Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Publication year range
1.
FASEB J ; 34(8): 9884-9898, 2020 08.
Article in English | MEDLINE | ID: mdl-32596871

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by altered epithelial cell phenotypes, which are associated with myofibroblast accumulation in the lung. Atypical alveolar epithelial cells in IPF express molecular markers of airway epithelium. Polymorphisms within and around Toll interacting protein (TOLLIP) are associated with the susceptibility to IPF and mortality. However, the functional role of TOLLIP in IPF is unknown. Using lung tissues from IPF and control subjects, we showed that expression of TOLLIP gene in the lung parenchyma is globally lower in IPF compared to controls. Lung cells expressing significant levels of TOLLIP include macrophages, alveolar type II, and basal cells. TOLLIP protein expression is lower in the parenchyma of IPF lungs but is expressed in the atypical epithelial cells of the distal fibrotic regions. Using overexpression and silencing approaches, we demonstrate that TOLLIP protects cells from bleomycin-induced apoptosis using primary bronchial epithelial cells and BEAS-2B cells. The protective effects are mediated by reducing mitochondrial reactive oxygen species (ROS) levels and upregulating autophagy. Therefore, global downregulation of the TOLLIP gene in IPF lungs may predispose injured lung epithelial cells to apoptosis and to the development of IPF.


Subject(s)
Apoptosis , Bleomycin/adverse effects , Bronchi/cytology , Epithelial Cells/cytology , Idiopathic Pulmonary Fibrosis/prevention & control , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Protective Agents , Antibiotics, Antineoplastic/adverse effects , Autophagy , Bronchi/drug effects , Bronchi/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Intracellular Signaling Peptides and Proteins/genetics , Mitochondria/drug effects , Reactive Oxygen Species/metabolism
2.
Clin Infect Dis ; 70(10): 2045-2053, 2020 05 06.
Article in English | MEDLINE | ID: mdl-31504285

ABSTRACT

BACKGROUND: Neisseria meningitidis (Nm) is a nasopharyngeal commensal carried by healthy individuals. However, invasive infections occurs in a minority of individuals, with devastating consequences. There is evidence that common polymorphisms are associated with invasive meningococcal disease (IMD), but the contributions of rare variants other than those in the complement system have not been determined. METHODS: We identified familial cases of IMD in the UK meningococcal disease study and the European Union Life-Threatening Infectious Disease Study. Candidate genetic variants were identified by whole-exome sequencing of 2 patients with familial IMD. Candidate variants were further validated by in vitro assays. RESULTS: Exomes of 2 siblings with IMD identified a novel heterozygous missense mutation in BPIFA1/SPLUNC1. Sequencing of 186 other nonfamilial cases identified another unrelated IMD patient with the same mutation. SPLUNC1 is an innate immune defense protein expressed in the nasopharyngeal epithelia; however, its role in invasive infections is unknown. In vitro assays demonstrated that recombinant SPLUNC1 protein inhibits biofilm formation by Nm, and impedes Nm adhesion and invasion of human airway cells. The dominant negative mutant recombinant SPLUNC1 (p.G22E) showed reduced antibiofilm activity, increased meningococcal adhesion, and increased invasion of cells, compared with wild-type SPLUNC1. CONCLUSIONS: A mutation in SPLUNC1 affecting mucosal attachment, biofilm formation, and invasion of mucosal epithelial cells is a new genetic cause of meningococcal disease.


Subject(s)
Glycoproteins/genetics , Meningococcal Infections/genetics , Meningococcal Infections/microbiology , Neisseria meningitidis , Phosphoproteins/genetics , Complement System Proteins , Epithelial Cells , Humans , Mutation , Neisseria meningitidis/genetics
3.
Chemistry ; 26(28): 6247-6256, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32166806

ABSTRACT

In the quest for new antibiotics, two novel engineered cationic antimicrobial peptides (eCAPs) have been rationally designed. WLBU2 and D8 (all 8 valines are the d-enantiomer) efficiently kill both Gram-negative and -positive bacteria, but WLBU2 is toxic and D8 nontoxic to eukaryotic cells. We explore protein secondary structure, location of peptides in six lipid model membranes, changes in membrane structure and pore evidence. We suggest that protein secondary structure is not a critical determinant of bactericidal activity, but that membrane thinning and dual location of WLBU2 and D8 in the membrane headgroup and hydrocarbon region may be important. While neither peptide thins the Gram-negative lipopolysaccharide outer membrane model, both locate deep into its hydrocarbon region where they are primed for self-promoted uptake into the periplasm. The partially α-helical secondary structure of WLBU2 in a red blood cell (RBC) membrane model containing 50 % cholesterol, could play a role in destabilizing this RBC membrane model causing pore formation that is not observed with the D8 random coil, which correlates with RBC hemolysis caused by WLBU2 but not by D8.


