Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 737
Filter
1.
Methods Mol Biol ; 2813: 137-144, 2024.
Article in English | MEDLINE | ID: mdl-38888776

ABSTRACT

Air-liquid interface (ALI) airway culture models serve as a powerful tool to emulate the characteristic features of the respiratory tract in vitro. These models are particularly valuable for studying emerging respiratory viral and bacterial infections. Here, we describe an optimized protocol to obtain the ALI airway culture models using normal human bronchial epithelial cells (NHBECs). The protocol outlined below enables the generation of differentiated mucociliary airway epithelial cultures by day 28 following exposure to air.


Subject(s)
Cell Culture Techniques , Epithelial Cells , Humans , Cell Culture Techniques/methods , Epithelial Cells/microbiology , Epithelial Cells/virology , Epithelial Cells/cytology , Bronchi/cytology , Respiratory Mucosa/cytology , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Air , Cells, Cultured , Communicable Diseases/microbiology
2.
Viruses ; 16(6)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38932231

ABSTRACT

Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1, DNAH11, LRRC-23, -49, and -51, and acidic sialomucin CD164L2. PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes (RSAD2, MX1, IFIT, and ISG15) and chemokine genes (CCL5 and CXCL10), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.


Subject(s)
Betacoronavirus 1 , Epithelial Cells , Gene Expression Profiling , Interferons , Animals , Swine , Epithelial Cells/virology , Epithelial Cells/immunology , Interferons/genetics , Interferons/metabolism , Interferons/immunology , Betacoronavirus 1/immunology , Betacoronavirus 1/genetics , Immunity, Innate , Virus Replication , Coronavirus Infections/immunology , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Cytokines/metabolism , Cytokines/genetics , Cytokines/immunology , Transcriptome , Respiratory Mucosa/virology , Respiratory Mucosa/immunology , Swine Diseases/virology , Swine Diseases/immunology , Swine Diseases/genetics , Cells, Cultured , Deltacoronavirus
4.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38693120

ABSTRACT

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


Subject(s)
CD47 Antigen , Epithelial Cells , Staphylococcal Infections , Staphylococcus aureus , Superinfection , CD47 Antigen/metabolism , CD47 Antigen/genetics , Humans , Animals , Superinfection/microbiology , Mice , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/virology , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Influenza, Human/metabolism , Influenza, Human/immunology , Influenza, Human/virology , Bacterial Adhesion , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Mice, Inbred C57BL , Bronchi/metabolism , Bronchi/cytology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Mice, Knockout , Influenza A Virus, H1N1 Subtype
5.
Methods Mol Biol ; 2808: 141-152, 2024.
Article in English | MEDLINE | ID: mdl-38743368

ABSTRACT

Measles virus (MeV) infection of airway surface epithelial cells provides a site for final amplification before being released back into the environment via coughing and sneezing. Multiple cell lines have served as models of polarized epithelia for MeV infection, such as Caco2 cells (intestinal derived human epithelia) or MDCK cells (kidney derived canine epithelia). In this chapter, we describe the materials and air-liquid interface (ALI) culture conditions for maintaining four different cell lines derived from human airway epithelial cells: 16HBE14o-, Calu-3, H358, and NuLi-1. We provide methods for confirming transepithelial electrical resistance (TER) and preparing samples for microscopy as well as expected results from apical or basolateral MeV delivery. Polarized human airway derived cells serve as tissue culture models for investigating targeted questions about how MeV exits a human host. In addition, these methods are generalizable to studies of other respiratory viruses or the biology of ALI airway epithelial cells.


Subject(s)
Cell Culture Techniques , Epithelial Cells , Measles virus , Humans , Measles virus/physiology , Epithelial Cells/virology , Epithelial Cells/cytology , Cell Culture Techniques/methods , Measles/virology , Cell Line , Dogs , Animals , Respiratory Mucosa/virology , Respiratory Mucosa/cytology , Electric Impedance
6.
Cell Rep ; 43(4): 114076, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38607917

