Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 13: 1125901, 2023.
Article in English | MEDLINE | ID: mdl-37009499

ABSTRACT

Introduction: Pseudomonas aeruginosa is known for its ability to form biofilms, which are dependent on the production of exopolysaccharides. During chronic colonization of the airway and biofilm formation, P. aeruginosa converts to a mucoid phenotype, indicating production of the exopolysaccharide alginate. The mucoid phenotype promotes resistance to phagocytic killing, but the mechanism has not been established. Methods and Results: To better understand the mechanism of phagocytic evasion conferred by alginate production, Human (THP-1) and murine (MH-S) macrophage cell lines were used to determine the effects of alginate production on macrophage binding, signaling and phagocytosis. Phagocytosis assays using mucoid clinical isolate FRD1 and its non-mucoid algD mutant showed that alginate production inhibited opsonic and non-opsonic phagocytosis, but exogenous alginate was not protective. Alginate caused a decrease in binding to murine macrophages. Blocking antibodies to CD11b and CD14 showed that these receptors were important for phagocytosis and were blocked by alginate. Furthermore, alginate production decreased the activation of signaling pathways required for phagocytosis. Mucoid and non-mucoid bacteria induced similar levels of MIP-2 from murine macrophages. Discussion: This study demonstrated for the first time that alginate on the bacterial surface inhibits receptor-ligand interactions important for phagocytosis. Our data suggest that there is a selection for alginate conversion that blocks the earliest steps in phagocytosis, leading to persistence during chronic pulmonary infections.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Mice , Animals , Humans , Pseudomonas aeruginosa/genetics , Phagocytosis , Macrophages , Biofilms , Cell Line , Pseudomonas Infections/microbiology
2.
Pharmaceutics ; 15(4)2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37111762

ABSTRACT

Treatments for acute respiratory distress syndrome are still unavailable, and the prevalence of the disease has only increased due to the COVID-19 pandemic. Mechanical ventilation regimens are still utilized to support declining lung function but also contribute to lung damage and increase the risk for bacterial infection. The anti-inflammatory and pro-regenerative abilities of mesenchymal stromal cells (MSCs) have shown to be a promising therapy for ARDS. We propose to utilize the regenerative effects of MSCs and the extracellular matrix (ECM) in a nanoparticle. Our mouse MSC (MMSC) ECM nanoparticles were characterized using size, zeta potential, and mass spectrometry to evaluate their potential as pro-regenerative and antimicrobial treatments. The nanoparticles had an average size of 273.4 nm (±25.6) and possessed a negative zeta potential, allowing them to surpass defenses and reach the distal regions of the lung. It was found that the MMSC ECM nanoparticles are biocompatible with mouse lung epithelial cells and MMSCs, increasing the wound healing rate of human lung fibroblasts while also inhibiting the growth of Pseudomonas aeruginosa, a common lung pathogen. Our MMSC ECM nanoparticles display characteristics of healing injured lungs while preventing bacterial infection, which can increase recovery time.

3.
PLoS One ; 16(6): e0252970, 2021.
Article in English | MEDLINE | ID: mdl-34133429

ABSTRACT

Pseudomonas aeruginosa secretes several endopeptidases, including elastase, alkaline proteinase (Apr), a lysine-specific endopeptidase (LysC), and an aminopeptidase (PaAP), all of which are important virulence factors. Activation of the endopeptidases requires removal of an inhibitory N-terminal propeptide. Activation of pro-PaAP, in contrast, requires C-terminal processing. The activating proteases of pro-PaAP and their cleavage site(s) have not yet been defined. Studying pro-PaAP processing in a wild type P. aeruginosa strain and strains lacking either elastase or both elastase and Apr, we detected three processing variants, each ~56 kDa in size (AP56). Activity assays and N- and C-terminal sequence analyses of these variants pointed at LysC as the principal activating protease, cleaving a Lys512-Ala513 peptide bond at the C-terminal end of pro-PaAP. Elastase and/or Apr are required for activation of LysC, suggesting both are indirectly involved in activation of PaAP. To shed light on the function(s) of the N-terminal domain of AP56, we purified recombinant AP56 and generated from it the 28 kDa catalytic domain (AP28). The kinetic constants (Km and Kcat) for hydrolysis of Leu-, Lys-, Arg- and Met-p-nitroanilide (pNA) derivatives by AP56 and AP28 were then determined. The catalytic coefficients (Kcat/Km) for hydrolysis of all four substrates by AP28 and AP56 were comparable, indicating that the non-catalytic domain is not involved in hydrolysis of small substrates. It may, however, regulate hydrolysis of natural peptides/proteins. Lys-pNA was hydrolyzed 2 to 3-fold more rapidly than Leu-pNA and ~8-fold faster than Arg- or Met-pNA, indicating that Lys-pNA was the preferred substrate.


