Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Sci Rep ; 10(1): 4135, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139768

ABSTRACT

Bacterial type III secretion systems (T3SSs) play an important role in pathogenesis of Gram-negative infections. Enteropathogenic and enterohemorrhagic Escherichia coli contain a well-defined T3SS but in addition a second T3SS termed E. coli T3SS 2 (ETT2) has been described in a number of strains of E. coli. The majority of pathogenic E. coli contain elements of a genetic locus encoding ETT2, but which has undergone significant mutational attrition rendering it without predicted function. Only a very few strains have been reported to contain an intact ETT2 locus. To investigate the occurrence of the ETT2 locus in strains of human pathogenic E. coli, we carried out genomic sequencing of 162 isolates obtained from patient blood cultures in Scotland. We found that 22 of 26 sequence type (ST) 69 isolates from this collection contained an intact ETT2 together with an associated eip locus which encodes putative secreted ETT2 effectors as well as eilA, a gene encoding a putative transcriptional regulator of ETT2 associated genes. Using a reporter gene for eilA activation, we defined conditions under which this gene was differentially activated. Analysis of published E. coli genomes with worldwide representation showed that ST69 contained an intact ETT2 in these strains as well. The conservation of the genes encoding ETT2 in human pathogenic ST69 strains strongly suggests it has importance in infection, although its exact functional role remains obscure.


Subject(s)
Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Type III Secretion Systems/metabolism , Escherichia coli Infections/genetics , Gene Expression Regulation, Bacterial , Humans , Mutation
2.
mSystems ; 4(4)2019 Aug 13.
Article in English | MEDLINE | ID: mdl-31409659

ABSTRACT

Streptococcus pneumoniae is the dominant cause of community-acquired pneumonia worldwide. Invasion of the pleural space is common and results in increased mortality. We set out to determine the bacterial and host factors that influence invasion of the pleural space. In a murine model of pneumococcal infection, we isolated neutrophil-dominated samples of bronchoalveolar and pleural fluid containing bacteria 48 hours after infection. Using dual RNA sequencing (RNA-seq), we characterized bacterial and host transcripts that were differentially regulated between these compartments and bacteria in broth and resting neutrophils, respectively. Pleural and lung samples showed upregulation of genes involved in the positive regulation of neutrophil extravasation but downregulation of genes mediating bacterial killing. Compared to the lung samples, cells within the pleural space showed marked upregulation of many genes induced by type I interferons, which are cytokines implicated in preventing bacterial transmigration across epithelial barriers. Differences in the bacterial transcripts between the infected samples and bacteria grown in broth showed the upregulation of genes in the bacteriocin locus, the pneumococcal surface adhesin PsaA, and the glycopeptide resistance gene vanZ; the gene encoding the ClpP protease was downregulated in infection. One hundred sixty-nine intergenic putative small bacterial RNAs were also identified, of which 43 (25.4%) small RNAs had been previously described. Forty-two of the small RNAs were upregulated in pleura compared to broth, including many previously identified as being important in virulence. Our results have identified key host and bacterial responses to invasion of the pleural space that can be potentially exploited to develop alternative antimicrobial strategies for the prevention and treatment of pneumococcal pleural disease.IMPORTANCE The factors that regulate the passage of bacteria between different anatomical compartments are unclear. We have used an experimental model of infection with Streptococcus pneumoniae to examine the host and bacterial factors involved in the passage of bacteria from the lung to the pleural space. The transcriptional profile of host and bacterial cells within the pleural space and lung was analyzed using deep sequencing of the entire transcriptome using the technique of dual RNA-seq. We found significant differences in the host and bacterial RNA profiles in infection, which shed light on the key factors that allow passage of this bacterium into the pleural space.