Subject(s)
Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Lipopolysaccharides/chemistry , Membrane Lipids/chemistry , Pseudomonas aeruginosa/chemistry , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/metabolism , Cell Membrane/metabolism , Hemolysis , Lipopolysaccharides/metabolism , Membrane Lipids/metabolism , Microbial Sensitivity Tests , Protein Structure, Secondary
4.
J Allergy Clin Immunol ; 144(5): 1228-1241.e9, 2019 11.
Article in English | MEDLINE | ID: mdl-31301373

ABSTRACT

BACKGROUND: 15-Lipoxygenase 1 (15LO1) is expressed in airway epithelial cells in patients with type 2-high asthma in association with eosinophilia. Chronic rhinosinusitis with nasal polyps (CRSwNP) is also associated with type 2 inflammation and eosinophilia. CCL26/eotaxin 3 has been reported to be regulated by 15LO1 in lower airway epithelial cells. However, its relation to 15LO1 in patients with CRSwNP or mechanisms for its activation are unclear. OBJECTIVE: We sought to evaluate 15LO1 and CCL26 expression in nasal epithelial cells (NECs) from patients with CRSwNP and healthy control subjects (HCs) and determine whether 15LO1 regulates CCL26 in NECs through extracellular signal-regulated kinase (ERK) activation. METHODS: 15LO1, CCL26, and phosphorylated ERK were evaluated in NECs from patients with CRSwNP and HCs. 15LO1/CCL26 and CCL26/cytokeratin 5 were colocalized by means of immunofluorescence. IL-13-stimulated NECs were cultured at an air-liquid interface with or without 15-lipoxygenase 1 gene (ALOX15) Dicer-substrate short interfering RNAs (DsiRNA) transfection, a specific 15LO1 enzymatic inhibitor, and 2 ERK inhibitors. Expression of 15LO1 and CCL26 mRNA and protein was analyzed by using quantitative RT-PCR, Western blotting, and ELISA. RESULTS: 15LO1 expression was increased in nasal polyp (NP) epithelial cells compared with middle turbinate epithelial cells from patients with CRSwNP and HCs. 15LO1 expression correlated with CCL26 expression and colocalized with CCL26 expression in basal cells of the middle turbinate and NPs from patients with CRSwNP. In primary NECs in vitro, IL-13 induced 15LO1 and CCL26 expression. 15LO1 knockdown and inhibition decreased IL-13-induced ERK phosphorylation and CCL26 expression. ERK inhibition (alone) similarly decreased IL-13-induced CCL26. Phosphorylated ERK expression was increased in NECs from CRSwNP subjects and positively correlated with both 15LO1 and CCL26 expression. CONCLUSIONS: 15LO1 expression is increased in NP epithelial cells and contributes to CCL26 expression through ERK activation. 15LO1 could be considered a novel therapeutic target for CRSwNP.


Subject(s)
Arachidonate 15-Lipoxygenase/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Nasal Polyps/metabolism , Respiratory Mucosa/metabolism , Rhinitis/metabolism , Sinusitis/metabolism , Turbinates/metabolism , Adult , Arachidonate 15-Lipoxygenase/genetics , Cells, Cultured , Chemokine CCL26/metabolism , Chronic Disease , Enzyme Activation , Female , Humans , Male , Middle Aged , Nasal Polyps/complications , RNA, Small Interfering/genetics , Respiratory Mucosa/pathology , Rhinitis/complications , Sinusitis/complications , Up-Regulation
5.
Biochemistry ; 55(21): 2979-91, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27145151

ABSTRACT

SPLUNC1 is an abundantly secreted innate immune protein in the mammalian respiratory tract that exerts bacteriostatic and antibiofilm effects, binds to lipopolysaccharide (LPS), and acts as a fluid-spreading surfactant. Here, we unravel the structural elements essential for the surfactant and antimicrobial functions of human SPLUNC1 (short palate lung nasal epithelial clone 1). A unique α-helix (α4) that extends from the body of SPLUNC1 is required for the bacteriostatic, surfactant, and LPS binding activities of this protein. Indeed, we find that mutation of just four leucine residues within this helical motif to alanine is sufficient to significantly inhibit the fluid spreading abilities of SPLUNC1, as well as its bacteriostatic actions against Gram-negative pathogens Burkholderia cenocepacia and Pseudomonas aeruginosa. Conformational flexibility in the body of SPLUNC1 is also involved in the bacteriostatic, surfactant, and LPS binding functions of the protein as revealed by disulfide mutants introduced into SPLUNC1. In addition, SPLUNC1 exerts antibiofilm effects against Gram-negative bacteria, although α4 is not involved in this activity. Interestingly, though, the introduction of surface electrostatic mutations away from α4 based on the unique dolphin SPLUNC1 sequence, and confirmed by crystal structure, is shown to impart antibiofilm activity against Staphylococcus aureus, the first SPLUNC1-dependent effect against a Gram-positive bacterium reported to date. Together, these data pinpoint SPLUNC1 structural motifs required for the antimicrobial and surfactant actions of this protective human protein.