Subject(s)
Aminopeptidases/metabolism , Metalloendopeptidases/metabolism , Pancreatic Elastase/metabolism , Proteolysis , Pseudomonas aeruginosa/enzymology , Amino Acid Sequence , Aminopeptidases/genetics , Catalytic Domain , Hydrolysis , Kinetics , Metalloendopeptidases/genetics , Pancreatic Elastase/genetics , Protein Domains , Pseudomonas aeruginosa/growth & development , Substrate Specificity
4.
PLoS One ; 14(6): e0216401, 2019.
Article in English | MEDLINE | ID: mdl-31158231

ABSTRACT

Mucoid mucA22 Pseudomonas aeruginosa (PA) is an opportunistic lung pathogen of cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) patients that is highly sensitive to acidified nitrite (A-NO2-). In this study, we first screened PA mutant strains for sensitivity or resistance to 20 mM A-NO2- under anaerobic conditions that represent the chronic stages of the aforementioned diseases. Mutants found to be sensitive to A-NO2- included PA0964 (pmpR, PQS biosynthesis), PA4455 (probable ABC transporter permease), katA (major catalase, KatA) and rhlR (quorum sensing regulator). In contrast, mutants lacking PA0450 (a putative phosphate transporter) and PA1505 (moaA2) were A-NO2- resistant. However, we were puzzled when we discovered that mucA22 mutant bacteria, a frequently isolated mucA allele in CF and to a lesser extent COPD, were more sensitive to A-NO2- than a truncated ΔmucA deletion (Δ157-194) mutant in planktonic and biofilm culture, as well as during a chronic murine lung infection. Subsequent transcriptional profiling of anaerobic, A-NO2--treated bacteria revealed restoration of near wild-type transcript levels of protective NO2- and nitric oxide (NO) reductase (nirS and norCB, respectively) in the ΔmucA mutant in contrast to extremely low levels in the A-NO2--sensitive mucA22 mutant. Proteins that were S-nitrosylated by NO derived from A-NO2- reduction in the sensitive mucA22 strain were those involved in anaerobic respiration (NirQ, NirS), pyruvate fermentation (UspK), global gene regulation (Vfr), the TCA cycle (succinate dehydrogenase, SdhB) and several double mutants were even more sensitive to A-NO2-. Bioinformatic-based data point to future studies designed to elucidate potential cellular binding partners for MucA and MucA22. Given that A-NO2- is a potentially viable treatment strategy to combat PA and other infections, this study offers novel developments as to how clinicians might better treat problematic PA infections in COPD and CF airway diseases.


Subject(s)
Bacterial Proteins/genetics , Biofilms , Lung/microbiology , Mutation , Nitrites/pharmacology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Bacterial Proteins/metabolism , Biofilms/drug effects , Chronic Disease , Humans , Hydrogen-Ion Concentration , Plankton/metabolism , Plankton/physiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism
5.
ACS Appl Mater Interfaces ; 11(23): 20699-20714, 2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31117452

ABSTRACT

Infection is a serious medical complication associated with health care environments. Despite advances, the 5-10% incidence of infections for hospital patients is well documented. Sources of pathogenic organisms include medical devices such as catheters and endotracheal tubes. Offering guidance for curbing the spread of such infections, a model antimicrobial coating is described herein that kills bacteria on contact but is compatible with human cells. To achieve these characteristics, a novel blend of a conventional biomedical grade polyurethane (Tecoflex) with mixed soft block polyurethane is described. The functional polyurethane (UP-C12-50-T) has a copolyoxetane soft block P-C12-50 with quaternary ammonium (C12) and PEG-like side chains and a conventional poly(tetramethylene oxide) (PTMO, T) soft block. DSC and DMA data point to limited miscibility of UP-C12-50-T with Tecoflex. The blend of Tecoflex with 10 wt % UP-C12-50-T designated UP-C12-50-T-10 radically changed surface properties. Evidence for surface concentration of the P-C12-50 soft block was obtained by atomic force microscopy (AFM), dynamic contact angles (DCAs), zeta potentials (ζ), and X-ray photoelectron spectroscopy (XPS). The antimicrobial effectiveness of the blend coatings was established by the ASTM E2149 "shake flask" test for challenges of E. coli and a methicillin resistant strain of S. epidermidis. Cytocompatibility was demonstrated with an in vitro test designed for direct contact (ISO 10993-5). Growth of human mesenchymal stem cells (MSCs) beside and under UP-C12-50-T-10 indicated remarkable biocompatibility for a composition that is also strongly antimicrobial. Overall, the results point to a model coating with a level of P-C12-50 that combines high antimicrobial effectiveness and low toxicity to human cells.


