Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; : e0136123, 2023 Sep 19.
Article in English | MEDLINE | ID: mdl-37724882

ABSTRACT

Amplification of measles virus (MeV) in human airway epithelia may contribute to its extremely high contagious nature. We use well-differentiated primary cultures of human airway epithelial cells (HAE) to model ex vivo how MeV spreads in human airways. In HAE, MeV spreads cell-to-cell for 3-5 days, but then, infectious center growth is arrested. What stops MeV spread in HAE is not understood, but interferon (IFN) is known to slow MeV spread in other in vitro and in vivo models. Here, we assessed the role of type I and type III IFN in arresting MeV spread in HAE. The addition of IFN-ß or IFN-λ1 to the medium of infected HAE slowed MeV infectious center growth, but when IFN receptor signaling was blocked, infectious center size was not affected. In contrast, blocking type-I IFN receptor signaling enhanced respiratory syncytial virus spread. HAE were also infected with MeV mutants defective for the V protein. The V protein has been demonstrated to interact with both MDA5 and STAT2 to inhibit activation of innate immunity; however, innate immune reactions were unexpectedly muted against the V-defective MeV in HAE. Minimal innate immunity activation was confirmed by deep sequencing, quantitative RT-PCR, and single-cell RNA-seq analyses of the transcription of IFN and IFN-stimulated genes. We conclude that in HAE, IFN-signaling can contribute to slowing infectious center growth; however, IFN-independent processes are most important for limiting cell-to-cell spread. IMPORTANCE Fundamental biological questions remain about the highly contagious measles virus (MeV). MeV amplifies within airway epithelial cells before spreading to the next host. This final step likely contributes to the ability of MeV to spread host-to-host. Over the course of 3-5 days post-infection of airway epithelial cells, MeV spreads directly cell-to-cell and forms infectious centers. Infectious center formation is unique to MeV. In this study, we show that interferon (IFN) signaling does not explain why MeV cell-to-cell spread is ultimately impeded within the cell layer. The ability of MeV to spread cell-to-cell in airway cells without appreciable IFN induction may contribute to its highly contagious nature. This study contributes to the understanding of a significant global health concern by demonstrating that infectious center formation occurs independent of the simplest explanation for limiting viral transmission within a host.

2.
J Infect Dis ; 2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37698016

ABSTRACT

BACKGROUND: Chronic pulmonary conditions such as asthma and COPD increase the risk of morbidity and mortality during infection with the Middle East respiratory syndrome coronavirus (MERS-CoV). We hypothesized that individuals with such comorbidities are more susceptible to MERS-CoV infection due to increased expression of its receptor, dipeptidyl peptidase 4 (DPP4). METHODS: We modeled chronic airway disease by treating primary human airway epithelia with the Th2 cytokine IL-13, examining how this impacted DPP4 protein levels along with MERS-CoV entry and replication. RESULTS: IL-13 exposure for 3 days led to increased DPP4 protein abundance, while a 21-day treatment increased DPP4 levels and caused goblet cell metaplasia. Surprisingly, despite this increase in receptor availability, MERS-CoV entry and replication were not significantly impacted by IL-13 treatment. CONCLUSIONS: Our results suggest that increased DPP4 abundance is likely not the primary mechanism leading to increased MERS severity in the setting of Th2 inflammation. Transcriptional profiling analysis highlighted the complexity of IL-13 induced changes in airway epithelia, including altered expression of genes involved in innate immunity, antiviral responses, and maintenance of the extracellular mucus barrier. These data suggest that additional factors likely interact with DPP4 abundance to determine MERS-CoV infection outcomes.

3.
mBio ; 12(4): e0097021, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34340553

