were used to genotype the isolates. Of the samples analysed, 12.7% showed rhinovirus positivity. All the three genotypes of rhinovirus were detected with genotype C (71.4%), being the predominant. Multiple strains of rhinovirus were found circulating. We showed for the first time the genotypes and strains of rhinovirus circulating in Nigeria. Further studies are required to highlight transmission patterns and disease severity among rhinovirus species in Nigeria.


Subject(s)
Respiratory Tract Infections/genetics , Rhinovirus/genetics , Viral Proteins/genetics , Child , Child, Preschool , Female , Genotype , Humans , Infant , Male , Phylogeny , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Rhinovirus/pathogenicity
14.
J Immunother Cancer ; 9(4)2021 04.
Article in English | MEDLINE | ID: mdl-33875611

ABSTRACT

BACKGROUND: While programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) antagonists have improved the prognosis for many patients with melanoma, around 60% fail therapy. PVSRIPO is a non-neurovirulent rhinovirus:poliovirus chimera that facilitates an antitumor immune response following cell entry via the poliovirus receptor CD155, which is expressed on tumor and antigen-presenting cells. Preclinical studies show that oncolytic virus plus anti-PD-1 therapy leads to a greater antitumor response than either agent alone, warranting clinical investigation. METHODS: An open-label phase I trial of intratumoral PVSRIPO in patients with unresectable melanoma (American Joint Committee on Cancer V.7 stage IIIB, IIIC, or IV) was performed. Eligible patients had disease progression on anti-PD-1 and V-raf murine sarcoma viral oncogene homolog B (BRAF)/mitogen activated protein kinase kinase (MEK) inhibitors (if BRAF mutant). The primary objective was to characterize the safety and tolerability of PVSRIPO. Twelve patients in four cohorts received a total of 1, 2 or 3 injections of PVSRIPO monotherapy, with 21 days between injections. RESULTS: PVSRIPO injections were well tolerated with no serious adverse events (SAEs) or dose-limiting toxicities (DLTs) reported. All adverse events (AEs) were grade (G) 1 or G2 (G1 pruritus most common at 58%); all but two PVSRIPO-treatment related AEs were localized to the injected or adjacent lesions (n=1 G1 hot flash, n=1 G1 fatigue). Four out of 12 patients (33%) achieved an objective response per immune-related response criteria (two observations, 4 weeks apart), including 4/6 (67%) who received three injections. In the four patients with in-transit disease, a pathological complete response (pCR) was observed in two (50%) patients. Following study completion, 11/12 patients (92%) reinitiated immune checkpoint inhibitor-based therapy, and 6/12 patients (50%) remained without progression at a median follow-up time of 18 months. CONCLUSION: Intratumoral PVSRIPO was well tolerated. Despite the limited number of PVSRIPO treatments relative to the overall lesion burden (67% patients>5 lesions), intratumoral PVSRIPO showed promising antitumor activity, with pCR in injected as well as non-injected lesions in select patients. TRIAL REGISTRATION NUMBER: NCT03712358.


Subject(s)
Melanoma/therapy , Oncolytic Virotherapy , Oncolytic Viruses/pathogenicity , Poliovirus/pathogenicity , Rhinovirus/pathogenicity , Skin Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Melanoma/immunology , Middle Aged , North Carolina , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/immunology , Poliovirus/immunology , Rhinovirus/immunology , Skin Neoplasms/immunology , Skin Neoplasms/virology , Time Factors , Treatment Outcome
15.
Viruses ; 13(4)2021 04 07.
Article in English | MEDLINE | ID: mdl-33916958

ABSTRACT

Rhinoviruses (RVs) are the etiological agents of upper respiratory tract infections, particularly the common cold. Infections in the lower respiratory tract is shown to cause severe disease and exacerbations in asthma and COPD patients. Viruses being obligate parasites, hijack host cell pathways such as programmed cell death to suppress host antiviral responses and prolong viral replication and propagation. RVs are non-enveloped positive sense RNA viruses with a lifecycle fully contained within the cytoplasm. Despite decades of study, the details of how RVs exit the infected cell are still unclear. There are some diverse studies that suggest a possible role for programmed cell death. In this review, we aimed to consolidate current literature on the impact of RVs on cell death to inform future research on the topic. We searched peer reviewed English language literature in the past 21 years for studies on the interaction with and modulation of cell death pathways by RVs, placing it in the context of the broader knowledge of these interconnected pathways from other systems. Our review strongly suggests a role for necroptosis and/or autophagy in RV release, with the caveat that all the literature is based on RV-A and RV-B strains, with no studies to date examining the interaction of RV-C strains with cell death pathways.