Subject(s)
Anti-Infective Agents/chemistry , Biocompatible Materials/chemistry , Oxides/chemistry , Polyethylene Glycols/chemistry , Polyurethanes/chemistry , Calorimetry, Differential Scanning , Chromatography, Gel , Humans , Microscopy, Atomic Force , Molecular Structure , Photoelectron Spectroscopy
6.
Macromol Biosci ; 17(9)2017 09.
Article in English | MEDLINE | ID: mdl-28605136

ABSTRACT

A preliminary study is reported for a polycation antimicrobial peptide (AMP) mimic against Propionibacterium acnes, which is associated with acne vulgaris, a common skin condition. Antibiotics are commonly used against P. acnes but buildup of resistance is well-known. Worse, antibiotic regimens build up resistance for more sensitive bacteria such as Staphylococcus epidermidis. The polycation AMP mimic C12-50, 1, is chosen for the present study as it has been previously shown to have high antimicrobial effectiveness. This study reports that C12-50 is active against P. acnes (strain ATCC 6919) with a minimum inhibitory concentration (MIC) of 6.3 µg mL-1 . To monitor resistance build-up ten passages are conducted with C12-50 against P. acnes. The MIC remains constant with no resistance buildup. Parallel studies with erythromycin confirm previously reported resistance buildup. The results point to a promising pathway to applications for polycation AMP mimics against P. acnes.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Propionibacterium acnes/drug effects , Acne Vulgaris , Anti-Infective Agents/pharmacology , Drug Resistance, Microbial , Humans , Microbial Sensitivity Tests , Polyamines/pharmacology , Polyelectrolytes
7.
Langmuir ; 32(12): 2975-84, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26948099

ABSTRACT

Real-time atomic force microscopy (AFM) was used for analyzing effects of the antimicrobial polycation copolyoxetane P[(C12)-(ME2Ox)-50/50], C12-50 on the membrane of a model bacterium, Escherichia coli (ATCC# 35218). AFM imaging showed cell membrane changes with increasing C12-50 concentration and time including nanopore formation and bulges associated with outer bacterial membrane disruption. A macroscale bactericidal concentration study for C12-50 showed a 4 log kill at 15 µg/mL with conditions paralleling imaging (1 h, 1x PBS, physiological pH, 25 °C). The dramatic changes from the control image to 1 h after introducing 15 µg/mL C12-50 are therefore reasonably attributed to cell death. At the highest concentration (60 µg/mL) further cell membrane disruption results in leakage of cytoplasm driven by detergent-like action. The sequence of processes for initial membrane disruption by the synthetic polycation C12-50 follows the carpet model posited for antimicrobial peptides (AMPs). However, the nanoscale details are distinctly different as C12-50 is a synthetic, water-soluble copolycation that is best modeled as a random coil. In a complementary AFM study, chemical force microscopy shows that incubating cells with C12-50 decreased the hydrophobicity across the entire cell surface at an early stage. This finding provides additional evidence indicating that C12-50 polycations initially bind with the cell membrane in a carpet-like fashion. Taken together, real time AFM imaging elucidates the mechanism of antimicrobial action for copolyoxetane C12-50 at the single cell level. In future work this approach will provide important insights into structure-property relationships and improved antimicrobial effectiveness for synthetic amphiphilic polycations.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Polyurethanes/pharmacology , Surface-Active Agents/pharmacology , Anti-Bacterial Agents/chemical synthesis , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Microscopy, Atomic Force , Polylysine/chemistry , Polyurethanes/chemistry , Surface-Active Agents/chemical synthesis
8.
J Biol Chem ; 290(20): 12451-62, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25817996

ABSTRACT

Pseudomonas aeruginosa is an opportunistic human pathogen that secretes the exopolysaccharide alginate during infection of the respiratory tract of individuals afflicted with cystic fibrosis and chronic obstructive pulmonary disease. Among the proteins required for alginate production, Alg44 has been identified as an inner membrane protein whose bis-(3',5')-cyclic dimeric guanosine monophosphate (c-di-GMP) binding activity post-translationally regulates alginate secretion. In this study, we report the 1.8 Å crystal structure of the cytoplasmic region of Alg44 in complex with dimeric self-intercalated c-di-GMP and characterize its dinucleotide-binding site using mutational analysis. The structure shows that the c-di-GMP binding region of Alg44 adopts a PilZ domain fold with a dimerization mode not previously observed for this family of proteins. Calorimetric binding analysis of residues in the c-di-GMP binding site demonstrate that mutation of Arg-17 and Arg-95 alters the binding stoichiometry between c-di-GMP and Alg44 from 2:1 to 1:1. Introduction of these mutant alleles on the P. aeruginosa chromosome show that the residues required for binding of dimeric c-di-GMP in vitro are also required for efficient alginate production in vivo. These results suggest that the dimeric form of c-di-GMP represents the biologically active signaling molecule needed for the secretion of an important virulence factor produced by P. aeruginosa.


Subject(s)
Alginates/chemistry , Cyclic GMP/analogs & derivatives , Protein Multimerization , Pseudomonas aeruginosa/chemistry , Virulence Factors/chemistry , Bacterial Proteins , Binding Sites , Crystallography, X-Ray , Cyclic GMP/chemistry , Cyclic GMP/genetics , Cyclic GMP/metabolism , Glucuronic Acid/chemistry , Glucuronic Acid/genetics , Glucuronic Acid/metabolism , Hexuronic Acids/chemistry , Humans , Membrane Proteins , Mutation , Protein Structure, Quaternary , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Genome Announc ; 3(2)2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25792066

ABSTRACT

We announce here the complete genome sequence of the Pseudomonas aeruginosa mucoid strain FRD1, isolated from the sputum of a cystic fibrosis patient. The complete genome of P. aeruginosa FRD1 is 6,712,339 bp. This genome will allow comparative genomics to be used to identify genes associated with virulence, especially those involved in chronic pulmonary infections.