Subject(s)
Anti-Infective Agents/pharmacology , Bronchi/drug effects , Burkholderia cenocepacia/drug effects , Glycoproteins/chemistry , Glycoproteins/metabolism , Lipopolysaccharides/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Pseudomonas aeruginosa/drug effects , Amino Acid Sequence , Biofilms/drug effects , Bronchi/cytology , Burkholderia cenocepacia/immunology , Cells, Cultured , Crystallization , Crystallography, X-Ray , Glycoproteins/genetics , Humans , Immunity, Innate/drug effects , Phosphoproteins/genetics , Protein Conformation , Pseudomonas aeruginosa/immunology , Pulmonary Surfactants/chemistry , Pulmonary Surfactants/metabolism
6.
Antimicrob Agents Chemother ; 60(12): 7252-7262, 2016 12.
Article in English | MEDLINE | ID: mdl-27671059

ABSTRACT

Pseudomonas aeruginosa is the major microorganism colonizing the respiratory epithelium in cystic fibrosis (CF) sufferers. The widespread use of available antibiotics has drastically reduced their efficacy, and antimicrobial peptides (AMPs) are a promising alternative. Among them, the frog skin-derived AMPs, i.e., Esc(1-21) and its diastereomer, Esc(1-21)-1c, have recently shown potent activity against free-living and sessile forms of P. aeruginosa Importantly, this pathogen also escapes antibiotics treatment by invading airway epithelial cells. Here, we demonstrate that both AMPs kill Pseudomonas once internalized into bronchial cells which express either the functional or the ΔF508 mutant of the CF transmembrane conductance regulator. A higher efficacy is displayed by Esc(1-21)-1c (90% killing at 15 µM in 1 h). We also show the peptides' ability to stimulate migration of these cells and restore the induction of cell migration that is inhibited by Pseudomonas lipopolysaccharide when used at concentrations mimicking lung infection. This property of AMPs was not investigated before. Our findings suggest new therapeutics that not only eliminate bacteria but also can promote reepithelialization of the injured infected tissue. Confocal microscopy indicated that both peptides are intracellularly localized with a different distribution. Biochemical analyses highlighted that Esc(1-21)-1c is significantly more resistant than the all-l peptide to bacterial and human elastase, which is abundant in CF lungs. Besides proposing a plausible mechanism underlying the properties of the two AMPs, we discuss the data with regard to differences between them and suggest Esc(1-21)-1c as a candidate for the development of a new multifunctional drug against Pseudomonas respiratory infections.


Subject(s)
Amphibian Proteins/pharmacology , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Respiratory Mucosa/microbiology , Amphibian Proteins/chemistry , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Cell Movement/drug effects , Cells, Cultured , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Humans , Lipopolysaccharides , Microscopy, Confocal , Pseudomonas Infections/microbiology
7.
J Virol ; 89(23): 11935-44, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26378172

ABSTRACT

UNLABELLED: Human alveolar epithelial cells (AECs) and alveolar macrophages (AMs) are the first lines of lung defense. Here, we report that AECs are the direct targets for H1N1 viruses that have circulated since the 2009 pandemic (H1N1pdm09). AMs are less susceptible to H1N1pdm09 virus, but they produce significantly more inflammatory cytokines than AECs from the same donor. AECs form an intact epithelial barrier that is destroyed by H1N1pdm09 infection. However, there is significant variation in the cellular permissiveness to H1N1pdm09 infection among different donors. AECs from obese donors appear to be more susceptible to H1N1pdm09 infection, whereas gender, smoking history, and age do not appear to affect AEC susceptibility. There is also a difference in response to different strains of H1N1pdm09 viruses. Compared to A/California04/09 (CA04), A/New York/1682/09 (NY1682) is more infectious and causes more epithelial barrier injury, although it stimulates less cytokine production. We further determined that a single amino acid residue substitution in NY1682 hemagglutinin is responsible for the difference in infectivity. In conclusion, this is the first study of host susceptibility of human lung primary cells and the integrity of the alveolar epithelial barrier to influenza. Further elucidation of the mechanism of increased susceptibility of AECs from obese subjects may facilitate the development of novel protection strategies against influenza virus infection. IMPORTANCE: Disease susceptibility of influenza is determined by host and viral factors. Human alveolar epithelial cells (AECs) form the key line of lung defenses against pathogens. Using primary AECs from different donors, we provided cellular level evidence that obesity might be a risk factor for increased susceptibility to influenza. We also compared the infections of two closely related 2009 pandemic H1N1 strains in AECs from the same donor and identified a key viral factor that affected host susceptibility, the dominance of which may be correlated with disease epidemiology. In addition, primary human AECs can serve as a convenient and powerful model to investigate the mechanism of influenza-induced lung injury and determine the effect of genetic and epigenetic factors on host susceptibility to pandemic influenza virus infection.