3.
PLoS Pathog ; 14(5): e1007099, 2018 05.
Article in English | MEDLINE | ID: mdl-29813133

ABSTRACT

Streptococcus pneumoniae is the major bacterial cause of community-acquired pneumonia, and the leading agent of childhood pneumonia deaths worldwide. Nasal colonization is an essential step prior to infection. The cytokine IL-17 protects against such colonization and vaccines that enhance IL-17 responses to pneumococcal colonization are being developed. The role of IL-17 in host defence against pneumonia is not known. To address this issue, we have utilized a murine model of pneumococcal pneumonia in which the gene for the IL-17 cytokine family receptor, Il17ra, has been inactivated. Using this model, we show that IL-17 produced predominantly from γδ T cells protects mice against death from the invasive TIGR4 strain (serotype 4) which expresses a relatively thin capsule. However, in pneumonia produced by two heavily encapsulated strains with low invasive potential (serotypes 3 and 6B), IL-17 significantly enhanced mortality. Neutrophil uptake and killing of the serotype 3 strain was significantly impaired compared to the serotype 4 strain and depletion of neutrophils with antibody enhanced survival of mice infected with the highly encapsulated SRL1 strain. These data strongly suggest that IL-17 mediated neutrophil recruitment to the lungs clears infection from the invasive TIGR4 strain but that lung neutrophils exacerbate disease caused by the highly encapsulated pneumococcal strains. Thus, whilst augmenting IL-17 immune responses against pneumococci may decrease nasal colonization, this may worsen outcome during pneumonia caused by some strains.


Subject(s)
Interleukin-17/immunology , Pneumonia, Pneumococcal/immunology , Receptors, Interleukin-17/genetics , Streptococcus pneumoniae/immunology , Animals , Bacteremia/immunology , Bacteremia/microbiology , Bacterial Capsules/immunology , Bacterial Capsules/ultrastructure , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/microbiology , Disease Models, Animal , Lung/cytology , Lung/enzymology , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nasopharynx/microbiology , Neutrophils/cytology , Neutrophils/immunology , Peroxidase/metabolism , Phagocytosis , Pneumonia, Pneumococcal/mortality , Pneumonia, Pneumococcal/prevention & control , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Specific Pathogen-Free Organisms , Streptococcus pneumoniae/ultrastructure
4.
BMC Genomics ; 18(1): 807, 2017 Oct 23.
Article in English | MEDLINE | ID: mdl-29058583

ABSTRACT

BACKGROUND: The bacterial pathogen Streptococcus pneumoniae colonizes the nasopharynx prior to causing disease, necessitating successful competition with the resident microflora. Cytokines of the IL-17 family are important in host defence against this pathogen but their effect on the nasopharyngeal microbiome is unknown. Here we analyse the influence of IL-17 on the composition and interactions of the nasopharyngeal microbiome before and after pneumococcal colonization. RESULTS: Using a murine model and 16S rRNA profiling, we found that a lack of IL-17 signalling led to profound alterations in the nasal but not lung microbiome characterized by decreased diversity and richness, increases in Proteobacteria and reduction in Bacteroidetes, Actinobacteria and Acidobacteria. Following experimental pneumococcal nasal inoculation, animals lacking IL-17 family signalling showed increased pneumococcal colonization, though both wild type and knockout animals showed as significant disruption of nasal microbiome composition, with increases in the proportion of Proteobacteria, even in animals that did not have persistent colonization. Sparse correlation analysis of the composition of the microbiome at various time points after infection showed strong positive interactions within the Firmicutes and Proteobacteria, but strong antagonism between members of these two phyla. CONCLUSIONS: These results show the powerful influence of IL-17 signalling on the composition of the nasal microbiome before and after pneumococcal colonization, and apparent lack of interspecific competition between pneumococci and other Firmicutes. IL-17 driven changes in nasal microbiome composition may thus be an important factor in successful resistance to pneumococcal colonization and potentially could be manipulated to augment host defence against this pathogen.