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has caused significant morbidity and mortality on a global scale. The etiologic agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), initiates host cell entry when its spike protein (S) binds to its receptor, angiotensin-converting enzyme 2 (ACE2). In airway epithelia, the spike protein is cleaved by the cell surface protease TMPRSS2, facilitating membrane fusion and entry at the cell surface. This dependence on TMPRSS2 and related proteases suggests that protease inhibitors might limit SARS-CoV-2 infection in the respiratory tract. Here, we tested two serine protease inhibitors, camostat mesylate and nafamostat mesylate, for their ability to inhibit entry of SARS-CoV-2 and that of a second pathogenic coronavirus, Middle East respiratory syndrome coronavirus (MERS-CoV). Both camostat and nafamostat reduced infection in primary human airway epithelia and in the Calu-3 2B4 cell line, with nafamostat exhibiting greater potency. We then assessed whether nafamostat was protective against SARS-CoV-2 in vivo using two mouse models. In mice sensitized to SARS-CoV-2 infection by transduction with human ACE2, intranasal nafamostat treatment prior to or shortly after SARS-CoV-2 infection significantly reduced weight loss and lung tissue titers. Similarly, prophylactic intranasal treatment with nafamostat reduced weight loss, viral burden, and mortality in K18-hACE2 transgenic mice. These findings establish nafamostat as a candidate for the prevention or treatment of SARS-CoV-2 infection and disease pathogenesis. IMPORTANCE The causative agent of COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), requires host cell surface proteases for membrane fusion and entry into airway epithelia. We tested the hypothesis that inhibitors of these proteases, the serine protease inhibitors camostat and nafamostat, block infection by SARS-CoV-2. We found that both camostat and nafamostat reduce infection in human airway epithelia, with nafamostat showing greater potency. We then asked whether nafamostat protects mice against SARS-CoV-2 infection and subsequent COVID-19 lung disease. We performed infections in mice made susceptible to SARS-CoV-2 infection by introducing the human version of ACE2, the SARS-CoV-2 receptor, into their airway epithelia. We observed that pretreating these mice with nafamostat prior to SARS-CoV-2 infection resulted in better outcomes, in the form of less virus-induced weight loss, viral replication, and mortality than that observed in the untreated control mice. These results provide preclinical evidence for the efficacy of nafamostat in treating and/or preventing COVID-19.


Subject(s)
Benzamidines/pharmacology , Esters/pharmacology , Guanidines/pharmacology , SARS-CoV-2/drug effects , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Virus Internalization/drug effects , Angiotensin-Converting Enzyme 2/genetics , Animals , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle East Respiratory Syndrome Coronavirus/drug effects , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment
4.
J Infect Dis ; 224(8): 1357-1361, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34289058

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 ) initiates entry into airway epithelia by binding its receptor, angiotensin-converting enzyme 2 (ACE2). METHODS: To explore whether interindividual variation in ACE2 abundance contributes to variability in coronavirus disease 2019 (COVID-19) outcomes, we measured ACE2 protein abundance in primary airway epithelial cultures derived from 58 human donor lungs. RESULTS: We found no evidence for sex- or age-dependent differences in ACE2 protein expression. Furthermore, we found that variations in ACE2 abundance had minimal effects on viral replication and induction of the interferon response in airway epithelia infected with SARS-CoV-2. CONCLUSIONS: Our results highlight the relative importance of additional host factors, beyond viral receptor expression, in determining COVID-19 lung disease outcomes.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/pathology , Receptors, Coronavirus/metabolism , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/analysis , Biological Variation, Population , Bronchi/cytology , Bronchi/pathology , Bronchi/virology , COVID-19/virology , Epithelial Cells , Female , Humans , Male , Primary Cell Culture , Receptors, Coronavirus/analysis , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Sex Factors , Virus Internalization
5.
Front Public Health ; 9: 756049, 2021.
Article in English | MEDLINE | ID: mdl-35059374

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) causes respiratory infection in humans, with symptom severity that ranges from asymptomatic to severe pneumonia. Known risk factors for severe MERS include male sex, older age, and the presence of various comorbidities. MERS-CoV gains entry into cells by binding its receptor, dipeptidyl peptidase 4 (DPP4), on the surface of airway epithelia. We hypothesized that expression of this receptor might be an additional determinant of outcomes in different individuals during MERS-CoV infection. To learn more about the role of DPP4 in facilitating MERS-CoV infection and spread, we used ELISA and immunofluorescent staining to characterize DPP4 expression in well-differentiated primary human airway epithelia (HAE). We noted wide inter-individual variation in DPP4 abundance, varying by as much as 1000-fold between HAE donors. This variability appears to influence multiple aspects of MERS-CoV infection and pathogenesis, with greater DPP4 abundance correlating with early, robust virus replication and increased cell sloughing. We also observed increased induction of interferon and some interferon-stimulated genes in response to MERS-CoV infection in epithelia with the greatest DPP4 abundance. Overall, our results indicate that inter-individual differences in DPP4 abundance are one host factor contributing to MERS-CoV replication and host defense responses, and highlight how HAE may serve as a useful model for identifying risk factors associated with heightened susceptibility to serious respiratory pathogens.