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 pandemic is characterized by the emergence of novel variants of concern (VOCs) that replace ancestral strains. Here, we dissect the complex selective pressures by evaluating variant fitness and adaptation in human respiratory tissues. We evaluate viral properties and host responses to reconstruct forces behind D614G through Omicron (BA.1) emergence. We observe differential replication in airway epithelia, differences in cellular tropism, and virus-induced cytotoxicity. D614G accumulates the most mutations after infection, supporting zoonosis and adaptation to the human airway. We perform head-to-head competitions and observe the highest fitness for Gamma and Delta. Under these conditions, RNA recombination favors variants encoding the B.1.617.1 lineage 3' end. Based on viral growth kinetics, Alpha, Gamma, and Delta exhibit increased fitness compared to D614G. In contrast, the global success of Omicron likely derives from increased transmission and antigenic variation. Our data provide molecular evidence to support epidemiological observations of VOC emergence.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/physiology , SARS-CoV-2/genetics , COVID-19/virology , COVID-19/transmission , Virus Replication , Mutation/genetics , Respiratory Mucosa/virology , Genetic Fitness , Animals , Epithelial Cells/virology , Chlorocebus aethiops , Adaptation, Physiological/genetics , Vero Cells
7.
PLoS Biol ; 22(4): e3002566, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38652717

ABSTRACT

Phage therapy is a therapeutic approach to treat multidrug-resistant (MDR) infections that employs lytic bacteriophages (phages) to eliminate bacteria. Despite the abundant evidence for its success as an antimicrobial in Eastern Europe, there is scarce data regarding its effects on the human host. Here, we aimed to understand how lytic phages interact with cells of the airway epithelium, the tissue site that is colonized by bacterial biofilms in numerous chronic respiratory disorders. Using a panel of Pseudomonas aeruginosa phages and human airway epithelial cells (AECs) derived from a person with cystic fibrosis (CF), we determined that interactions between phages and epithelial cells depend on specific phage properties as well as physiochemical features of the microenvironment. Although poor at internalizing phages, the airway epithelium responds to phage exposure by changing its transcriptional profile and secreting antiviral and proinflammatory cytokines that correlate with specific phage families. Overall, our findings indicate that mammalian responses to phages are heterogenous and could potentially alter the way that respiratory local defenses aid in bacterial clearance during phage therapy. Thus, besides phage receptor specificity in a particular bacterial isolate, the criteria to select lytic phages for therapy should be expanded to include mammalian cell responses.


Subject(s)
Cystic Fibrosis , Cytokines , Epithelial Cells , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/virology , Epithelial Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/immunology , Cytokines/metabolism , Cystic Fibrosis/therapy , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Phage Therapy , Bacteriophages/physiology , Bacteriophages/genetics , Respiratory Mucosa/virology , Respiratory Mucosa/metabolism , Respiratory Mucosa/immunology , Pseudomonas Infections/therapy , Pseudomonas Infections/immunology , Pseudomonas Phages/metabolism , Biofilms
8.
Daru ; 32(1): 215-235, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38652363

ABSTRACT

PURPOSE: Identifying the molecular mechanisms behind SARS-CoV-2 disparities and similarities will help find new treatments. The present study determines networks' shared and non-shared (specific) crucial elements in response to HCoV-229E and SARS-CoV-2 viruses to recommend candidate medications. METHODS: We retrieved the omics data on respiratory cells infected with HCoV-229E and SARS-CoV-2, constructed PPIN and GRN, and detected clusters and motifs. Using a drug-gene interaction network, we determined the similarities and disparities of mechanisms behind their host response and drug-repurposed. RESULTS: CXCL1, KLHL21, SMAD3, HIF1A, and STAT1 were the shared DEGs between both viruses' protein-protein interaction network (PPIN) and gene regulatory network (GRN). The NPM1 was a specific critical node for HCoV-229E and was a Hub-Bottleneck shared between PPI and GRN in HCoV-229E. The HLA-F, ADCY5, TRIM14, RPF1, and FGA were the seed proteins in subnetworks of the SARS-CoV-2 PPI network, and HSPA1A and RPL26 proteins were the seed in subnetworks of the PPI network of HCOV-229E. TRIM14, STAT2, and HLA-F played the same role for SARS-CoV-2. Top enriched KEGG pathways included cell cycle and proteasome in HCoV-229E and RIG-I-like receptor, Chemokine, Cytokine-cytokine, NOD-like receptor, and TNF signaling pathways in SARS-CoV-2. We suggest some candidate medications for COVID-19 patient lungs, including Noscapine, Isoetharine mesylate, Cycloserine, Ethamsylate, Cetylpyridinium, Tretinoin, Ixazomib, Vorinostat, Venetoclax, Vorinostat, Ixazomib, Venetoclax, and epoetin alfa for further in-vitro and in-vivo investigations. CONCLUSION: We suggested CXCL1, KLHL21, SMAD3, HIF1A, and STAT1, ADCY5, TRIM14, RPF1, and FGA, STAT2, and HLA-F as critical genes and Cetylpyridinium, Cycloserine, Noscapine, Ethamsylate, Epoetin alfa, Isoetharine mesylate, Ribavirin, and Tretinoin drugs to study further their importance in treating COVID-19 lung complications.