10.
Microbiology (Reading) ; 161(Pt 1): 30-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25336469

ABSTRACT

The ECF (extracytoplasmic function) alternative sigma factor, σ(22) (AlgT/U), is required for expression of the algD promoter of the operon for alginate biosynthesis in Pseudomonas aeruginosa. Alginate production promotes chronic pulmonary infections by this opportunistic pathogen in patients with cystic fibrosis and chronic obstructive pulmonary disease. σ(22) is normally sequestered, but its deregulation and activation occur either by mutation in mucA (encoding an anti-sigma factor) or in response to envelope stress, such as inhibition of peptidoglycan synthesis. The σ(22) stress response system includes many genes in addition to those for alginate. In the present study, we characterized an intergenic region between ORFs PA2559 and PA2560 in PAO1 for a σ(22)-dependent, stress-responsive transcript, described here as PA2559.1. Northern analysis and transcript end-mapping indicated the PA2559.1 transcript was ~310 nt in length. Examination of the DNA sequence upstream of +1 revealed a σ(22) core promoter motif, GAATTT-N16-TCTGT, and site-directed mutagenesis confirmed this to be a σ(22)-dependent promoter that was highly activated during cell wall stress. PA2559.1 also contained an ORF that demonstrated increased translational activity upon cell wall stress. As determined by mutant analysis, the protein encoded by PA2559.1 was shown to play a positive role in the σ(22)-dependent activation of the algD promoter under stress in both sessile (i.e. biofilm) and planktonic conditions. Thus, it appeared to act as a stress response facilitator and so was named SrfA. The sequence of SrfA was found to be novel in nature and extremely well conserved only in P. aeruginosa, suggesting that it is of high evolutionary importance in this species.


Subject(s)
Bacterial Proteins/genetics , Cell Wall/metabolism , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Sigma Factor/metabolism , Stress, Physiological , Alginates , Amino Acid Sequence , Bacterial Proteins/chemistry , Base Sequence , DNA Mutational Analysis , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Gene Order , Glucuronic Acid/biosynthesis , Hexuronic Acids , Molecular Sequence Data , Mutation , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Transcription, Genetic
11.
PLoS Pathog ; 10(8): e1004334, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25165982

ABSTRACT

The O-acetylation of polysaccharides is a common modification used by pathogenic organisms to protect against external forces. Pseudomonas aeruginosa secretes the anionic, O-acetylated exopolysaccharide alginate during chronic infection in the lungs of cystic fibrosis patients to form the major constituent of a protective biofilm matrix. Four proteins have been implicated in the O-acetylation of alginate, AlgIJF and AlgX. To probe the biological function of AlgJ, we determined its structure to 1.83 Å resolution. AlgJ is a SGNH hydrolase-like protein, which while structurally similar to the N-terminal domain of AlgX exhibits a distinctly different electrostatic surface potential. Consistent with other SGNH hydrolases, we identified a conserved catalytic triad composed of D190, H192 and S288 and demonstrated that AlgJ exhibits acetylesterase activity in vitro. Residues in the AlgJ signature motifs were found to form an extensive network of interactions that are critical for O-acetylation of alginate in vivo. Using two different electrospray ionization mass spectrometry (ESI-MS) assays we compared the abilities of AlgJ and AlgX to bind and acetylate alginate. Binding studies using defined length polymannuronic acid revealed that AlgJ exhibits either weak or no detectable polymer binding while AlgX binds polymannuronic acid specifically in a length-dependent manner. Additionally, AlgX was capable of utilizing the surrogate acetyl-donor 4-nitrophenyl acetate to catalyze the O-acetylation of polymannuronic acid. Our results, combined with previously published in vivo data, suggest that the annotated O-acetyltransferases AlgJ and AlgX have separate and distinct roles in O-acetylation. Our refined model for alginate acetylation places AlgX as the terminal acetlytransferase and provides a rationale for the variability in the number of proteins required for polysaccharide O-acetylation.