Subject(s)
Middle East Respiratory Syndrome Coronavirus , Respiratory Tract Infections , Aged , Humans , Immunity , Male , Middle East Respiratory Syndrome Coronavirus/genetics , Virus Replication
6.
Am J Respir Cell Mol Biol ; 62(1): 104-111, 2020 01.
Article in English | MEDLINE | ID: mdl-31242392

ABSTRACT

Although chronic bacterial infections and inflammation are associated with progressive lung disease in patients with cystic fibrosis (CF), much less is known regarding the contributions of respiratory viral infections to this process. Clinical studies suggest that antiviral host defenses may be compromised in individuals with CF, and CF airway epithelia exhibit impaired antiviral responses in vitro. Here, we used the CF pig model to test the hypothesis that the antiviral activity of respiratory secretions is reduced in CF. We developed an in vitro assay to measure the innate antiviral activity present in airway surface liquid (ASL) from CF and non-CF pigs. We found that tracheal and nasal ASL from newborn non-CF pigs exhibited dose-dependent inhibitory activity against several enveloped and encapsidated viruses, including Sendai virus, respiratory syncytial virus, influenza A, and adenovirus. Importantly, we found that the anti-Sendai virus activity of nasal ASL from newborn CF pigs was significantly diminished relative to non-CF littermate controls. This diminution of extracellular antiviral defenses appears to be driven, at least in part, by the differences in pH between CF and non-CF ASL. These data highlight the novel antiviral properties of native airway secretions and suggest the possibility that defects in extracellular antiviral defenses contribute to CF pathogenesis.


Subject(s)
Antiviral Agents/immunology , Body Fluids/immunology , Cystic Fibrosis/immunology , Immunity, Innate/immunology , Lung/immunology , Animals , Body Fluids/virology , Cystic Fibrosis/virology , Hydrogen-Ion Concentration , Lung/virology , Respiratory Mucosa/immunology , Respiratory Mucosa/virology , Swine , Trachea/immunology , Trachea/virology , Virus Diseases/immunology , Virus Diseases/virology , Viruses/immunology
7.
Am J Respir Crit Care Med ; 194(7): 845-854, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27027566

ABSTRACT

RATIONALE: Studies suggest that inappropriate responses to proinflammatory stimuli might contribute to inflammation in cystic fibrosis (CF) lungs. However, technical challenges have made it difficult to distinguish whether altered responses in CF airways are an intrinsic defect or a secondary effect of chronic disease in their tissue of origin. The CF pig model provides an opportunity to study the inflammatory responses of CF airways at birth, before the onset of infection and inflammation. OBJECTIVES: To test the hypothesis that acute inflammatory responses are perturbed in porcine CF airways. METHODS: We investigated the inflammatory responses of newborn CF and non-CF pig airways following a 4-hour exposure to heat-killed Staphylococcus aureus, in vivo and in vitro. MEASUREMENTS AND MAIN RESULTS: Following an in vivo S. aureus challenge, markers of inflammation were similar between CF and littermate control animals through evaluation of bronchoalveolar lavage and tissues. However, transcriptome analysis revealed genotype-dependent differences as CF pigs showed a diminished host defense response compared with their non-CF counterparts. Furthermore, CF pig airways exhibited an increase in apoptotic pathways and a suppression of ciliary and flagellar biosynthetic pathways. Similar differences were observed in cultured airway epithelia from CF and non-CF pigs exposed to the stimulus. CONCLUSIONS: Transcriptome profiling suggests that acute inflammatory responses are dysregulated in the airways of newborn CF pigs.