Subject(s)
Epithelial Cells/virology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/physiopathology , Lung/cytology , Macrophages/metabolism , Obesity/complications , Pulmonary Alveoli/cytology , Adiposity , Cytokines/biosynthesis , Disease Susceptibility , Flow Cytometry , Hemagglutinins/genetics , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/complications , Influenza, Human/virology , Lung/virology , Macrophages/virology , Species Specificity , Statistics, Nonparametric
8.
Am J Physiol Lung Cell Mol Physiol ; 309(2): L158-67, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26001778

ABSTRACT

Suppression of type 17 immunity by type I interferon (IFN) during influenza A infection has been shown to enhance susceptibility to secondary bacterial pneumonia. Although this mechanism has been described in coinfection with gram-positive bacteria, it is unclear whether similar mechanisms may impair lung defense against gram-negative infections. Furthermore, precise delineation of the duration of type I IFN-associated susceptibility to bacterial infection remains underexplored. Therefore, we investigated the effects of preceding influenza A virus infection on subsequent challenge with the gram-negative bacteria Escherichia coli or Pseudomonas aeruginosa and the temporal association between IFN expression with susceptibility to Staphylococcus aureus challenge in a mouse model of influenza and bacterial coinfection. Here we demonstrate that preceding influenza A virus led to increased lung E. coli and P. aeruginosa bacterial burden, which was associated with suppression of type 17 immunity and attenuation of antimicrobial peptide expression. Enhanced susceptibility to S. aureus coinfection ceased at day 14 of influenza infection, when influenza-associated type I IFN levels had returned to baseline levels, further suggesting a key role for type I IFN in coinfection pathogenesis. These findings further implicate type I IFN-associated suppression of type 17 immunity and antimicrobial peptide production as a conserved mechanism for enhanced susceptibility to both gram-positive and gram-negative bacterial coinfection during influenza infection.


Subject(s)
Escherichia coli Infections/microbiology , Influenza A virus/pathogenicity , Orthomyxoviridae Infections/microbiology , Pneumonia, Bacterial/microbiology , Pneumonia/microbiology , Receptor, Interferon alpha-beta/physiology , Staphylococcal Infections/microbiology , Animals , Antimicrobial Cationic Peptides/metabolism , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Disease Susceptibility , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/virology , Influenza A virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumonia/immunology , Pneumonia/virology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/virology , Staphylococcal Infections/immunology , Staphylococcal Infections/virology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity
9.
Amino Acids ; 47(12): 2505-19, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26162435

ABSTRACT

Naturally occurring antimicrobial peptides (AMPs) represent promising future antibiotics. We have previously isolated esculentin-1a(1-21)NH2, a short peptide derived from the frog skin AMP esculentin-1a, with a potent anti-Pseudomonal activity. Here, we investigated additional functions of the peptide and properties responsible for these activities. For that purpose, we synthesized the peptide, as well as its structurally altered analog containing two D-amino acids. The peptides were then biophysically and biologically investigated for their cytotoxicity and immunomodulating activities. The data revealed that compared to the wild-type, the diastereomer: (1) is significantly less toxic towards mammalian cells, in agreement with its lower α-helical structure, as determined by circular dichroism spectroscopy; (2) is more effective against the biofilm form of Pseudomonas aeruginosa (responsible for lung infections in cystic fibrosis sufferers), while maintaining a high activity against the free-living form of this important pathogen; (3) is more stable in serum; (4) has a higher activity in promoting migration of lung epithelial cells, and presumably in healing damaged lung tissue, and (5) disaggregates and detoxifies the bacterial lipopolysaccharide (LPS), albeit less than the wild-type. Light scattering studies revealed a correlation between anti-LPS activity and the ability to disaggregate the LPS. Besides shedding light on the multifunction properties of esculentin-1a(1-21)NH2, the D-amino acid containing isomer may serve as an attractive template for the development of new anti-Pseudomonal compounds with additional beneficial properties. Furthermore, together with other studies, incorporation of D-amino acids may serve as a general approach to optimize the future design of new AMPs.


Subject(s)
Amino Acids/chemistry , Amphibian Proteins/chemistry , Antimicrobial Cationic Peptides/chemistry , Pseudomonas aeruginosa/drug effects , Amino Acid Sequence , Amphibian Proteins/chemical synthesis , Animals , Antimicrobial Cationic Peptides/chemical synthesis , Biofilms/drug effects , Cell Line , Cell Movement/drug effects , Cell Survival , Circular Dichroism , Epithelial Cells/drug effects , Humans , Lipopolysaccharides/chemistry , Macrophages/drug effects , Mice , Microbial Sensitivity Tests , Protein Structure, Tertiary , Pulmonary Alveoli/cytology , RAW 264.7 Cells , Ranidae , Skin/chemistry , Stereoisomerism
10.
J Immunol ; 191(8): 4259-68, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048904