Subject(s)
Antiviral Agents , Coronavirus 229E, Human , Drug Repositioning , Protein Interaction Maps , SARS-CoV-2 , Systems Biology , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Coronavirus 229E, Human/genetics , Coronavirus 229E, Human/drug effects , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Nucleophosmin , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , Gene Regulatory Networks/drug effects , COVID-19
9.
Immunology ; 172(3): 329-342, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38354831

ABSTRACT

Alterations in airway epithelial homeostasis increase viral respiratory infections risk. Viral infections frequently are associated with chronic obstructive pulmonary disease (COPD) exacerbations, events that dramatically promote disease progression. Mechanism promoting the main respiratory viruses entry and virus-evocated innate and adaptive immune responses have now been elucidated, and an oxidative stress central role in these pathogenic processes has been recognized. Presence of reactive oxygen species in macrophages and other cells allows them to eliminate virus, but its excess alters the balance between innate and adaptive immune responses and proteases/anti-proteases and leads to uncontrolled inflammation, tissue damage, and hypercoagulability. Different upper and lower airway cell types also play a role in viral entry and infection. Carbocysteine is a muco-active drug with anti-oxidant and anti-inflammatory properties used for the management of several chronic respiratory diseases. Although the use of anti-oxidants has been proposed as an effective strategy in COPD exacerbations management, the molecular mechanisms that explain carbocysteine efficacy have not yet been fully clarified. The present review describes the most relevant features of the common respiratory virus pathophysiology with a focus on epithelial cells and oxidative stress role and reports data supporting a putative role of carbocysteine in viral respiratory infections.


Subject(s)
Carbocysteine , Oxidative Stress , Respiratory Mucosa , Respiratory Tract Infections , Virus Diseases , Humans , Carbocysteine/therapeutic use , Carbocysteine/pharmacology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Oxidative Stress/drug effects , Respiratory Mucosa/virology , Respiratory Mucosa/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/drug effects , Virus Diseases/immunology , Virus Diseases/drug therapy , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy
10.
J Virol ; 98(3): e0157623, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38323814

ABSTRACT

Adenovirus (AdV) infection of the respiratory epithelium is common but poorly understood. Human AdV species C types, such as HAdV-C5, utilize the Coxsackie-adenovirus receptor (CAR) for attachment and subsequently integrins for entry. CAR and integrins are however located deep within the tight junctions in the mucosa where they would not be easily accessible. Recently, a model for CAR-independent AdV entry was proposed. In this model, human lactoferrin (hLF), an innate immune protein, aids the viral uptake into epithelial cells by mediating interactions between the major capsid protein, hexon, and yet unknown host cellular receptor(s). However, a detailed understanding of the molecular interactions driving this mechanism is lacking. Here, we present a new cryo-EM structure of HAdV-5C hexon at high resolution alongside a hybrid structure of HAdV-5C hexon complexed with human lactoferrin (hLF). These structures reveal the molecular determinants of the interaction between hLF and HAdV-C5 hexon. hLF engages hexon primarily via its N-terminal lactoferricin (Lfcin) region, interacting with hexon's hypervariable region 1 (HVR-1). Mutational analyses pinpoint critical Lfcin contacts and also identify additional regions within hLF that critically contribute to hexon binding. Our study sheds more light on the intricate mechanism by which HAdV-C5 utilizes soluble hLF/Lfcin for cellular entry. These findings hold promise for advancing gene therapy applications and inform vaccine development. IMPORTANCE: Our study delves into the structural aspects of adenovirus (AdV) infections, specifically HAdV-C5 in the respiratory epithelium. It uncovers the molecular details of a novel pathway where human lactoferrin (hLF) interacts with the major capsid protein, hexon, facilitating viral entry, and bypassing traditional receptors such as CAR and integrins. The study's cryo-EM structures reveal how hLF engages hexon, primarily through its N-terminal lactoferricin (Lfcin) region and hexon's hypervariable region 1 (HVR-1). Mutational analyses identify critical Lfcin contacts and other regions within hLF vital for hexon binding. This structural insight sheds light on HAdV-C5's mechanism of utilizing soluble hLF/Lfcin for cellular entry, holding promise for gene therapy and vaccine development advancements in adenovirus research.