Subject(s)
Cystic Fibrosis/immunology , Lung/immunology , Staphylococcus aureus/immunology , Animals , Animals, Newborn , Apoptosis/genetics , Cell Proliferation/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Disease Progression , Epithelium/immunology , Genotype , In Vitro Techniques , Inflammation/genetics , Inflammation/immunology , Models, Animal , Respiratory Mucosa/immunology , Signal Transduction/genetics , Swine , Transcriptome/genetics
8.
Dis Model Mech ; 8(5): 501-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25765466

ABSTRACT

Otitis media (inflammation of the middle ear) is one of the most common diseases of early childhood. Susceptibility to otitis is influenced by a number of factors, including the actions of innate immune molecules secreted by the epithelia lining the nasopharynx, middle ear and Eustachian tube. The SPLUNC1 (short palate, lung, nasal epithelial clone 1) protein is a highly abundant secretory product of the mammalian nasal, oral and respiratory mucosa that is thought to play a multifunctional role in host defense. In this study we investigated Splunc1 expression in the ear of the mouse, and examined whether this protein contributes to overall host defense in the middle ear and/or Eustachian tube. We found that Splunc1 is highly expressed in both the surface epithelium and in submucosal glands in these regions in wild-type mice. In mice lacking Splunc1, we noted histologically an increased frequency of otitis media, characterized by the accumulation of leukocytes (neutrophils with scattered macrophages), proteinaceous fluid and mucus in the middle ear lumens. Furthermore, many of these mice had extensive remodeling of the middle ear wall, suggesting a chronic course of disease. From these observations, we conclude that loss of Splunc1 predisposes mice to the development of otitis media. The Splunc1(-/-) mouse model should help investigators to better understand both the biological role of Splunc1 as well as host defense mechanisms in the middle ear.


Subject(s)
Glycoproteins/deficiency , Otitis Media/pathology , Phosphoproteins/deficiency , Animals , Bacteria/metabolism , Disease Models, Animal , Disease Susceptibility , Ear, Middle/metabolism , Ear, Middle/microbiology , Ear, Middle/pathology , Eustachian Tube/pathology , Fungi/physiology , Glycoproteins/metabolism , Mice, Inbred C3H , Penetrance , Phosphoproteins/metabolism
9.
Am J Respir Crit Care Med ; 190(2): 165-74, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24940638

ABSTRACT

RATIONALE: Cathepsin S (CTSS) activity is increased in bronchoalveolar lavage (BAL) fluid from patients with cystic fibrosis (CF). This activity contributes to lung inflammation via degradation of antimicrobial proteins, such as lactoferrin and members of the ß-defensin family. OBJECTIVES: In this study, we investigated the hypothesis that airway epithelial cells are a source of CTSS, and mechanisms underlying CTSS expression in the CF lung. METHODS: Protease activity was determined using fluorogenic activity assays. Protein and mRNA expression were analyzed by ELISA, Western blotting, and reverse-transcriptase polymerase chain reaction. MEASUREMENTS AND MAIN RESULTS: In contrast to neutrophil elastase, CTSS activity was detectable in 100% of CF BAL fluid samples from patients without Pseudomonas aeruginosa infection. In this study, we identified epithelial cells as a source of pulmonary CTSS activity with the demonstration that CF airway epithelial cells express and secrete significantly more CTSS than non-CF control cells in the absence of proinflammatory stimulation. Furthermore, levels of the transcription factor IRF-1 correlated with increased levels of its target gene CTSS. We discovered that miR-31, which is decreased in the CF airways, regulates IRF-1 in CF epithelial cells. Treating CF bronchial epithelial cells with a miR-31 mimic decreased IRF-1 protein levels with concomitant knockdown of CTSS expression and secretion. CONCLUSIONS: The miR-31/IRF-1/CTSS pathway may play a functional role in the pathogenesis of CF lung disease and may open up new avenues for exploration in the search for an effective therapeutic target.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Cathepsins/metabolism , Cystic Fibrosis/genetics , Interferon Regulatory Factor-1/metabolism , MicroRNAs/metabolism , Respiratory Mucosa/metabolism , Adolescent , Biomarkers/metabolism , Blotting, Western , Bronchoalveolar Lavage Fluid/microbiology , Cell Line , Child , Child, Preschool , Cystic Fibrosis/metabolism , Cystic Fibrosis/microbiology , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , Infant , Peptide Hydrolases/metabolism , Pseudomonas Infections/complications , Pseudomonas Infections/diagnosis , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/isolation & purification , Respiratory Mucosa/microbiology , Reverse Transcriptase Polymerase Chain Reaction
10.
Am J Physiol Lung Cell Mol Physiol ; 305(3): L256-66, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23709621