ABSTRACT

The airway epithelium is the first line of host defense against pathogens. The short palate, lung, and nasal epithelium clone (SPLUNC)1 protein is secreted in respiratory tracts and is a member of the bacterial/permeability increasing (BPI) fold-containing protein family, which shares structural similarities with BPI-like proteins. On the basis of its homology with BPIs and restricted expression of SPLUNC1 in serous cells of submucosal glands and surface epithelial cells of the upper respiratory tract, SPLUNC1 is thought to possess antimicrobial activity in host defense. SPLUNC1 is also reported to have surfactant properties, which may contribute to anti-biofilm defenses. The objective of this study was to determine the in vivo functions of SPLUNC1 following Pseudomonas aeruginosa infection and to elucidate the underlying mechanism by using a knockout (KO) mouse model with a genetic ablation of Splunc1. Splunc1 KO mice showed accelerated mortality and increased susceptibility to P. aeruginosa infection with significantly decreased survival rates, increased bacterial burdens, exaggerated tissue injuries, and elevated proinflammatory cytokine levels as compared with those of their wild-type littermates. Increased neutrophil infiltration in Splunc1 KO mice was accompanied by elevated chemokine levels, including Cxcl1, Cxcl2, and Ccl20. Furthermore, the expression of several epithelial secretory proteins and antimicrobial molecules was considerably suppressed in the lungs of Splunc1 KO mice. The deficiency of Splunc1 in mouse airway epithelium also results in increased biofilm formation of P. aeruginosa. Taken together, our results support that the ablation of Splunc1 in mouse airways affects the mucociliary clearance, resulting in decreased innate immune response during Pseudomonas-induced respiratory infection.


Subject(s)
Glycoproteins/immunology , Phosphoproteins/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Respiratory Tract Infections/immunology , Animals , Bacterial Load/immunology , Biofilms/growth & development , Chemokine CCL20/biosynthesis , Chemokine CXCL1/biosynthesis , Chemokine CXCL2/biosynthesis , Cytokines/biosynthesis , Glycoproteins/deficiency , Glycoproteins/genetics , Lung/immunology , Lung/microbiology , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Pseudomonas Infections/mortality , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Tract Infections/metabolism , Survival Rate
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.
Am J Respir Cell Mol Biol ; 49(5): 759-67, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23721177

ABSTRACT

Carbon nanotubes (CNTs; allotropes of carbon with a cylindrical nanostructure) have emerged as one of the most commonly used types of nanomaterials, with numerous applications in industry and biomedicine. However, the inhalation of CNTs has been shown to elicit pulmonary toxicity, accompanied by a robust inflammatory response with an early-onset fibrotic phase. Epithelial host-defense proteins represent an important component of the pulmonary innate immune response to foreign inhalants such as particles and bacteria. The short palate, lung, and nasal epithelium clone-1 (SPLUNC1) protein, a member of the bactericidal/permeability-increasing-fold (BPIF)-containing protein family, is a 25-kD secretory protein that is expressed in nasal, oropharyngeal, and lung epithelia, and has been shown to have multiple functions, including antimicrobial and chemotactic activities, as well as surfactant properties. This study sought to assess the importance of SPLUNC1-mediated pulmonary responses in airway epithelial secretions, and to explore the biological relevance of SPLUNC1 to inhaled particles in a single-walled carbon nanotube (SWCNT) model. Using Scgb1a1-hSPLUNC1 transgenic mice, we observed that SPLUNC1 significantly modified host inflammatory responses by increasing leukocyte recruitment and enhancing phagocytic activity. Furthermore, we found that transgenic mice were more susceptible to SWCNT exposure at the acute phase, but showed resistance against lung fibrogenesis through pathological changes in the long term. The binding of SPLUNC1 also attenuated SWCNT-induced TNF-α secretion by RAW 264.7 macrophages. Taken together, our data indicate that SPLUNC1 is an important component of mucosal innate immune defense against pulmonary inhaled particles.


Subject(s)
Glycoproteins/metabolism , Lung/metabolism , Nanotubes, Carbon/toxicity , Phosphoproteins/metabolism , Pneumonia/metabolism , Pulmonary Fibrosis/prevention & control , Animals , Cell Line , Chemotaxis , Glycoproteins/genetics , Immunity, Innate , Immunity, Mucosal , Inflammation Mediators/metabolism , Inhalation Exposure , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lung/immunology , Lung/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Transgenic , Phagocytosis , Phosphoproteins/genetics , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
13.
J Biol Chem ; 287(33): 27948-58, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22700966