Subject(s)
Alginates/metabolism , Bacterial Proteins/metabolism , Pseudomonas aeruginosa/enzymology , Acetylation , Bacterial Proteins/chemistry , Base Sequence , Glucuronic Acid/metabolism , Hexuronic Acids/metabolism , Molecular Sequence Data , Protein Structure, Quaternary
12.
Biomacromolecules ; 15(2): 456-67, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24422429

ABSTRACT

The alkyl chain length of quaternary ammonium/PEG copolyoxetanes has been varied to discern effects on solution antimicrobial efficacy, hemolytic activity and cytotoxicity. Monomers 3-((4-bromobutoxy)methyl)-3-methyloxetane (BBOx) and 3-((2-(2-methoxyethoxy)ethoxy)methyl)-3-methyloxetane (ME2Ox) were used to prepare precursor P[(BBOx)(ME2Ox)-50:50-4 kDa] copolyoxetane via cationic ring opening polymerization. The 1:1 copolymer composition and Mn (4 kDa) were confirmed by (1)H NMR spectroscopy. After C-Br substitution by a series of tertiary amines, ionic liquid Cx-50 copolyoxetanes were obtained, where 50 is the mole percent of quaternary repeat units and "x" is quaternary alkyl chain length (2, 6, 8, 10, 12, 14, or 16 carbons). Modulated differential scanning calorimetry (MDSC) studies showed Tgs between -40 and -60 °C and melting endotherms for C14-50 and C16-50. Minimum inhibitory concentrations (MIC) were determined for Escherichia coli , Staphylococcus aureus , and Pseudomonas aeruginosa . A systematic dependence of MIC on alkyl chain length was found. The most effective antimicrobials were in the C6-50 to C12-50 range. C8-50 had better overall performance with MICs of 4 µg/mL, E. coli ; 2 µg/mL, S. aureus ; and 24 µg/mL, P. aeruginosa . At 5 × MIC, C8-50 effected >99% kill in 1 h against S. aureus , E. coli , and P. aeruginosa challenges of 10(8) cfu/mL; log reductions (1 h) were 7, 3, and 5, respectively. To provide additional insight into polycation interactions with bacterial membranes, a geometric model based on the dimensions of E. coli is described that provides an estimate of the maximum number of polycations that can chemisorb. Chain dimensions were estimated for polycation C8-50 with a molecular weight of 5 kDa. Considering the approximations for polycation chemisorption (PCC), it is surprising that a calculation based on geometric considerations gives a C8-50 concentration within a factor of 2 of the MIC, 4.0 (±1.2) µg/mL for E. coli . Cx-50 copolyoxetane cytotoxicity was low for human red blood cells, human dermal fibroblasts (HDF), and human foreskin fibroblasts (HFF). Selectivities for bacterial kill over cell lysis were among the highest ever reported for polycations indicating good prospects for biocompatibility.


Subject(s)
Anti-Bacterial Agents/pharmacology , Fibroblasts/drug effects , Hemolysis/drug effects , Polyethylene Glycols/pharmacology , Polymers/pharmacology , Propylene Glycols/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cell Line , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Humans , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polymers/chemical synthesis , Polymers/chemistry , Propylene Glycols/chemical synthesis , Propylene Glycols/chemistry , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Structure-Activity Relationship
13.
J Biol Chem ; 288(31): 22299-314, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23779107

ABSTRACT

The exopolysaccharide alginate, produced by mucoid Pseudomonas aeruginosa in the lungs of cystic fibrosis patients, undergoes two different chemical modifications as it is synthesized that alter the properties of the polymer and hence the biofilm. One modification, acetylation, causes the cells in the biofilm to adhere better to lung epithelium, form microcolonies, and resist the effects of the host immune system and/or antibiotics. Alginate biosynthesis requires 12 proteins encoded by the algD operon, including AlgX, and although this protein is essential for polymer production, its exact role is unknown. In this study, we present the X-ray crystal structure of AlgX at 2.15 Å resolution. The structure reveals that AlgX is a two-domain protein, with an N-terminal domain with structural homology to members of the SGNH hydrolase superfamily and a C-terminal carbohydrate-binding module. A number of residues in the carbohydrate-binding module form a substrate recognition "pinch point" that we propose aids in alginate binding and orientation. Although the topology of the N-terminal domain deviates from canonical SGNH hydrolases, the residues that constitute the Ser-His-Asp catalytic triad characteristic of this family are structurally conserved. In vivo studies reveal that site-specific mutation of these residues results in non-acetylated alginate. This catalytic triad is also required for acetylesterase activity in vitro. Our data suggest that not only does AlgX protect the polymer as it passages through the periplasm but that it also plays a role in alginate acetylation. Our results provide the first structural insight for a wide group of closely related bacterial polysaccharide acetyltransferases.


Subject(s)
Alginates/metabolism , Bacterial Proteins/physiology , Pseudomonas aeruginosa/metabolism , Acetylation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Glucuronic Acid/metabolism , Hexuronic Acids/metabolism , Models, Molecular , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation
14.
Curr Microbiol ; 65(6): 770-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22986819

ABSTRACT

While much is known about the transcriptional regulation of the 12 gene alginate biosynthesis operon from the algD promoter in Pseudomonas aeruginosa, there has been little investigation into the possibility of other transcription starts within this operon, especially those genes dealing with the epimerization and acetylation of the alginate polymer. In this study, we utilized quantitative reverse transcription polymerase chain reaction, a ß-galactosidase reporter assay and sequence scanning to identify two putative promoters within the alginate biosynthesis operon upstream of the alginate epimerase gene algG and the alginate acetylation gene algI. These data support the possibility of differential regulation within the operon to alter polymer structure under varying environmental conditions.