ABSTRACT

The airway mucosa and the alveolar surface form dynamic interfaces between the lung and the external environment. The epithelial cells lining these barriers elaborate a thin liquid layer containing secreted peptides and proteins that contribute to host defense and other functions. The goal of this study was to develop and apply methods to define the proteome of porcine lung lining liquid, in part, by leveraging the wealth of information in the Sus scrofa database of Ensembl gene, transcript, and protein model predictions. We developed an optimized workflow for detection of secreted proteins in porcine bronchoalveolar lavage (BAL) fluid and in methacholine-induced tracheal secretions [airway surface liquid (ASL)]. We detected 674 and 3,858 unique porcine-specific proteins in BAL and ASL, respectively. This proteome was composed of proteins representing a diverse range of molecular classes and biological processes, including host defense, molecular transport, cell communication, cytoskeletal, and metabolic functions. Specifically, we detected a significant number of secreted proteins with known or predicted roles in innate and adaptive immunity, microbial killing, or other aspects of host defense. In greatly expanding the known proteome of the lung lining fluid in the pig, this study provides a valuable resource for future studies using this important animal model of pulmonary physiology and disease.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Proteome/analysis , Respiratory Mucosa/chemistry , Animals , Animals, Newborn , Body Fluids , Databases, Protein , Epithelial Cells/chemistry , Epithelial Cells/cytology , Lung/metabolism , Mass Spectrometry , Methacholine Chloride , Respiratory Mucosa/cytology , Sus scrofa , Trachea/metabolism
11.
Am J Pathol ; 182(5): 1519-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23499554

ABSTRACT

Epithelial host defense proteins comprise a critical component of the pulmonary innate immune response to infection. The short palate, lung, nasal epithelium clone (PLUNC) 1 (SPLUNC1) protein is a member of the bactericidal/permeability-increasing (BPI) fold-containing (BPIF) protein family, sharing structural similarities with BPI-like proteins. SPLUNC1 is a 25 kDa secretory protein that is expressed in nasal, oropharyngeal, and lung epithelia, and has been implicated in airway host defense against Pseudomonas aeruginosa and other organisms. SPLUNC1 is reported to have surfactant properties, which may contribute to anti-biofilm defenses. The objective of this study was to assess the importance of SPLUNC1 surfactant activity in airway epithelial secretions and to explore its biological relevance in the context of a bacterial infection model. Using cultured airway epithelia, we confirmed that SPLUNC1 is critically important for maintenance of low surface tension in airway fluids. Furthermore, we demonstrated that recombinant SPLUNC1 (rSPLUNC1) significantly inhibited Klebsiella pneumoniae biofilm formation on airway epithelia. We subsequently found that Splunc1(-/-) mice were significantly more susceptible to infection with K. pneumoniae, confirming the likely in vivo relevance of this anti-biofilm effect. Our data indicate that SPLUNC1 is a crucial component of mucosal innate immune defense against pulmonary infection by a relevant airway pathogen, and provide further support for the novel hypothesis that SPLUNC1 protein prevents bacterial biofilm formation through its ability to modulate surface tension of airway fluids.


Subject(s)
Glycoproteins/metabolism , Host-Pathogen Interactions/immunology , Klebsiella Infections/immunology , Klebsiella pneumoniae/physiology , Lung/pathology , Phosphoproteins/metabolism , Respiratory Tract Infections/immunology , Animals , Base Sequence , Biofilms/growth & development , Cytokines/biosynthesis , Disease Susceptibility/immunology , Disease Susceptibility/microbiology , Disease Susceptibility/pathology , Disease Susceptibility/physiopathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glycoproteins/deficiency , Glycoproteins/genetics , Humans , Inflammation Mediators/metabolism , Klebsiella Infections/microbiology , Klebsiella Infections/pathology , Klebsiella Infections/physiopathology , Klebsiella pneumoniae/growth & development , Lung/immunology , Lung/microbiology , Lung/physiopathology , Mice , Molecular Sequence Data , Phosphoproteins/deficiency , Phosphoproteins/genetics , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/physiopathology , Surface Tension , Up-Regulation
12.
Arch Oral Biol ; 57(2): 197-204, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21925642