ABSTRACT

Cigarette smoke (CS) exposure is associated with increased mucus production and chronic obstructive pulmonary disease (COPD). MUC5AC is the major inducible mucus gene in the airway. The purpose of this investigation was to elucidate the mechanisms of CS-induced activation of MUC5AC gene transcription. We observed that the region -3724/-3224 of the MUC5AC promoter is critical for CS-induced gene transcriptional activity and that this region contains two Sp1 binding sites. Using a lung-relevant model, we observed that CS increased nuclear Sp1 protein expression. Consequently, CS exposure resulted in enhanced Sp1-DNA binding activity and Sp1 trans-activation. Co-transfection of the MUC5AC-luc reporter with Sp1 expression plasmids resulted in significantly increased MUC5AC-luc activity, whereas co-treatment with mithramycin A, a Sp1 inhibitor, abolished CS-induced MUC5AC promoter activity. Using mobility shift assay and chromatin immunoprecipitation, we demonstrated that two Sp1 binding sites in the MUC5AC promoter are functional and responsive to CS exposure. A mutation of either Sp1 binding site in the MUC5AC promoter significantly decreased CS-induced promoter activity. Together, these data indicate that CS induces MUC5AC gene transcription predominantly through increased Sp1 nuclear protein levels and increased Sp1 binding to its promoter region.


Subject(s)
Lung/metabolism , Mucin 5AC/biosynthesis , Response Elements , Smoking/adverse effects , Sp1 Transcription Factor/metabolism , Transcriptional Activation , Cell Line , Female , Humans , Lung/pathology , Male , Mucin 5AC/genetics , Mutation , Plicamycin/analogs & derivatives , Plicamycin/pharmacology , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics
14.
Infect Immun ; 81(1): 285-91, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23132494

ABSTRACT

The human short PLUNC1 (SPLUNC1) protein has been identified as a component of the pulmonary antimicrobial response based on its structural similarity to the bactericidal/permeability-increasing (BPI) protein. Using a genetically modified mouse model, we recently verified the antimicrobial activity of SPLUNC1 against Pseudomonas aeruginosa in vivo. To further define the mechanism of epithelial SPLUNC1-mediated antibacterial action, we carried out studies to determine how SPLUNC1 protects the host from acute respiratory infections. P. aeruginosa treated with recombinant human SPLUNC1 protein showed decreased growth in vitro. This antibacterial activity was due to growth inhibition, as a consequence of a SPLUNC1-induced increase in bacterial cell permeability. Removal of SPLUNC1 allowed the recovery of P. aeruginosa and suggested no permanent cell injury or direct killing of bacteria. Further investigation showed coating of bacterial cells by SPLUNC1. We suggest that this "bacterial cell coating" is necessary for the bacteriostatic function of SPLUNC1. Additionally, we demonstrated a novel role for SPLUNC1 as a chemoattractant that facilitated migration of macrophages and neutrophils. Taking the findings together, we propose synergistic roles for human SPLUNC1 as an antibacterial agent with bacteriostatic and chemotactic activities.


Subject(s)
Glycoproteins/immunology , Glycoproteins/metabolism , Phosphoproteins/immunology , Phosphoproteins/metabolism , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/metabolism , Animals , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/immunology , Blood Proteins/metabolism , Cell Line, Tumor , HL-60 Cells , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Neutrophils/immunology , Neutrophils/metabolism , Permeability , Protein Binding/immunology , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism
15.
J Immunol ; 187(1): 382-90, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21632717

ABSTRACT

Epithelial antimicrobial activity may protect the lung against inhaled pathogens. The bactericidal/permeability-increasing protein family has demonstrated antimicrobial activity in vitro. PLUNC (palate, lung, and nasal epithelium associated) is a 25-kDa secreted protein that shares homology with bactericidal/permeability-increasing proteins and is expressed in nasopharyngeal and respiratory epithelium. The objective of this study was to determine whether PLUNC can limit Pseudomonas aeruginosa infection in mice. Transgenic mice (Scgb1a1-hPLUNC) were generated in which human PLUNC (hPLUNC) was directed to the airway epithelium with the Scgb1a1 promoter. The hPLUNC protein (hPLUNC) was detected in the epithelium throughout the trachea and bronchial airways and in bronchoalveolar lavage fluid. Bronchoalveolar lavage fluid from transgenic mice exhibited higher antibacterial activity than that from wild type littermates in vitro. After in vivo P. aeruginosa challenge, Scgb1a1-hPLUNC transgenic mice displayed enhanced bacterial clearance. This was accompanied by a decrease in neutrophil infiltration and cytokine levels. More importantly, the overexpressed hPLUNC in Scgb1a1-hPLUNC transgenic mouse airway significantly enhanced mouse survival against P. aeruginosa-induced respiratory infection. These data indicate that PLUNC is a novel antibacterial protein that likely plays a critical role in airway epithelium-mediated innate immune response.