Subject(s)
Alginates/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Operon , Promoter Regions, Genetic/genetics , Pseudomonas aeruginosa/metabolism , Acetylation , Bacterial Proteins/genetics , Base Sequence , Culture Media , Genes, Reporter , Molecular Sequence Data , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , beta-Galactosidase/genetics
15.
mBio ; 3(3)2012.
Article in English | MEDLINE | ID: mdl-22589289

ABSTRACT

UNLABELLED: The Pseudomonas aeruginosa extracytoplasmic functioning (ECF) sigma factor σ(22) is encoded by algT/algU and is inhibited by anti-sigma factor MucA. σ(22) was originally discovered for its essential role in the expression of the exopolysaccharide alginate by mucoid strains associated with chronic pulmonary infection. However, σ(22) is now known to also have a large regulon associated with the response to cell wall stress. Our recent transcriptome analysis identified 293 open reading frames (ORFs) in the σ(22) stress stimulon that include genes for outer envelope biogenesis and remodeling, although most of the genes have undefined functions. To better understand the σ(22)-dependent stress response, mutants affected in 27 genes of the σ(22) stimulon were examined and expression was studied with lacZ fusions. Mutants constructed in the 27 genes showed no major change in response to cell wall-acting antibiotics or growth at elevated temperatures nor in alginate production. The mutants were examined for their effects on the expression of the σ(22)-dependent promoter of the alginate biosynthetic operon (PalgD) as a measure of σ(22) derepression from MucA. By testing PalgD expression under both planktonic and sessile growth conditions, 11 genes were found to play a role in the stress response that activates σ(22). Some mutations caused an increase or a decrease in the response to cell wall stress. Interestingly, mutations in 7 of the 11 genes caused constitutive PalgD expression under nonstressed conditions and thus showed that these genes are involved in maintaining envelope homeostasis. Mutations in PA0062 and PA1324 showed constitutive PalgD expression during both the planktonic and the sessile modes of growth. However, the PA5178 mutation caused constitutive PalgD expression only during planktonic growth. In contrast, mutations in PA2717, PA0567, PA3040, and PA0920 caused constitutive PalgD expression only in the sessile/biofilm mode of growth. This provides evidence that the σ(22) stimulon for cell envelope homeostasis overlaps with biofilm control mechanisms. IMPORTANCE: During chronic lung infections, such as in cystic fibrosis patients, Pseudomonas aeruginosa produces the exopolysaccharide alginate and forms biofilms that shield the organisms from the immune response and increase resistance to antibiotics. Activation of alginate genes is under the control of an extracytoplasmic stress response system that releases an alternative sigma factor (σ(22)) in response to cell wall stress and then activates expression of a large regulon. In this study, a mutant analysis of 27 members of the regulon showed that 11 play a role in envelope homeostasis and affect the stress response system itself. Interestingly, some genes demonstrate effects only in either the planktonic (free-swimming) or the sessile (biofilm) mode of growth, which leads to persistence and antibiotic tolerance. The studies presented here provide an important initial step in dissecting the mechanisms that regulate a critical signal transduction pathway that impacts P. aeruginosa pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/metabolism , Gene Expression Regulation, Bacterial , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/genetics , Regulon , Sigma Factor/metabolism , Bacterial Proteins/genetics , Cell Membrane/genetics , Operon , Pseudomonas aeruginosa/metabolism , Sigma Factor/genetics
16.
Graefes Arch Clin Exp Ophthalmol ; 250(2): 223-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21947265

ABSTRACT

BACKGROUND: Therapy of S. aureus keratitis is increasingly challenging due to emerging resistant strains. Staphylolysin (LasA protease) is a staphylolytic endopeptidase secreted by Pseudomonas aeruginosa. The purpose of the current study was to study the effect of treatment with staphylolysin on experimental keratitis caused by various Staphylococcus aureus strains. METHODS: The therapeutic effect was studied in a keratitis model induced in rabbits by intrastromal injections of 10(3) S. aureus cells of three different methicillin-resistant S. aureus (MRSA) strains and one methicillin-susceptible S. aureus strain (MSSA). Topical treatment with either staphylolysin or bovine serum albumin (BSA; control) was applied every half hour for 5 h, starting at 4 h after infection. Corneas were removed for bacterial quantification. Histopathological analysis was performed on MSSA-infected rabbits, killed at either one or 84 h after completion of treatment and on uninfected eyes 1 h after treatment termination. RESULTS: The number of bacteria in the staphylolysin-treated corneas was significantly reduced in all infections with the four S. aureus strains studied as compared to controls: the staphylolysin-treated eyes infected with MRSA strains were either completely sterilized or showed a 3-4 orders of magnitude decrease in the number of cfu/cornea (p = 0.004 to 0.005); all of the staphylolysin-treated MSSA-infected eyes were sterile. Histopathological analysis of the methicillin-sensitive (MSSA) strain-infected eyes at 84 h after completion of treatment showed moderate inflammation in the staphylolysin-treated eyes as compared with extensive abscess formation in the control group. The uninfected corneas showed only mild stromal edema in both the staphylolysin and BSA-treated groups. CONCLUSIONS: Staphylolysin provided long-lasting protection against several strains of S. aureus, evident by both its strong anti-bacterial activity and beneficial histopathological results of treatment.