ABSTRACT

The short palate lung and nasal epithelial clone 1 (SPLUNC1) protein may be differentially expressed in oral infections, oral inflammatory disorders, or oral malignancies and may be involved in innate immune responses in the oral cavity. However, the actual concentration of SPLUNC1 in saliva has not previously been determined. In this study, we determined the concentrations of SPLUNC1 in saliva using a particle-based antibody capture and detection immunoassay. A commercial goat anti-rhSPLUNC1 polyclonal antibody (AF1897) was linked to fluorescent polystyrene microspheres and used as the capture antibody. A commercial mouse IgG2b anti-rhSPLUNC1 monoclonal antibody (MAB1897) was biotinylated and used as the detection antibody. Western blot and 2-dimensional fluorescence difference gel electrophoresis (2-D DIGE) analysis of immunoprecipitated rhSPLUNC1 and SPLUNC1 from saliva were used to show that the capture AF1897 and detection MAB1897 antibodies both recognized SPLUNC1. Protein concentrations in saliva from 20 subjects ranged from 0.9 to 23.9mg/ml; SPLUNC1 concentrations ranged from 34.7ng/ml to 13.8µg/ml; and SPLUNC concentrations normalized per mg of total salivary protein ranged from 4.7ng/ml to 5.3µg/ml. These results show that SPLUNC1 is detected in saliva in a variety of concentrations. This immunoassay may prove to be useful in determining the concentration of SPLUNC1 in saliva for assessing its role in the pathogenesis of oral infections, oral inflammatory disorders, or oral malignancies.


Subject(s)
Epithelial Cells/immunology , Gingival Crevicular Fluid/immunology , Glycoproteins/immunology , Immunoassay/methods , Mouth/immunology , Phosphoproteins/immunology , Respiratory System/immunology , Saliva/immunology , Adult , Blotting, Western , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunologic Techniques , Male , Middle Aged , Saliva/metabolism
13.
Biochem Soc Trans ; 39(4): 1012-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21787339

ABSTRACT

PLUNC (palate, lung and nasal epithelium clone) protein is an abundant secretory product of epithelia throughout the mammalian conducting airways. Despite its homology with the innate immune defence molecules BPI (bactericidal/permeability-increasing protein) and LBP (lipopolysaccharide-binding protein), it has been difficult to define the functions of PLUNC. Based on its marked hydrophobicity and expression pattern, we hypothesized that PLUNC is an airway surfactant. We found that purified recombinant human PLUNC exhibited potent surfactant activity by several different measures, and experiments with airway epithelial cell lines and primary cultures indicate that native PLUNC makes a significant contribution to the overall surface tension in airway epithelial secretions. Interestingly, we also found that physiologically relevant concentrations of PLUNC-inhibited Pseudomonas aeruginosa biofilm formation in vitro without acting directly as a bactericide. This finding suggests that PLUNC protein may inhibit biofilm formation by airway pathogens, perhaps through its dispersant properties. Our data, along with reports from other groups on activity against some airway pathogens, expand on an emerging picture of PLUNC as a multifunctional protein, which plays a novel role in airway defences at the air/liquid interface.


Subject(s)
Glycoproteins/metabolism , Phosphoproteins/metabolism , Pulmonary Surfactants/metabolism , Respiratory System/metabolism , Animals , Bacterial Infections/immunology , Biofilms , Humans , Hydrophobic and Hydrophilic Interactions , Immunity, Innate
14.
Sci Transl Med ; 2(29): 29ra31, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20427821

ABSTRACT

Lung disease causes most of the morbidity and mortality in cystic fibrosis (CF). Understanding the pathogenesis of this disease has been hindered, however, by the lack of an animal model with characteristic features of CF. To overcome this problem, we recently generated pigs with mutated CFTR genes. We now report that, within months of birth, CF pigs spontaneously developed hallmark features of CF lung disease, including airway inflammation, remodeling, mucus accumulation, and infection. Their lungs contained multiple bacterial species, suggesting that the lungs of CF pigs have a host defense defect against a wide spectrum of bacteria. In humans, the temporal and causal relations between inflammation and infection have remained uncertain. To investigate these processes, we studied newborn pigs. Their lungs showed no inflammation but were less often sterile than controls. Moreover, after introduction of bacteria into their lungs, pigs with CF failed to eradicate bacteria as effectively as wild-type pigs. These results suggest that impaired bacterial elimination is the pathogenic event that initiates a cascade of inflammation and pathology in CF lungs. Our finding that pigs with CF have a host defense defect against bacteria within hours of birth provides an opportunity to further investigate CF pathogenesis and to test therapeutic and preventive strategies that could be deployed before secondary consequences develop.