Subject(s)
Adenoviruses, Human , Capsid Proteins , Lactoferrin , Receptors, Virus , Virus Internalization , Humans , Adenovirus Infections, Human/metabolism , Adenovirus Infections, Human/virology , Adenoviruses, Human/chemistry , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Adenoviruses, Human/ultrastructure , Binding Sites/genetics , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Lactoferrin/chemistry , Lactoferrin/genetics , Lactoferrin/metabolism , Lactoferrin/ultrastructure , Models, Biological , Mutation , Protein Binding , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Receptors, Virus/ultrastructure , Solubility , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology
11.
J Virol ; 97(2): e0147822, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36656015

ABSTRACT

Little is known about the relationships between symptomatic early severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral load and upper airway mucosal gene expression and immune response. To examine the association of symptomatic SARS-CoV-2 early viral load with upper airway mucosal gene expression, we profiled the host mucosal transcriptome from nasopharyngeal swab samples from 68 adults with symptomatic, mild-to-moderate coronavirus disease 19 (COVID-19). We measured SARS-CoV-2 viral load using reverse transcription-quantitative PCR (RT-qPCR). We then examined the association of SARS-CoV-2 viral load with upper airway mucosal immune response. We detected SARS-CoV-2 in all samples and recovered >80% of the genome from 95% of the samples from symptomatic COVID-19 adults. The respiratory virome was dominated by SARS-CoV-2, with limited codetection of other respiratory viruses, with the human Rhinovirus C being identified in 4 (6%) samples. This limited codetection of other respiratory viral pathogens may be due to the implementation of public health measures, like social distancing and masking practices. We observed a significant positive correlation between SARS-CoV-2 viral load and interferon signaling (OAS2, OAS3, IFIT1, UPS18, ISG15, ISG20, IFITM1, and OASL), chemokine signaling (CXCL10 and CXCL11), and adaptive immune system (IFITM1, CD300E, and SIGLEC1) genes in symptomatic, mild-to-moderate COVID-19 adults, when adjusting for age, sex, and race. Interestingly, the expression levels of most of these genes plateaued at a cycle threshold (CT) value of ~25. Overall, our data show that the early nasal mucosal immune response to SARS-CoV-2 infection is viral load dependent, potentially modifying COVID-19 outcomes. IMPORTANCE Several prior studies have shown that SARS-CoV-2 viral load can predict the likelihood of disease spread and severity. A higher detectable SARS-CoV-2 plasma viral load was associated with worse respiratory disease severity. However, the relationship between SARS-CoV-2 viral load, airway mucosal gene expression, and immune response remains elusive. We profiled the nasal mucosal transcriptome from nasal samples collected from adults infected with SARS-CoV-2 during spring 2020 with mild-to-moderate symptoms using a comprehensive metatranscriptomics method. We observed a positive correlation between SARS-CoV-2 viral load, interferon signaling, chemokine signaling, and adaptive immune system in adults with COVID-19. Our data suggest that early nasal mucosal immune response to SARS-CoV-2 infection was viral load dependent and may modify COVID-19 outcomes.


Subject(s)
COVID-19 , Gene Expression , Respiratory Mucosa , SARS-CoV-2 , Viral Load , Adult , Humans , Chemokines/physiology , COVID-19/immunology , COVID-19/virology , Gene Expression/immunology , Immunity, Mucosal/immunology , Interferons/physiology , SARS-CoV-2/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/virology
12.
Proc Natl Acad Sci U S A ; 119(28): e2202370119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35749382