Subject(s)
Corneal Ulcer/drug therapy , Endotoxins/therapeutic use , Eye Infections, Bacterial/drug therapy , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/drug therapy , Administration, Topical , Animals , Colony Count, Microbial , Cornea/microbiology , Corneal Ulcer/microbiology , Disease Models, Animal , Endotoxins/administration & dosage , Eye Infections, Bacterial/microbiology , Rabbits , Staphylococcal Infections/microbiology , Treatment Outcome
17.
Gene ; 498(2): 242-53, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22088575

ABSTRACT

Infection by the opportunistic pathogen Pseudomonas aeruginosa is a leading cause of morbidity and mortality seen in cystic fibrosis (CF) patients. This is mainly due to the genotypic and phenotypic changes of the bacteria that cause conversion from a typical nonmucoid to a mucoid form in the CF lung. Mucoid conversion is indicative of overproduction of a capsule-like polysaccharide called alginate. The alginate-overproducing (Alg(+)) mucoid phenotype seen in the CF isolates is extremely unstable. Low oxygen tension growth of mucoid variants readily selects for nonmucoid variants. The switching off mechanism has been mapped to the algT/U locus, and the molecular basis for this conversion was partially attributed to mutations in the algT/U gene itself. To further characterize molecular changes resulting in the unstable phenotype, an isogenic PAO1 derivative that is constitutively Alg(+) due to the replacement of the mucA with mucA22 (PDO300) was used. The mucA22 allele is common in mucoid CF isolates. Thirty-four spontaneous nonmucoid variants, or sap (suppressor of alginate production) mutants, of PDO300 were isolated under low oxygen tension. About 40% of the sap mutants were rescued by a plasmid carrying algT/U (Group A). The remaining sap mutants were not (Group B). The members of Group B fall into two subsets: one similar to PAO1, and another comparable to PDO300. Sequence analysis of the algT/U and mucA genes in Group A shows that mucA22 is intact, whereas algT/U contains mutations. Genetic complementation and sequencing of one Group B sap mutant, sap22, revealed that the nonmucoid phenotype was due to the presence of a mutation in PA3257. PA3257 encodes a putative periplasmic protease. Mutation of PA3257 resulted in decreased algT/U expression. Thus, inhibition of algT/U is a primary mechanism for alginate synthesis suppression.


Subject(s)
Alginates/metabolism , Bacterial Proteins/genetics , Pseudomonas aeruginosa/genetics , Sigma Factor/genetics , Amino Acid Sequence , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Base Sequence , DNA Transposable Elements , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Glucuronic Acid/metabolism , Hexuronic Acids/metabolism , Microbial Sensitivity Tests , Molecular Sequence Data , Mutation , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Sigma Factor/metabolism , beta-Lactamases/genetics , beta-Lactamases/metabolism
18.
PLoS One ; 6(11): e27091, 2011.
Article in English | MEDLINE | ID: mdl-22069491

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that causes both acute pneumonitis in immunocompromised patients and chronic lung infections in individuals with cystic fibrosis and other bronchiectasis. Over 75% of clinical isolates of P. aeruginosa secrete elastase B (LasB), an elastolytic metalloproteinase that is encoded by the lasB gene. Previously, in vitro studies have demonstrated that LasB degrades a number of components in both the innate and adaptive immune systems. These include surfactant proteins, antibacterial peptides, cytokines, chemokines and immunoglobulins. However, the contribution of LasB to lung infection by P. aeruginosa and to inactivation of pulmonary innate immunity in vivo needs more clarification. In this study, we examined the mechanisms underlying enhanced clearance of the ΔlasB mutant in mouse lungs. The ΔlasB mutant was attenuated in virulence when compared to the wild-type strain PAO1 during lung infection in SP-A+/+ mice. However, the ΔlasB mutant was as virulent as PAO1 in the lungs of SP-A⁻/⁻ mice. Detailed analysis showed that the ΔlasB mutant was more susceptible to SP-A-mediated opsonization but not membrane permeabilization. In vitro and in vivo phagocytosis experiments revealed that SP-A augmented the phagocytosis of ΔlasB mutant bacteria more efficiently than the isogenic wild-type PAO1. The ΔlasB mutant was found to have a severely reduced ability to degrade SP-A, consequently making it unable to evade opsonization by the collectin during phagocytosis. These results suggest that P. aeruginosa LasB protects against SP-A-mediated opsonization by degrading the collectin.