Subject(s)
Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Lung/microbiology , Lung/pathology , Swine/growth & development , Swine/microbiology , Animals , Animals, Newborn , Cystic Fibrosis/complications , Cystic Fibrosis/diagnostic imaging , Disease Models, Animal , Ileus/surgery , Inflammation/complications , Inflammation/pathology , Lung/abnormalities , Lung/diagnostic imaging , Meconium , Mucus/metabolism , Pancreatic Diseases/pathology , Radiography, Thoracic , Survival Analysis , Time Factors
15.
PLoS One ; 5(2): e9098, 2010 Feb 09.
Article in English | MEDLINE | ID: mdl-20161732

ABSTRACT

BACKGROUND: The PLUNC ("Palate, lung, nasal epithelium clone") protein is an abundant secretory product of epithelia present throughout the conducting airways of humans and other mammals, which is evolutionarily related to the lipid transfer/lipopolysaccharide binding protein (LT/LBP) family. Two members of this family--the bactericidal/permeability increasing protein (BPI) and the lipopolysaccharide binding protein (LBP)--are innate immune molecules with recognized roles in sensing and responding to Gram negative bacteria, leading many to propose that PLUNC may play a host defense role in the human airways. METHODOLOGY/PRINCIPAL FINDINGS: Based on its marked hydrophobicity, we hypothesized that PLUNC may be an airway surfactant. We found that purified recombinant human PLUNC greatly enhanced the ability of aqueous solutions to spread on a hydrophobic surface. Furthermore, we discovered that PLUNC significantly reduced surface tension at the air-liquid interface in aqueous solutions, indicating novel and biologically relevant surfactant properties. Of note, surface tensions achieved by adding PLUNC to solutions are very similar to measurements of the surface tension in tracheobronchial secretions from humans and animal models. Because surfactants of microbial origin can disperse matrix-encased bacterial clusters known as biofilms [1], we hypothesized that PLUNC may also have anti-biofilm activity. We found that, at a physiologically relevant concentration, PLUNC inhibited biofilm formation by the airway pathogen Pseudomonas aeruginosa in an in vitro model. CONCLUSIONS/SIGNIFICANCE: Our data suggest that the PLUNC protein contributes to the surfactant properties of airway secretions, and that this activity may interfere with biofilm formation by an airway pathogen.


Subject(s)
Biofilms/growth & development , Glycoproteins/physiology , Phosphoproteins/physiology , Pulmonary Surfactants/metabolism , Amino Acid Sequence , Biofilms/drug effects , Cells, Cultured , Circular Dichroism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Immunoblotting , Lung/cytology , Lung/metabolism , Lung/microbiology , Molecular Sequence Data , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid
16.
Contrib Microbiol ; 15: 147-163, 2008.
Article in English | MEDLINE | ID: mdl-18511860

ABSTRACT

The epithelium of the respiratory tract forms a large surface area that maintains intimate contact with the environment. Through the act of breathing, this mucosal surface encounters an array of pathogens and toxic particulates. In response to these challenges many strategies have evolved to protect the host. These include the barrier functions of the epithelium, cough, mucociliary clearance, resident professional phagocytes, and the secretion of a number of proteins and peptides with host defense functions. Thus, the surface and submucosal gland epithelium of the conducting airways is a constitutive primary participant in innate immunity. In addition, this tissue may serve the function of a secondary amplifier of innate immune responses following neurohumoral input, stimulation with cytokines from cells such as alveolar macrophages, or engagement of pattern recognition receptors. Here, we provide an overview of the airway epithelium's role in pulmonary innate immunity, especially in the context of bacterial and viral infections, emphasizing findings from human cells and selected animal models. We also provide examples of human disease states caused by impaired epithelial defenses in the lung.


Subject(s)
Cystic Fibrosis/immunology , Cystic Fibrosis/microbiology , Epithelium/immunology , Immunity, Innate , Kartagener Syndrome/immunology , Kartagener Syndrome/microbiology , Respiratory System/immunology , Animals , Anti-Infective Agents/immunology , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/pharmacology , Cystic Fibrosis/genetics , Epithelium/microbiology , Epithelium/virology , Humans , Kartagener Syndrome/genetics , Receptors, Immunologic/immunology , Respiratory System/microbiology , Respiratory System/virology
17.
J Leukoc Biol ; 83(5): 1201-6, 2008 May.
Article in English | MEDLINE | ID: mdl-18245229