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections initiate in the bronchi of the upper respiratory tract and are able to disseminate to the lower respiratory tract, where infections can cause an acute respiratory distress syndrome with a high degree of mortality in elderly patients. We used reconstituted primary bronchial epithelia from adult and child donors to follow the SARS-CoV-2 infection dynamics. We show that, in epithelia from adult donors, infections initiate in multiciliated cells and spread within 24 to 48 h throughout the whole epithelia. Syncytia formed of ciliated and basal cells appeared at the apical side of the epithelia within 3 to 4 d and were released into the apical lumen, where they contributed to the transmittable virus dose. A small number of reconstituted epithelia were intrinsically more resistant to virus infection, limiting virus spread to different degrees. This phenotype was more frequent in epithelia derived from children versus adults and correlated with an accelerated release of type III interferon. Treatment of permissive adult epithelia with exogenous type III interferon restricted infection, while type III interferon gene knockout promoted infection. Furthermore, a transcript analysis revealed that the inflammatory response was specifically attenuated in children. Taken together, our findings suggest that apical syncytia formation is an underappreciated source of virus propagation for tissue or environmental dissemination, whereas a robust type III interferon response such as commonly seen in young donors restricted SARS-CoV-2 infection. Thus, the combination of interferon restriction and attenuated inflammatory response in children might explain the epidemiological observation of age-related susceptibility to COVID-19.


Subject(s)
Bronchi , COVID-19 , Giant Cells , Interferons , Respiratory Mucosa , SARS-CoV-2 , Aged , Bronchi/immunology , Bronchi/virology , COVID-19/immunology , COVID-19/virology , Child , Disease Susceptibility , Giant Cells/immunology , Giant Cells/virology , Humans , Interferons/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , SARS-CoV-2/immunology , Interferon Lambda
13.
mBio ; 13(4): e0105522, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35699372

ABSTRACT

Influenza A virus (IAV) causes significant morbidity and mortality in the human population. Tethered mucin 1 (MUC1) is highly expressed in airway epithelium, the primary site of IAV replication, and also by other cell types that influence IAV infection, including macrophages. MUC1 has the potential to influence infection dynamics through physical interactions and/or signaling activity, yet MUC1 modulation and its impact during viral pathogenesis remain unclear. Thus, we investigated MUC1-IAV interactions in an in vitro model of human airway epithelium (HAE). Our data indicate that a recombinant IAV hemagglutinin (H3) and H3N2 virus can bind endogenous HAE MUC1. Notably, infection of HAE with H1N1 or H3N2 IAV strains does not trigger MUC1 shedding but instead stimulates an increase in cell-associated MUC1 protein. We observed a similar increase after type I or III interferon (IFN) stimulation; however, inhibition of IFN signaling during H1N1 infection only partially abrogated this increase, indicating that multiple soluble factors contribute to MUC1 upregulation during the antiviral response. In addition to HAE, primary human monocyte-derived macrophages also upregulated MUC1 protein in response to IFN treatment and conditioned media from IAV-infected HAE. Then, to determine the impact of MUC1 on IAV pathogenesis, we developed HAE genetically depleted of MUC1 and found that MUC1 knockout cultures exhibited enhanced viral growth compared to control cultures for several IAV strains. Together, our data support a model whereby MUC1 inhibits productive uptake of IAV in HAE. Infection then stimulates MUC1 expression on multiple cell types through IFN-dependent and -independent mechanisms that further impact infection dynamics. IMPORTANCE Influenza A virus (IAV) targets airway epithelial cells for infection. Large, heavily glycosylated molecules known as tethered mucins extend from the airway epithelial cell surface and may physically restrict pathogen access to underlying cells. Additionally, tethered mucin 1 (MUC1) can be differentially phosphorylated based on external stimuli and can influence inflammation. Given MUC1's multifunctional capability, we sought to define its role during IAV infection. Here, we demonstrate that IAV directly interacts with MUC1 in a physiologically relevant model of human airway epithelium (HAE) and find that MUC1 protein expression is elevated throughout the epithelium and in primary human monocyte-derived macrophages in response to antiviral signals produced during infection. Using CRISPR/Cas9-modified HAE, we demonstrated more efficient IAV infection when MUC1 is genetically ablated. Our data suggest that MUC1 physically restricts IAV uptake and represents a dynamic component of the host response that acts to inhibit viral spread, yielding new insight into mucin-mediated antiviral defense.