Subject(s)
Interleukin-17/metabolism , Microbiota , Nasal Mucosa/metabolism , Pneumococcal Infections/genetics , Streptococcus pneumoniae/physiology , Animals , Genetic Variation , Interleukin-17/genetics , Lung/cytology , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/cytology , Nasal Mucosa/microbiology , Pneumococcal Infections/microbiology , Receptors, Interleukin-17/metabolism , Ribotyping , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/isolation & purification , Streptococcus pneumoniae/pathogenicity
5.
Innate Immun ; 23(5): 413-423, 2017 07.
Article in English | MEDLINE | ID: mdl-28399692

ABSTRACT

Neutrophils play an important role in the innate immune response to infection with Streptococcus pneumoniae, the pneumococcus. Pneumococci are phagocytosed by neutrophils and undergo killing after ingestion. Other cellular processes may also be induced, including autophagy and the formation of neutrophil extracellular traps (NETs), which may play a role in bacterial eradication. We set out to determine how these different processes interacted following pneumococcal infection of neutrophils, and the role of the major pneumococcal toxin pneumolysin in these various pathways. We found that pneumococci induced autophagy in neutrophils in a type III phosphatidylinositol-3 kinase dependent fashion that also required the autophagy gene Atg5. Pneumolysin did not affect this process. Phagocytosis was inhibited by pneumolysin but enhanced by autophagy, while killing was accelerated by pneumolysin but inhibited by autophagy. Pneumococci induced extensive NET formation in neutrophils that was not influenced by pneumolysin but was critically dependent on autophagy. While pneumolysin did not affect NET formation, it had a potent inhibitory effect on bacterial trapping within NETs. These findings show a complex interaction between phagocytosis, killing, autophagy and NET formation in neutrophils following pneumococcal infection that contribute to host defence against this pathogen.


Subject(s)
Autophagy-Related Protein 5/metabolism , Extracellular Traps/immunology , Neutrophils/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/immunology , Streptolysins/immunology , Autophagy , Autophagy-Related Protein 5/genetics , Bacterial Proteins/immunology , Bacteriolysis , Cells, Cultured , Humans , Immunity, Innate , Neutrophil Activation , Phagocytosis , Reactive Oxygen Species/metabolism
6.
PLoS One ; 9(2): e90263, 2014.
Article in English | MEDLINE | ID: mdl-24587305

ABSTRACT

BACKGROUND: We set out to determine the magnitude of antigen-specific memory T helper cell responses to Pseudomonas aeruginosa in healthy humans and patients with cystic fibrosis. METHODS: Peripheral blood human memory CD4(+) T cells were co-cultured with dendritic cells that had been infected with different strains of Pseudomonas aeruginosa. The T helper response was determined by measuring proliferation, immunoassay of cytokine output, and immunostaining of intracellular cytokines. RESULTS: Healthy individuals and patients with cystic fibrosis had robust antigen-specific memory CD4(+) T cell responses to Pseudomonas aeruginosa that not only contained a Th1 and Th17 component but also Th22 cells. In contrast to previous descriptions of human Th22 cells, these Pseudomonal-specific Th22 cells lacked the skin homing markers CCR4 or CCR10, although were CCR6(+). Healthy individuals and patients with cystic fibrosis had similar levels of Th22 cells, but the patient group had significantly fewer Th17 cells in peripheral blood. CONCLUSIONS: Th22 cells specific to Pseudomonas aeruginosa are induced in both healthy individuals and patients with cystic fibrosis. Along with Th17 cells, they may play an important role in the pulmonary response to this microbe in patients with cystic fibrosis and other conditions.


Subject(s)
Cystic Fibrosis/complications , Cystic Fibrosis/immunology , Pseudomonas Infections/complications , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adult , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Dendritic Cells/immunology , Dendritic Cells/microbiology , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunologic Memory , Lymphocyte Activation , Male , T-Cell Antigen Receptor Specificity/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult
7.
MMWR Morb Mortal Wkly Rep ; 63(4): 73-6, 2014 Jan 31.
Article in English | MEDLINE | ID: mdl-24476978