ABSTRACT

Airway epithelia and neutrophils are frequently recruited to release host defense factors in response to a variety of pulmonary pathogens. One abundant product of airway epithelia is palate, lung, nasal epithelium clone (PLUNC), a proposed innate immune protein expressed in submucosal glands and surface airway epithelia. In this study, we report the expression of PLUNC in human neutrophils, a previously unrecognized source of this protein. Immunoblots performed on polymorphonuclear cell (PMN) lysates and PMN subcellular fractions indicated that PLUNC was present in the specific granules of the neutrophil. Furthermore, secretion assays demonstrated that PLUNC protein was released by neutrophils upon stimulation with secretogogues, including formyl methionyl leucyl phenylalanine and the calcium ionophore A23187. Although recombinant PLUNC protein failed to exhibit antibacterial activity in our studies, its storage and secretion by a professional phagocytic cell support the hypothesis that PLUNC participates in an aspect of the inflammatory response that contributes to host defense. These studies suggest that PLUNC expression is less restricted than previously believed, and highlight new avenues of research for the study of PLUNC function.


Subject(s)
Glycoproteins/metabolism , Neutrophils/metabolism , Phosphoproteins/metabolism , Respiratory Mucosa/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Line , Cells, Cultured , Cytoplasmic Granules/metabolism , Electrophoresis, Polyacrylamide Gel , Glycoproteins/genetics , Glycoproteins/isolation & purification , Humans , Inflammation/physiopathology , Mammals , Phosphoproteins/genetics , Phosphoproteins/isolation & purification , Recombinant Proteins/metabolism , Respiratory Mucosa/cytology , Transfection
18.
Immunol Rev ; 190: 137-45, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12493011

ABSTRACT

The completion of draft sequences of the human and mouse genomes offers many opportunities for gene discovery in the field of immunology through the application of the methods of computational genomics. One arm of the innate immune system includes the antimicrobial peptides that protect multicellular organisms from a diverse spectrum of microorganisms. The beta-defensins comprise an important family of mammalian antimicrobial peptides. To better define the beta-defensin gene family, we developed an approach to search genomic databases for conserved motifs present in the beta-defensin family using HMMER, a computational search tool based on hidden Markov models (HMMs), in combination with the basic local alignment search tool. The approach was first used to identify candidate second-exon coding regions, and later applied to finding associated first exons. This strategy discovered 28 new human and 43 new mouse beta-defensin genes in five syntenic chromosomal regions. Within each syntenic cluster, the gene sequences and organization were similar, suggesting that each cluster pair arose from a common ancestor and was retained because of conserved functions. These findings demonstrate an important proof-of-principle for a genome-wide search strategy to identify genes with conserved structural motifs. Such an approach may be readily adopted to address other questions of relevance to immunology.


Subject(s)
Genomics , Immunity, Innate/genetics , Animals , Computational Biology , Exons , Humans , Markov Chains , Mice , Multigene Family , Species Specificity , beta-Defensins/genetics
19.
Proc Natl Acad Sci U S A ; 99(4): 2129-33, 2002 Feb 19.
Article in English | MEDLINE | ID: mdl-11854508

ABSTRACT

The innate immune system includes antimicrobial peptides that protect multicellular organisms from a diverse spectrum of microorganisms. beta-Defensins comprise one important family of mammalian antimicrobial peptides. The annotation of the human genome fails to reveal the expected diversity, and a recent query of the draft sequence with the blast search engine found only one new beta-defensin gene (DEFB3). To define better the beta-defensin gene family, we adopted a genomics approach that uses hmmer, a computational search tool based on hidden Markov models, in combination with blast. This strategy identified 28 new human and 43 new mouse beta-defensin genes in five syntenic chromosomal regions. Within each syntenic cluster, the gene sequences and organization were similar, suggesting each cluster pair arose from a common ancestor and was retained because of conserved functions. Preliminary analysis indicates that at least 26 of the predicted genes are transcribed. These results demonstrate the value of a genomewide search strategy to identify genes with conserved structural motifs. Discovery of these genes represents a new starting point for exploring the role of beta-defensins in innate immunity.


Subject(s)
beta-Defensins/chemistry , beta-Defensins/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Computers , Conserved Sequence , Contig Mapping , Cysteine/chemistry , Exons , Humans , Markov Chains , Mice , Models, Genetic , Molecular Sequence Data , Multigene Family , Phylogeny , Sequence Homology, Amino Acid , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...