Subject(s)
Influenza A virus , Influenza, Human , Mucin-1 , Antiviral Agents/pharmacology , Epithelium , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza A virus/physiology , Influenza, Human/metabolism , Interferons/pharmacology , Mucin-1/genetics , Mucin-1/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Virus Replication
14.
Viruses ; 14(2)2022 01 28.
Article in English | MEDLINE | ID: mdl-35215873

ABSTRACT

Highly pathogenic avian influenza (HPAI) outbreaks have become increasingly frequent in wild bird populations and have caused mass mortality in many wild bird species. The 2020/2021 epizootic was the largest and most deadly ever reported in Europe, and many new bird species tested positive for HPAI virus for the first time. This study investigated the tropism of HPAI virus in wild birds. We tested the pattern of virus attachment of 2020 H5N8 virus to intestinal and respiratory tissues of key bird species; and characterized pathology of naturally infected Eurasian wigeons (Mareca penelope) and barnacle geese (Branta leucopsis). This study determined that 2020 H5N8 virus had a high level of attachment to the intestinal epithelium (enterotropism) of dabbling ducks and geese and retained attachment to airway epithelium (respirotropism). Natural HPAI 2020 H5 virus infection in Eurasian wigeons and barnacle geese also showed a high level of neurotropism, as both species presented with brain lesions that co-localized with virus antigen expression. We concluded that the combination of respirotropism, neurotropism, and possibly enterotropism, contributed to the successful adaptation of 2020/2021 HPAI H5 viruses to wild waterbird populations.


Subject(s)
Ducks/virology , Geese/virology , Influenza A Virus, H5N8 Subtype/pathogenicity , Influenza in Birds/virology , Viral Tropism , Animals , Animals, Wild/virology , Brain/virology , Host Adaptation , Influenza A Virus, H5N8 Subtype/isolation & purification , Influenza A Virus, H5N8 Subtype/physiology , Intestinal Mucosa/virology , RNA, Viral/analysis , Respiratory Mucosa/virology , Virus Attachment
15.
Viruses ; 14(2)2022 02 05.
Article in English | MEDLINE | ID: mdl-35215919

ABSTRACT

SARS-CoV-2 can efficiently infect both children and adults, albeit with morbidity and mortality positively associated with increasing host age and presence of co-morbidities. SARS-CoV-2 continues to adapt to the human population, resulting in several variants of concern (VOC) with novel properties, such as Alpha and Delta. However, factors driving SARS-CoV-2 fitness and evolution in paediatric cohorts remain poorly explored. Here, we provide evidence that both viral and host factors co-operate to shape SARS-CoV-2 genotypic and phenotypic change in primary airway cell cultures derived from children. Through viral whole-genome sequencing, we explored changes in genetic diversity over time of two pre-VOC clinical isolates of SARS-CoV-2 during passage in paediatric well-differentiated primary nasal epithelial cell (WD-PNEC) cultures and in parallel, in unmodified Vero-derived cell lines. We identified a consistent, rich genetic diversity arising in vitro, variants of which could rapidly rise to near fixation within two passages. Within isolates, SARS-CoV-2 evolution was dependent on host cells, with paediatric WD-PNECs showing a reduced diversity compared to Vero (E6) cells. However, mutations were not shared between strains. Furthermore, comparison of both Vero-grown isolates on WD-PNECs disclosed marked growth attenuation mapping to the loss of the polybasic cleavage site (PBCS) in Spike, while the strain with mutations in Nsp12 (T293I), Spike (P812R) and a truncation of Orf7a remained viable in WD-PNECs. Altogether, our work demonstrates that pre-VOC SARS-CoV-2 efficiently infects paediatric respiratory epithelial cells, and its evolution is restrained compared to Vero (E6) cells, similar to the case of adult cells. We highlight the significant genetic plasticity of SARS-CoV-2 while uncovering an influential role for collaboration between viral and host cell factors in shaping viral evolution and ultimately fitness in human respiratory epithelium.


Subject(s)
Evolution, Molecular , Respiratory Mucosa/virology , SARS-CoV-2/genetics , Animals , Cells, Cultured , Child , Chlorocebus aethiops , Genotype , Humans , Mutation , Nose/cytology , Nose/virology , Phenotype , SARS-CoV-2/classification , SARS-CoV-2/growth & development , Vero Cells , Whole Genome Sequencing
16.
Life Sci Alliance ; 5(4)2022 04.
Article in English | MEDLINE | ID: mdl-35110354