Subject(s)
Cell Death , Picornaviridae Infections/virology , Rhinovirus/pathogenicity , Virus Replication , Autophagy , Humans , Necroptosis , Picornaviridae Infections/complications , Rhinovirus/physiology
16.
Epidemiol Infect ; 149: e96, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33849679

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is pandemic. Prevention and control strategies require an improved understanding of SARS-CoV-2 dynamics. We did a rapid review of the literature on SARS-CoV-2 viral dynamics with a focus on infective dose. We sought comparisons of SARS-CoV-2 with other respiratory viruses including SARS-CoV-1 and Middle East respiratory syndrome coronavirus. We examined laboratory animal and human studies. The literature on infective dose, transmission and routes of exposure was limited specially in humans, and varying endpoints were used for measurement of infection. Despite variability in animal studies, there was some evidence that increased dose at exposure correlated with higher viral load clinically, and severe symptoms. Higher viral load measures did not reflect coronavirus disease 2019 severity. Aerosol transmission seemed to raise the risk of more severe respiratory complications in animals. An accurate quantitative estimate of the infective dose of SARS-CoV-2 in humans is not currently feasible and needs further research. Our review suggests that it is small, perhaps about 100 particles. Further work is also required on the relationship between routes of transmission, infective dose, co-infection and outcomes.


Subject(s)
COVID-19/transmission , SARS-CoV-2/pathogenicity , Viral Load , Adenoviridae/pathogenicity , Animals , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/virology , Chlorocebus aethiops , Communicable Disease Control , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/virology , Cricetinae , Enterovirus/pathogenicity , Ferrets , Humans , Macaca mulatta , Mice , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Orthomyxoviridae/pathogenicity , Respiratory Syncytial Viruses/pathogenicity , Rhinovirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe Acute Respiratory Syndrome/epidemiology , Severe Acute Respiratory Syndrome/transmission , Severe Acute Respiratory Syndrome/virology , Virus Diseases/epidemiology , Virus Diseases/transmission , Virus Diseases/virology
18.
Allergol Immunopathol (Madr) ; 49(1): 153-158, 2021.
Article in English | MEDLINE | ID: mdl-33528944

ABSTRACT

Although there is increasing evidence showing that infants with viral bronchiolitis exhibit a high degree of heterogeneity, a core uncertainty shared by many clinicians is with regard to understanding which patients are most likely to benefit from bronchodilators such as albuterol. Based on our review, we concluded that older infants with rhinovirus (RV) bronchiolitis, especially those with a nasopharyngeal microbiome dominated by Haemophilus influenzae; those affected during nonpeak months or during non-respiratory syncytial virus (RSV) predominant months; those with wheezing at presentation; those with clinical characteristics such as atopic dermatitis or a family history of asthma in a first-degree relative; and those infants infected with RSV genotypes ON1 and BA, have the greatest likelihood of benefiting from albuterol. Presently, this patient profile could serve as the basis for rational albuterol administration in patients with viral bronchiolitis, at least on a therapeutic trial basis, and it could also be the starting point for future targeted randomized clinical trials (RCTs) on the use of albuterol among a subset of infants with bronchiolitis.


Subject(s)
Albuterol/therapeutic use , Bronchiolitis, Viral/drug therapy , Bronchodilator Agents/therapeutic use , Bronchiolitis, Viral/diagnosis , Bronchiolitis, Viral/virology , Humans , Infant , Nasopharynx/microbiology , Phenotype , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/pathogenicity , Rhinovirus/pathogenicity , Seasons
19.
Braz J Microbiol ; 52(1): 289-298, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33410102