Subject(s)
Bacterial Proteins/metabolism , Lung/microbiology , Metalloendopeptidases/metabolism , Phagocytosis/physiology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/enzymology , Pulmonary Surfactant-Associated Protein A/physiology , Animals , Bacterial Proteins/genetics , Blotting, Western , Cell Membrane Permeability , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Lung/immunology , Lung/metabolism , Macrophages/cytology , Macrophages/metabolism , Metalloendopeptidases/genetics , Mice , Mice, Inbred C3H , Mice, Knockout , Mutation/genetics , Opsonin Proteins/metabolism , Pseudomonas Infections/pathology , Pseudomonas Infections/prevention & control
19.
Proc Natl Acad Sci U S A ; 108(32): 13083-8, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21778407

ABSTRACT

Pseudomonas aeruginosa is the predominant pathogen associated with chronic lung infection among cystic fibrosis patients. During colonization of the lung, P. aeruginosa converts to a mucoid phenotype characterized by the overproduction of the exopolysaccharide alginate. Secretion of newly synthesized alginate across the outer membrane is believed to occur through the outer membrane protein AlgE. Here we report the 2.3 Å crystal structure of AlgE, which reveals a monomeric 18-stranded ß-barrel characterized by a highly electropositive pore constriction formed by an arginine-rich conduit that likely acts as a selectivity filter for the negatively charged alginate polymer. Interestingly, the pore constriction is occluded on either side by extracellular loop L2 and an unusually long periplasmic loop, T8. In halide efflux assays, deletion of loop T8 (ΔT8-AlgE) resulted in a threefold increase in anion flux compared to the wild-type or ΔL2-AlgE supporting the idea that AlgE forms a transport pathway through the membrane and suggesting that transport is regulated by T8. This model is further supported by in vivo experiments showing that complementation of an algE deletion mutant with ΔT8-AlgE impairs alginate production. Taken together, these studies support a mechanism for exopolysaccharide export across the outer membrane that is distinct from the Wza-mediated translocation observed in canonical capsular polysaccharide export systems.


Subject(s)
Bacterial Proteins/chemistry , Cell Membrane/metabolism , Pseudomonas aeruginosa/metabolism , Alginates , Bacterial Proteins/metabolism , Biological Transport , Conserved Sequence , Glucuronic Acid/metabolism , Hexuronic Acids , Models, Molecular , Periplasm/metabolism , Pliability , Polysaccharides/metabolism , Porins/metabolism , Porosity , Protein Structure, Secondary , Structural Homology, Protein , Substrate Specificity
20.
Biomacromolecules ; 12(3): 757-69, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21280645

ABSTRACT

This study focuses on the solution antimicrobial effectiveness of a novel class of copolyoxetanes with quaternary ammonium and PEG-like side chains. A precursor P[(BBOx-m)(ME2Ox)] copolyoxetane was prepared by cationic ring-opening copolymerization of 3-((4-bromobutoxy)methyl)-3-methyloxetane (BBOx) and 3-((2-(2-methoxyethoxy)ethoxy)methyl)-3-methyloxetane (ME2Ox) to give random copolymers with 14-100 (m) mol % BBOx. Reaction of P[(BBOx-m)(ME2Ox)] with dodecyl dimethylamine gave the corresponding quaternary P[(C12-m)(ME2Ox)] polycation salts, designated C12-m, as viscous liquids in 100% yield. BBOx/ME2Ox and C12/ME2Ox ratios were obtained by (1)H NMR spectroscopy. C12-m molecular weights (M(n), 3.5-21.9 kDa) were obtained from (1)H NMR end group analysis. DSC studies up to 150 °C showed only thermal transitions between -69 and -34 °C assigned to T(g) values. Antibacterial activity for the C12-m copolyoxetanes was tested by determining minimum inhibitory concentrations (MICs) against Gram(+) Staphylococcus aureus and Gram(-) Escherichia coli and Pseudomonas aeruginosa . MIC decreased with increasing C12 mol percent, reaching a minimum in the range C12-43 to C12-60. Overall, the antimicrobial with consistently low MICs for the three tested pathogenic bacteria was C12-43: (bacteria, MIC, µg/mL) E. coli (6), S. aureus (5), and P. aeruginosa (33). For C12-43, minimum biocidal concentration (MBC) to reach 99.99% kill in 24 h required 1.5× MIC for S. aureus and 2× MIC for E. coli and P. aeruginosa . At 5× MIC against a challenge of 10(8) cfu/mL, C12-43 kills ≥99% S. aureus , E. coli , and P. aeruginosa within 1 h. C12-m copolyoxetane cytotoxicity toward human red blood cells was low, indicating good prospects for biocompatibility. The tunability of C12-m copolyoxetane compositions, effective antimicrobial behavior against Gram(+) and Gram(-) bacteria, and promising biocompatibility offer opportunities for further modification and potential applications as therapeutic agents.


Subject(s)
Anti-Infective Agents/chemistry , Biocompatible Materials/chemistry , Epoxy Compounds , Polymers , Propylene Glycols , Bacteria/drug effects , Microbial Sensitivity Tests , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...