ABSTRACT

Increasingly, the need to strengthen global capacity to prevent, detect, and respond to public health threats around the globe is being recognized. CDC, in partnership with the World Health Organization (WHO), has committed to building capacity by assisting member states with strengthening their national capacity for integrated disease surveillance and response as required by International Health Regulations (IHR). CDC and other U.S. agencies have reinforced their pledge through creation of global health security (GHS) demonstration projects. One such project was conducted during March-September 2013, when the Uganda Ministry of Health (MoH) and CDC implemented upgrades in three areas: 1) strengthening the public health laboratory system by increasing the capacity of diagnostic and specimen referral networks, 2) enhancing the existing communications and information systems for outbreak response, and 3) developing a public health emergency operations center (EOC) (Figure 1). The GHS demonstration project outcomes included development of an outbreak response module that allowed reporting of suspected cases of illness caused by priority pathogens via short messaging service (SMS; i.e., text messaging) to the Uganda District Health Information System (DHIS-2) and expansion of the biologic specimen transport and laboratory reporting system supported by the President's Emergency Plan for AIDS Relief (PEPFAR). Other enhancements included strengthening laboratory management, establishing and equipping the EOC, and evaluating these enhancements during an outbreak exercise. In 6 months, the project demonstrated that targeted enhancements resulted in substantial improvements to the ability of Uganda's public health system to detect and respond to health threats.


Subject(s)
Capacity Building/organization & administration , Disease Outbreaks/prevention & control , Global Health , International Cooperation , Population Surveillance , Centers for Disease Control and Prevention, U.S. , Humans , Uganda , United States , World Health Organization
9.
PLoS One ; 7(3): e33408, 2012.
Article in English | MEDLINE | ID: mdl-22428045

ABSTRACT

BACKGROUND: E. coli O157 carries two genes encoding the effector proteins NleH1 and NleH2 which are 87% identical. Despite the similarity between the proteins, the promoter regions upstream of the genes encoding the effectors are more divergent suggesting that the actual expression of the genes may be differentially regulated. This was tested by creating reporter fusions and examining their expression in different genetic backgrounds, media and on contact with host cells. The function of the proteins was also tested following transfection into host cells. PRINCIPAL FINDINGS: Expression of both NleH1 and NleH2 was enhanced when cultured under conditions that stimulated expression of the Type Three Secretion System (T3SS) and was influenced by the regulators Ler and GrlA. Maximal expression of NleH1 required 531 bp of the upstream untranslated region but NleH2 required only 113 bp. Interestingly, contact with host cells strongly repressed expression of both NleH1 and NleH2. Following transfection, both proteins produced only minor effects on NF-κB activation when assessed using a NF-κB luciferase reporter assay, a result that is consistent with the recent report demonstrating the dependence on RPS3 for NleH1 modulation of NF-κB. SIGNIFICANCE: This study demonstrates the importance of considering gene regulation when studying bacterial effector proteins. Despite their sequence similarity, NleH1 and NleH2 are expressed differentially and may, therefore, be translocated at distinct times during an infection.


Subject(s)
Bacterial Secretion Systems/genetics , Escherichia coli O157/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics , DNA Primers/genetics , HEK293 Cells , Humans , Luciferases , Microscopy, Fluorescence , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction , Trans-Activators/metabolism
10.
Future Microbiol ; 7(1): 33-46, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22191445

ABSTRACT

Serotype 1 Streptococcus pneumoniae is among the most commonly isolated serotype in invasive pneumococcal disease but is rarely found causing asymptomatic nasopharyngeal colonization. Compared to infection by other serotypes, infection caused by serotype 1 is more likely to be identified in young patients without comorbidities but is generally associated with a lower mortality. Empyema and extrapulmonary manifestations are common. Outbreaks of serotype 1 disease have been reported in closed communities and epidemics are particularly common in sub-Saharan Africa. The serotype 1 capsular polysaccharide is a zwitterionic structure that enables it to function as a T-cell dependent antigen under some circumstances, in contrast to other pneumococcal capsular polysaccharides that are T-cell independent antigens. There are also differences in the key virulence factor pneumolysin in some serotype 1 isolates. The clinical significance of these differences remains to be determined.