Subject(s)
Anti-Bacterial Agents/pharmacology , Glycoproteins/physiology , Phosphoproteins/physiology , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Animals , Anti-Bacterial Agents/administration & dosage , Female , Glycoproteins/biosynthesis , Glycoproteins/genetics , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/physiology , Mice , Mice, Inbred Strains , Mice, Transgenic , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Uteroglobin/biosynthesis , Uteroglobin/genetics , Uteroglobin/physiology
16.
Am J Pathol ; 178(5): 2159-67, 2011 May.
Article in English | MEDLINE | ID: mdl-21514430

ABSTRACT

Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein is highly expressed in normal airways, but is dramatically decreased in allergic and cigarette smoke exposure settings. We have previously demonstrated SPLUNC1 in vitro antibacterial property against Mycoplasma pneumoniae (Mp). However, its in vivo biological functions remain unclear. The objectives of this study were to determine the in vivo functions of SPLUNC1 following bacterial (eg, Mp) infection, and to examine the underlying mechanisms. We generated SPLUNC1-deficient mice and utilized transgenic mice overexpressing human SPLUNC1 exclusively within the airway epithelium. These mice were infected with Mp and, twenty-four hours post infection, their host defense responses were compared to littermate controls. Mp levels and inflammatory cells increased in the lungs of SPLUNC1(-/-) mice as compared to wild type controls. SPLUNC1 deficiency was shown to contribute to impaired neutrophil activation. In contrast, mice overexpressing hSPLUNC1 exclusively in airway epithelial cells demonstrated lower Mp levels. Furthermore, neutrophil elastase activity was significantly increased in mice overexpressing hSPLUNC1. Lastly, we demonstrated that SPLUNC1 enhanced Mp-induced human neutrophil elastase (HNE) activity, and HNE directly inhibited the growth of Mp. Our findings demonstrate a critical in vivo role of SPLUNC1 in host defense against bacterial infection, and likely provide a novel therapeutic approach to restore impaired lung innate immune responses to bacteria in patients with chronic lung diseases.


Subject(s)
Glycoproteins/immunology , Immunity, Innate/immunology , Mycoplasma pneumoniae/immunology , Phosphoproteins/immunology , Pneumonia, Mycoplasma/immunology , Animals , Blotting, Western , Glycoproteins/metabolism , Humans , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mycoplasma pneumoniae/metabolism , Phosphoproteins/metabolism , Pneumonia, Mycoplasma/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Front Microbiol ; 13: 889791, 2022.
Article in English | MEDLINE | ID: mdl-35694289

ABSTRACT

The threat of antibiotic resistance warrants the discovery of agents with novel antimicrobial mechanisms. Antimicrobial peptides (AMPs) directly disrupting bacterial membranes may overcome resistance to traditional antibiotics. AMP development for clinical use has been mostly limited to topical application to date. We developed a rational framework for systematically addressing this challenge using libraries composed of 86 novel Trp- and Arg-rich engineered peptides tested against clinical strains of the most common multidrug-resistant bacteria known as ESKAPE pathogens. Structure-function correlations revealed minimum lengths (as low as 16 residues) and Trp positioning for maximum antibacterial potency with mean minimum inhibitory concentration (MIC) of 2-4 µM and corresponding negligible toxicity to mammalian cells. Twelve peptides were selected based on broad-spectrum activity against both gram-negative and -positive bacteria and <25% toxicity to mammalian cells at maximum test concentrations. Most of the selected PAX remained active against the colistin-resistant clinical strains. Of the selected peptides, the shortest (the 16-residue E35) was further investigated for antibacterial mechanism and proof-of-concept in vivo efficacy. E35 killed an extensively-resistant isolate of Pseudomonas aeruginosa (PA239 from the CDC, also resistant to colistin) by irreversibly disrupting the cell membranes as shown by propidium iodide incorporation, using flow cytometry and live cell imaging. As proof of concept, in vivo toxicity studies showed that mice tolerated a systemic dose of up to 30 mg/kg peptide and were protected with a single 5 mg/kg intravenous (IV) dose against an otherwise lethal intraperitoneal injection of PA239. Efficacy was also demonstrated in an immune-compromised Klebsiella pneumoniae infection model using a daily dose of 4mg/kg E35 systemically for 2 days. This framework defines the determinants of efficacy of helical AMPs composed of only cationic and hydrophobic amino acids and provides a path for a potential departure from the restriction to topical use of AMPs toward systemic application.

18.
ACS Chem Biol ; 17(4): 987-997, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35290019

ABSTRACT

The emergence of resistance to clinically used antibiotics by bacteria presents a significant problem in public health. Natural antimicrobial peptides (AMPs) are a valuable source of antibiotics that act by a mechanism less prone to the evolutionary development of resistance. In an effort to realize the promise of AMPs while overcoming limitations such as poor biostability, researchers have sought sequence-defined oligomers with artificial amide-based backbones that show membrane-disrupting functions similar to natural agents. Most of this precedent has focused on short peptidomimetic analogues of unstructured chains or secondary folds; however, the natural antimicrobial arsenal includes a number of small- and medium-sized proteins that act via an ordered tertiary structure. Generating proteomimetic analogues of these scaffolds poses a challenge due to the increased complexity of the target for mimicry. Here, we report the development of heterogeneous-backbone variants of lasiocepsin, a 27-residue disulfide-rich AMP found in bee venom that adopts a compact tertiary fold. Iterative cycles of design, synthesis, and biological evaluation yielded analogues of the natural domain with ∼30 to 40% artificial backbone content, comparable antibacterial activity, reduced host cell toxicity, and improved stability to proteolytic degradation. High-resolution structures determined for several variants by NMR provide insights into the interplay among backbone composition, tertiary fold, and biological properties. Collectively, the results reported here broaden the scope of protein functional mimicry by artificial backbone analogues of tertiary folding patterns and suggest protein backbone engineering as a means to tune protein function by exerting site-specific control over protein folded structure.