ABSTRACT

BACKGROUND: There are limited effective prophylactic/early treatments for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Viral entry requires spike protein binding to the angiotensin-converting enzyme-2 receptor and cleavage by transmembrane serine protease 2 (TMPRSS2), a cell surface serine protease. Targeting of TMPRSS2 by either androgen blockade or direct inhibition is in clinical trials in early SARS-CoV-2 infection. METHODS: We used differentiated primary human airway epithelial cells at the air-liquid interface to test the impact of targeting TMPRSS2 on the prevention of SARS-CoV-2 infection. RESULTS: We first modelled the systemic delivery of compounds. Enzalutamide, an oral androgen receptor antagonist, had no impact on SARS-CoV-2 infection. By contrast, camostat mesylate, an orally available serine protease inhibitor, blocked SARS-CoV-2 entry. However, oral camostat is rapidly metabolised in the circulation, with poor airway bioavailability. We therefore modelled local airway administration by applying camostat to the apical surface of differentiated airway cultures. We demonstrated that a brief exposure to topical camostat effectively restricts SARS-CoV-2 infection. CONCLUSION: These experiments demonstrate a potential therapeutic role for topical camostat for pre- or post-exposure prophylaxis of SARS-CoV-2, which can now be evaluated in a clinical trial.


Subject(s)
Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/administration & dosage , Administration, Topical , Androgens/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Antiviral Agents/pharmacology , COVID-19/prevention & control , COVID-19/virology , Cells, Cultured , Epithelial Cells , Esters/pharmacology , Gene Expression , Goblet Cells/immunology , Goblet Cells/metabolism , Guanidines/pharmacology , Host-Pathogen Interactions/drug effects , Humans , Serine Endopeptidases/genetics , Signal Transduction , Virus Internalization/drug effects , Virus Replication/drug effects
17.
Sci Rep ; 12(1): 2966, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194053

ABSTRACT

With highly active anti-retroviral therapy (HAART), higher incidence of airway abnormalities is common in the HIV population consistent with the concept of accelerated lung "aging". Our previous findings demonstrated that HIV induces human airway basal cells (BC) into destructive and inflammatory phenotypes. Since BC function as stem/progenitor cells of the small airway epithelium (SAE), responsible for self-renewal and differentiation of SAE, we hypothesized that BC from people living with HIV (PLWH) may have altered differentiation capacity that contribute to premature aging. The data demonstrates that BC from PLWH have impaired capacity to differentiate in vitro and senescent phenotypes including shortened telomeres, increased expression of ß-galactosidase and cell cycle inhibitors, and mitochondrial dysfunction. In vitro studies demonstrated that BC senescence is partly due to adverse effects of HAART on BC. These findings provide an explanation for higher incidence of airway dysfunction and accelerated lung aging observed in PLWH.


Subject(s)
Cell Differentiation , HIV Infections/metabolism , HIV-1/metabolism , Lung/metabolism , Respiratory Mucosa/metabolism , Stem Cells/metabolism , Adult , Female , Humans , Lung/virology , Male , Middle Aged , Respiratory Mucosa/virology , Stem Cells/virology , Telomere Shortening
18.
Nat Commun ; 13(1): 719, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169114

ABSTRACT

There is an urgent need for potent and selective antivirals against SARS-CoV-2. Pfizer developed PF-07321332 (PF-332), a potent inhibitor of the viral main protease (Mpro, 3CLpro) that can be dosed orally and that is in clinical development. We here report that PF-332 exerts equipotent in vitro activity against the four SARS-CoV-2 variants of concerns (VoC) and that it can completely arrest replication of the alpha variant in primary human airway epithelial cells grown at the air-liquid interface. Treatment of Syrian Golden hamsters with PF-332 (250 mg/kg, twice daily) completely protected the animals against intranasal infection with the beta (B.1.351) and delta (B.1.617.2) SARS-CoV-2 variants. Moreover, treatment of SARS-CoV-2 (B.1.617.2) infected animals with PF-332 completely prevented transmission to untreated co-housed sentinels.


Subject(s)
COVID-19 Drug Treatment , Disease Models, Animal , Lactams/administration & dosage , Leucine/administration & dosage , Nitriles/administration & dosage , Proline/administration & dosage , SARS-CoV-2/drug effects , Viral Protease Inhibitors/administration & dosage , A549 Cells , Administration, Oral , Animals , COVID-19/prevention & control , COVID-19/transmission , COVID-19/virology , Chlorocebus aethiops , Coronavirus 3C Proteases/antagonists & inhibitors , Cricetinae , Humans , Lactams/pharmacokinetics , Leucine/pharmacokinetics , Mesocricetus , Nitriles/pharmacokinetics , Proline/pharmacokinetics , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , SARS-CoV-2/enzymology , SARS-CoV-2/physiology , Vero Cells , Viral Protease Inhibitors/pharmacokinetics , Virus Replication/drug effects
19.
Viruses ; 14(1)2022 01 14.
Article in English | MEDLINE | ID: mdl-35062352