Subject(s)
Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Africa South of the Sahara/epidemiology , Animals , Bacterial Capsules/chemistry , Bacterial Capsules/immunology , Humans , Pneumococcal Infections/epidemiology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Virulence Factors/genetics , Virulence Factors/immunology
11.
Cell Microbiol ; 13(3): 388-401, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20955240

ABSTRACT

IL-1ß is produced from inactive pro-IL-1ß by activation of caspase-1 brought about by a multi-subunit protein platform called the inflammasome. Many bacteria can trigger inflammasome activity through flagellin activation of the host protein NLRC4. However, strains of the common human pathogen Pseudomonas aeruginosa lacking flagellin can still activate the inflammasome. We set out to identify what non-flagellin components could produce this activation. Using mass spectroscopy, we identified an inflammasome-activating factor from P. aeruginosa as pilin, the major component of the type IV bacterial pilus. Purified pilin introduced into mouse macrophages by liposomal delivery activated caspase-1 and led to secretion of mature IL-1ß, as did recombinant pilin purified from Escherichia coli. This was dependent on caspase-1 but not on the host inflammasome proteins NLRC4, NLRP3 or ASC. Mutants of P. aeruginosa strain PA103 lacking pilin did not activate the inflammasome following infection of macrophages with live bacteria. Type III secretion remained intact in the absence of pili, showing this was not due to a lack of effector delivery. Our observations show pilin is a novel activator of the inflammasome in addition to flagellin and the recently described PrgJ protein family, the basal body rod component of the type III apparatus.


Subject(s)
Fimbriae Proteins/immunology , Fimbriae Proteins/metabolism , Inflammasomes/immunology , Macrophages/immunology , Pseudomonas aeruginosa/immunology , Animals , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , CARD Signaling Adaptor Proteins , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Cytoskeletal Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Escherichia coli/metabolism , Fimbriae Proteins/genetics , Flagellin/genetics , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Pseudomonas aeruginosa/genetics
12.
J Biol Chem ; 285(14): 10508-18, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20097760

ABSTRACT

Many Gram-negative bacteria possess a type III secretion system (TTSS( paragraph sign)) that can activate the NLRC4 inflammasome, process caspase-1 and lead to secretion of mature IL-1beta. This is dependent on the presence of intracellular flagellin. Previous reports have suggested that this activation is independent of extracellular K(+) and not accompanied by leakage of K(+) from the cell, in contrast to activation of the NLRP3 inflammasome. However, non-flagellated strains of Pseudomonas aeruginosa are able to activate NLRC4, suggesting that formation of a pore in the cell membrane by the TTSS apparatus may be sufficient for inflammasome activation. Thus, we set out to determine if extracellular K(+) influenced P. aeruginosa inflammasome activation. We found that raising extracellular K(+) prevented TTSS NLRC4 activation by the non-flagellated P. aeruginosa strain PA103DeltaUDeltaT at concentrations above 90 mm, higher than those reported to inhibit NLRP3 activation. Infection was accompanied by efflux of K(+) from a minority of cells as determined using the K(+)-sensitive fluorophore PBFI, but no formation of a leaky pore. We obtained exactly the same results following infection with Salmonella typhimurium, previously described as independent of extracellular K(+). The inhibitory effect of raised extracellular K(+) on NLRC4 activation thus reflects a requirement for a decrease in intracellular K(+) for this inflammasome component as well as that described for NLRP3.


Subject(s)
Bacterial Proteins/metabolism , Inflammation/immunology , Interleukin-1beta/metabolism , Potassium/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Animals , Caspase 1/metabolism , Immunoblotting , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Salmonella Infections/immunology , Salmonella Infections/metabolism
13.
Cell Microbiol ; 8(8): 1294-309, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16882033

ABSTRACT

Type III secretion is a widespread method whereby Gram-negative bacteria introduce toxins into eukaryotic cells. These toxins mimic or subvert a normal cellular process by interacting with a specific target, although how toxins reach their site of action is unclear. We set out to investigate the intracellular localization of a type III toxin of Pseudomonas aeruginosa called ExoU, which has phospholipase activity and requires a eukaryotic factor for activity. We found that ExoU is localized to the plasma membrane and undergoes modification within the cell by addition of two ubiquitin molecules at lysine-178. A region of five amino acids at position 679-683 near the C-terminus of the ExoU protein controls both membrane localization and ubiquitinylation. Site-directed mutagenesis identified a tryptophan at position 681 as crucial for these effects. We found that the same region at position 679-683 was also required for cell toxicity produced by ExoU as well as in vitro phospholipase activity. Localization of the phospholipase ExoU to the plasma membrane is thus required for activation and allows efficient utilization of adjacent substrate phospholipids.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Pseudomonas aeruginosa/pathogenicity , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/toxicity , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Base Sequence , Cell Membrane/metabolism , Cell Membrane/microbiology , DNA, Bacterial/genetics , HeLa Cells , Humans , Lysine/chemistry , Models, Biological , Molecular Weight , Mutagenesis, Site-Directed , Pseudomonas aeruginosa/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Transfection , Ubiquitin/chemistry , Ubiquitin/metabolism
14.
Microbiology (Reading) ; 152(Pt 8): 2273-2285, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16849794