ABSTRACT

INTRODUCTION: Human rhinovirus (HRV) is one of the most common human viral pathogens related to infections of the upper and lower respiratory tract, which can result in bronchiolitis and pneumonia. However, the relevance of HRV in human health was under-estimated for long time due to the absence of molecular targets for influenza and influenza-like syndrome surveillance in Brasília, Brazil. OBJECTIVES: The main objective of this study was analyze the clinical characteristics and outcomes of HRV infections in comparison with patients without HRV and other common respiratory viruses. MATERIALS AND METHODS: For this purpose, new specific primer sets were designed based on the high throughput sequencing analysis in previous study. These primers were used for HRV detection by RT-qPCR and Sanger sequencing of amplified cDNA of 5' genomic region. The phylogenetic tree with representative HRV isolates was constructed using the Mega X software. Statistical analysis considering the patient profiles were performed using IBM SPSS program with non-parametric tests. RESULTS: The most prevalent virus in negative samples was rhinovirus (n = 40), including three rhinovirus species (rhinovirus A, B, and C). The odds ratio associated with HRV infection was 2.160 for patients younger than 2 years and 4.367 for people living in rural areas. The multiple analysis showed lower chance of patients with HRV presenting respiratory distress. CONCLUSION: In this study, it was reported the predominance of rhinoviruses in cases of respiratory illness for negative patients for the influenza and influenza-like syndrome surveillance, being rhinorrhea, the most significant symptom associated with the disease.


Subject(s)
Picornaviridae Infections/epidemiology , Picornaviridae Infections/virology , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Rhinovirus/genetics , Brazil/epidemiology , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Phylogeny , Picornaviridae Infections/diagnosis , Rhinovirus/classification , Rhinovirus/pathogenicity , Viruses/classification , Viruses/genetics , Viruses/isolation & purification
20.
Front Immunol ; 12: 793517, 2021.
Article in English | MEDLINE | ID: mdl-34975898

ABSTRACT

EphA2 receptor and its ephrin ligands are involved in virus infection, epithelial permeability, and chemokine secretion. We hypothesized that ephrinA1/ephA2 signaling participates in rhinovirus (RV)-induced antiviral immune response in sinonasal mucosa of patients with chronic rhinosinusitis (CRS). Therefore, we investigated the expression of ephrinA1/ephA2 in normal and inflamed sinonasal mucosa and evaluated whether they regulate chemokine secretion and the production of antiviral immune mediators including interferons (IFNs) in RV-infected human primary sinonasal epithelial cells. For this purpose, the expression and distribution of ephrinA1/ephA2 in sinonasal mucosa were evaluated with RT-qPCR, immunofluorescence, and western blot. Their roles in chemokine secretion and the production of antiviral immune mediators such as type I and III IFNs, and interferon stimulated genes were evaluated by stimulating ephA2 with ephrinA1 and inactivating ephA2 with ephA2 siRNA or inhibitor in cells exposed to RV and poly(I:C). We found that ephrinA1/ephA2 were expressed in normal mucosa and their levels increased in inflamed sinonasal mucosa of CRS patients. RV infection or poly(I:C) treatment induced chemokine secretion which were attenuated by blocking the action of ephA2 with ephA2 siRNA or inhibitor. The production of antiviral immune mediators enhanced by rhinovirus or poly (I:C) is increased by blocking ephA2 compared with that of cells stimulated by either rhinovirus or poly(I:C) alone. In addition, blocking ephA2 attenuated RV replication in cultured cells. Taken together, these results describe a novel role of ephrinA1/ephA2 signaling in antiviral innate immune response in sinonasal epithelium, suggesting their participation in RV-induced development and exacerbations of CRS.


Subject(s)
Common Cold/metabolism , Ephrin-A1/metabolism , Epithelial Cells/metabolism , Nasal Mucosa/metabolism , Receptor, EphA2/metabolism , Rhinitis/metabolism , Rhinovirus/pathogenicity , Sinusitis/metabolism , Case-Control Studies , Cells, Cultured , Chronic Disease , Common Cold/immunology , Common Cold/virology , Cytokines/metabolism , Ephrin-A1/genetics , Ephrin-A2/genetics , Ephrin-A2/metabolism , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/virology , Host-Pathogen Interactions , Humans , Immunity, Innate , Nasal Mucosa/drug effects , Nasal Mucosa/immunology , Nasal Mucosa/virology , Poly I-C/pharmacology , Receptor, EphA2/genetics , Rhinitis/immunology , Rhinovirus/growth & development , Rhinovirus/immunology , Signal Transduction , Sinusitis/immunology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...