ABSTRACT

Respiratory disease in horses is caused by a multifactorial complex of infectious agents and environmental factors. An important pathogen in horses is equine herpesvirus type 1 (EHV-1). During co-evolution with this ancient alphaherpesvirus, the horse's respiratory tract has developed multiple antiviral barriers. However, these barriers can become compromised by environmental threats. Pollens and mycotoxins enhance mucosal susceptibility to EHV-1 by interrupting cell junctions, allowing the virus to reach its basolateral receptor. Whether bacterial toxins also play a role in this impairment has not been studied yet. Here, we evaluated the role of α-hemolysin (Hla) and adenylate cyclase (ACT), toxins derived from the facultative pathogenic bacterium Staphylococcus aureus (S. aureus) and the primary pathogen Bordetella bronchiseptica (B. bronchiseptica), respectively. Equine respiratory mucosal explants were cultured at an air-liquid interface and pretreated with these toxins, prior to EHV-1 inoculation. Morphological analysis of hematoxylin-eosin (HE)-stained sections of the explants revealed a decreased epithelial thickness upon treatment with both toxins. Additionally, the Hla toxin induced detachment of epithelial cells and a partial loss of cilia. These morphological changes were correlated with increased EHV-1 replication in the epithelium, as assessed by immunofluorescent stainings and confocal microscopy. In view of these results, we argue that the ACT and Hla toxins increase the susceptibility of the epithelium to EHV-1 by disrupting the epithelial barrier function. In conclusion, this study is the first to report that bacterial exotoxins increase the horse's sensitivity to EHV-1 infection. Therefore, we propose that horses suffering from infection by S. aureus or B. bronchiseptica may be more susceptible to EHV-1 infection.


Subject(s)
Bacterial Toxins/pharmacology , Bordetella bronchiseptica/metabolism , Herpesviridae Infections/drug therapy , Herpesviridae Infections/veterinary , Herpesvirus 1, Equid/drug effects , Horse Diseases/virology , Respiratory Tract Diseases/virology , Staphylococcus aureus/metabolism , Animals , Epithelial Cells/virology , Hemolysin Proteins , Horses , Respiratory Mucosa/drug effects , Respiratory Mucosa/virology , Virus Replication/drug effects
20.
Medicina (Kaunas) ; 58(1)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35056429

ABSTRACT

COPD is a chronic lung disorder characterized by a progressive and irreversible airflow obstruction, and persistent pulmonary inflammation. It has become a global epidemic affecting 10% of the population, and is the third leading cause of death worldwide. Respiratory viruses are a primary cause of COPD exacerbations, often leading to secondary bacterial infections in the lower respiratory tract. COPD patients are more susceptible to viral infections and associated severe disease, leading to accelerated lung function deterioration, hospitalization, and an increased risk of mortality. The airway epithelium plays an essential role in maintaining immune homeostasis, and orchestrates the innate and adaptive responses of the lung against inhaled and pathogen insults. A healthy airway epithelium acts as the first line of host defense by maintaining barrier integrity and the mucociliary escalator, secreting an array of inflammatory mediators, and initiating an antiviral state through the interferon (IFN) response. The airway epithelium is a major site of viral infection, and the interaction between respiratory viruses and airway epithelial cells activates host defense mechanisms, resulting in rapid virus clearance. As such, the production of IFNs and the activation of IFN signaling cascades directly contributes to host defense against viral infections and subsequent innate and adaptive immunity. However, the COPD airway epithelium exhibits an altered antiviral response, leading to enhanced susceptibility to severe disease and impaired IFN signaling. Despite decades of research, there is no effective antiviral therapy for COPD patients. Herein, we review current insights into understanding the mechanisms of viral evasion and host IFN antiviral defense signaling impairment in COPD airway epithelium. Understanding how antiviral mechanisms operate in COPD exacerbations will facilitate the discovery of potential therapeutic interventions to reduce COPD hospitalization and disease severity.


Subject(s)
Interferons/immunology , Pulmonary Disease, Chronic Obstructive , Respiratory Mucosa/immunology , Viruses , Epithelium , Humans , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/virology , Respiratory Mucosa/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...