ABSTRACT

Pseudomonas aeruginosa secretes a number of toxins by a type III system, and these are important in virulence. One of them, ExoS, is a bifunctional toxin, with a GTPase-activating protein domain, as well as ADP ribosyltransferase (ADPRT) activity. These two domains have numerous potential cellular targets, but the overall mechanism of ExoS action remains unclear. The effects of ExoS in a simple eukaryotic system, the yeast Saccharomyces cerevisiae, using a tetracycline-regulated expression system were studied. This system allowed controlled expression of ExoS in yeast, which was not possible using a galactose-induced system. ExoS was found to be an extremely potent inhibitor of yeast growth, and to be largely dependent on the activity of its ADPRT domain. ExoS produced a dramatic alteration in actin distribution, with the appearance of large aggregates of cortical actin, and thickened disorganized cables, entirely dependent on the ADPRT domain. This phenotype is suggestive of actin stabilization, which was verified by showing that the cortical aggregates of actin induced by ExoS were resistant to treatment with latrunculin A, an agent that prevents actin polymerization. ExoS increased the numbers of mating projections produced following growth arrest with mating pheromone, and prevented subsequent DNA replication, an effect that is again dependent on the ADPRT domain. Following pheromone removal, ExoS produced altered development of the mating projections, which became elongated with a swollen bud-like tip. These results suggest alternative pathways for ExoS action in eukaryotic cells that may result from activation of small GTPases, and this yeast expression system is well suited to explore these pathways.


Subject(s)
ADP Ribose Transferases/physiology , Saccharomyces cerevisiae/metabolism , ADP Ribose Transferases/chemistry , Actins/analysis , Actins/metabolism , Bacterial Toxins/chemistry , Base Sequence , Cytoskeleton/chemistry , DNA/biosynthesis , Molecular Sequence Data , Tetracycline/pharmacology , cdc42 GTP-Binding Protein, Saccharomyces cerevisiae/metabolism
15.
Am J Respir Cell Mol Biol ; 31(3): 302-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15191914

ABSTRACT

The physiological role of the duodenal peptide secretin is as a potent stimulant of electrolyte and water movement in pancreatic and biliary epithelium, via activation of G protein-coupled secretin receptors (hSCTR). However, the distribution and potential function of hSCTR in human lung has not previously been addressed. Using real-time quantitative reverse transcriptase-polymerase chain reaction profiling, in situ hybridization, and immunohistochemistry, we demonstrated that the hSCTR is abundantly expressed within the distal regions of human lung (tertiary bronchus and parenchyma), with negligible expression detected in more proximal regions (trachea, primary, and secondary bronchus). Expression was observed predominantly on the basolateral membrane of the bronchial epithelial layer, with some expression also observed in bronchial smooth muscle. In primary cultures of human tertiary bronchial epithelial cells, secretin was demonstrated to potently stimulate channel-mediated Cl- efflux in a concentration-dependent manner. Secretin was also shown to cause concentration-dependent relaxation of human tertiary bronchial smooth muscle. In summary, these data demonstrate that secretin receptors are present in human lung, and that activation of these receptors with human secretin potently stimulates concentration-dependent Cl- efflux from bronchial epithelial cells and bronchorelaxation.


Subject(s)
Lung/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Secretin/metabolism , Bronchi/cytology , Bronchi/metabolism , Bronchodilator Agents/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Chloride Channels/drug effects , Chloride Channels/metabolism , Chlorides/metabolism , Dose-Response Relationship, Drug , Duodenum/metabolism , Humans , Lung/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled , Receptors, Gastrointestinal Hormone/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Secretin/genetics , Secretin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...