Subject(s)
Bee Venoms , Disulfides , Anti-Bacterial Agents/pharmacology , Antimicrobial Peptides , Disulfides/chemistry , Peptides, Cyclic , Proteins/chemistry
19.
J Phys Chem B ; 126(36): 6922-6935, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36067064

ABSTRACT

In an effort to combat rising antimicrobial resistance, our labs have rationally designed cationic, helical, amphipathic antimicrobial peptides (AMPs) as alternatives to traditional antibiotics since AMPs incur bacterial resistance in weeks, rather than days. One highly positively charged AMP, WLBU2 (+13e), (RRWV RRVR RWVR RVVR VVRR WVRR), has been shown to be effective in killing both Gram-negative (G(-)) and Gram-positive (G(+)) bacteria by directly perturbing the bacterial membrane nonspecifically. Previously, we used two equilibrium experimental methods: synchrotron X-ray diffuse scattering (XDS) providing lipid membrane thickness and neutron reflectometry (NR) providing WLBU2 depth of penetration into three lipid model membranes (LMMs). The purpose of the present study is to use the results from the scattering experiments to guide molecular dynamics (MD) simulations to investigate the detailed biophysics of the interactions of WLBU2 with LMMs of Gram-negative outer and inner membranes, and Gram-positive cell membranes, to elucidate the mechanisms of bacterial killing. Instead of coarse-graining, backmapping, or simulating without bias for several microseconds, all-atom (AA) simulations were guided by the experimental results and then equilibrated for ∼0.5 µs. Multiple replicas of the inserted peptide were run to probe stability and reach a combined time of at least 1.2 µs for G(-) and also 2.0 µs for G(+). The simulations with experimental comparisons help rule out certain structures and orientations and propose the most likely set of structures, orientations, and effects on the membrane. The simulations revealed that water, phosphates, and ions enter the hydrocarbon core when WLBU2 is positioned there. For an inserted peptide, the three types of amino acids, arginine, tryptophan, and valine (R, W, V), are arranged with the 13 Rs extending from the hydrocarbon core to the phosphate group, Ws are located at the interface, and Vs are more centrally located. For a surface state, R, W, and V are positioned relative to the bilayer interface as expected from their hydrophobicities, with Rs closest to the phosphate group, Ws close to the interface, and Vs in between. G(-) and G(+) LMMs are thinned ∼1 Å by the addition of WLBU2. Our results suggest a dual anchoring mechanism for WLBU2 both in the headgroup and in the hydrocarbon region that promotes a defect region where water and ions can flow across the slightly thinned bacterial cell membrane.


Subject(s)
Antimicrobial Peptides , Molecular Dynamics Simulation , Antimicrobial Cationic Peptides/chemistry , Bacteria/metabolism , Lipid Bilayers/chemistry , Lipids , Phosphates , Water
20.
Biochem Soc Trans ; 39(4): 1051-5, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21787346

ABSTRACT

PLUNC (palate, lung and nasal epithelium clone)-associated gene originally referred to one gene, but now has been extended to represent a gene family that consists of a number of genes with peptide sequence homologies and predicted structural similarities. PLUNC-like proteins display sequence homology with BPI (bactericidal/permeability-increasing protein), a 456-residue cationic protein produced by precursors of polymorphonuclear leucocytes that have been shown to possess both bactericidal and LPS (lipopolysaccharide)-binding activities. The human PLUNC is also known as LUNX (lung-specific X protein), NASG (nasopharyngeal carcinoma-related protein) and SPURT (secretory protein in upper respiratory tract). The gene originally named PLUNC is now recognized as SPLUNC1. Its gene product SPLUNC1 is a secretory protein that is abundantly expressed in cells of the surface epithelium in the upper respiratory tracts and secretory glands in lung, and in the head and the neck region. The functional role of SPLUNC1 in innate immunity has been suggested but not clearly defined. The present review describes recent findings that support antimicrobial and anti-inflammatory functions of SPLUNC1 in Gram-negative bacteria-induced respiratory infection.


Subject(s)
Glycoproteins/metabolism , Gram-Negative Bacterial Infections/immunology , Immunity, Innate , Phosphoproteins/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Glycoproteins/pharmacology , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/metabolism , Lung/immunology , Lung/metabolism , Lung/microbiology , Organ Specificity , Phosphoproteins/pharmacology , Protein Binding , Respiratory Mucosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL