Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Infect Dis ; 210(7): 1029-41, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24706936

ABSTRACT

We evaluated the protective effects of fermented dairy products (FDPs) in an infection model, using the mouse pathogen Citrobacter rodentium (CR). Treatment of mice with FDP formulas A, B, and C or a control product did not affect CR colonization, organ specificity, or attaching and effacing lesion formation. Fermented dairy product A (FDP-A), but neither the supernatant from FDP-A nor ß-irradiated (IR) FDP-A, caused a significant reduction in colonic crypt hyperplasia and CR-associated pathology. Profiling the gut microbiota revealed that IR-FDP-A promoted higher levels of phylotypes belonging to Alcaligenaceae and a decrease in Lachnospiraceae (Ruminococcus) during CR infection. Conversely, FDP-A prevented a decrease in Ruminococcus and increased Turicibacteraceae (Turicibacter). Importantly, loss of Ruminococcus and Turicibacter has been associated with susceptibility to dextran sodium sulfate-induced colitis. Our results demonstrate that viable bacteria in FDP-A reduced CR-induced colonic crypt hyperplasia and prevented the loss of key bacterial genera that may contribute to disease pathology.


Subject(s)
Biota , Citrobacter rodentium/growth & development , Colon/pathology , Dairy Products , Diet/methods , Enterobacteriaceae Infections/prevention & control , Hyperplasia/prevention & control , Animals , Colon/microbiology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Female , Hyperplasia/pathology , Mice, Inbred C57BL
2.
Appl Ergon ; 45(2): 188-97, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23820665

ABSTRACT

This contribution to the Ken D. Eason special issue is an illustration of the value of socio-technical analysis applied at an organizational level. We provide a brief historical overview of socio-technical IS research and review studies investigating the impact of IT on organizational structures in the last five decades, identifying a dominating (new) research theme in each decade. A key overall impact of IT in all decades has been a dramatic decrease in transaction costs making it increasingly easier for organizations to source from external providers. A five level taxonomy of sourcing arrangement is developed together with a framework of organizational activities, and a number of significant cases are offered of how organizations are sourcing practically all types of business processes, including innovation. We argue that future IT will further accelerate the movement towards more sourcing, eventually leading to a new type of organization that we call the Ambient organization.


Subject(s)
Information Systems , Models, Organizational , Outsourced Services , Sociology , Technology , Humans , Information Systems/economics , Internet , Organizational Innovation
3.
Trends Microbiol ; 21(8): 430-41, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23870533

ABSTRACT

Injection of effector proteins by a type III secretion system (T3SS) is a common infection strategy employed by many important human pathogens, including enteric Escherichia coli, Salmonella, Yersinia, and Shigella, to subvert cell signaling and host responses. In recent years, great advances have been made in understanding how the T3SS effectors function and execute the diverse infection strategies employed by these pathogens. In this review, we focus on effectors that subvert signaling pathways that impact on endosomal trafficking, cell survival, and innate immunity, particularly phagocytosis, nuclear factor-κB (NF-κB), and mitogen-activated protein (MAP) kinase pathways and the inflammasome.


Subject(s)
Apoptosis , Endosomes/metabolism , Enterobacteriaceae/physiology , Host-Pathogen Interactions , Immunity, Innate , Membrane Transport Proteins/metabolism , Virulence Factors/metabolism , Animals , Enterobacteriaceae/immunology , Humans , Inflammasomes/metabolism , MAP Kinase Signaling System , NF-kappa B/metabolism , Phagocytosis , Protein Transport , Signal Transduction
4.
Hum Gene Ther Methods ; 23(6): 393-407, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23194172

ABSTRACT

Adenoviral vectors deleted of all their viral genes (helper-dependent [HD]) are efficient gene-transfer vehicles. Because transgene expression is rapidly lost in actively dividing cells, we investigated the feasibility of using phage φC31 integrase (φC31-Int) to integrate an HD carrying an attB site and the puromycin resistance gene into human cells (HeLa) and murine myoblasts (C2C12) by co-infection with a second HD-expressing φC31-Int. Because the HD genome is linear, we also investigated whether its circularization, through expression of Cre using a third HD, affects integration. Efficacy and specificity were determined by scoring the number of puromycin-resistant colonies and by sequencing integration sites. Unexpectedly, circularization of HD was unnecessary and it even reduced the integration efficacy. The maximum integration efficacy achieved was 0.5% in HeLa cells and 0.1% in C2C12 myoblasts. Up to 76% of the integration events occurred at pseudo attP sites and previously characterized hotspots were found. A small (two- to three-fold) increase in the number of γ-H2AX positive foci, accompanied by no noticeable change in γ-H2AX expression, indicated the low genotoxicity of φC31-Int. In conclusion, integration of HD mediated by φC31-Int is an attractive alternative to engineer cells, because it permits site-specific integration of large DNA fragments with low genotoxicity.


Subject(s)
Adenoviridae/genetics , Bacteriophages/enzymology , Genetic Vectors/genetics , Integrases/genetics , Animals , Base Sequence , Cell Line , Genetic Loci , Genetic Vectors/metabolism , HEK293 Cells , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Mice , Transduction, Genetic
5.
Cell Microbiol ; 14(7): 1051-70, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22372637

ABSTRACT

Enteropathogenic and enterohaemorrhagic Escherichia coli (EPEC and EHEC) are diarrheagenic pathogens that colonize the gut mucosa via attaching-and-effacing lesion formation. EPEC and EHEC utilize a type III secretion system (T3SS) to translocate effector proteins that subvert host cell signalling to sustain colonization and multiplication. EspH, a T3SS effector that modulates actin dynamics, was implicated in the elongation of the EHEC actin pedestals. In this study we found that EspH is necessary for both efficient pedestal formation and pedestal elongation during EPEC infection. We report that EspH induces actin polymerization at the bacterial attachment sites independently of the Tir tyrosine residues Y474 and Y454, which are implicated in binding Nck and IRSp53/ITRKS respectively. Moreover, EspH promotes recruitment of neural Wiskott-Aldrich syndrome protein (N-WASP) and the Arp2/3 complex to the bacterial attachment site, in a mechanism involving the C-terminus of Tir and the WH1 domain of N-WASP. Dominant negative of WASP-interacting protein (WIP), which binds the N-WASP WH1 domain, diminished EspH-mediated actin polymerization. This study implicates WIP in EPEC-mediated actin polymerization and pedestal elongation and represents the first instance whereby N-WASP is efficiently recruited to the EPEC attachment sites independently of the Tir:Nck and Tir:IRTKS/IRSp53 pathways. Our study reveals the intricacies of Tir and EspH-mediated actin signalling pathways that comprise of distinct, convergent and synergistic signalling cascades.


Subject(s)
Actins/metabolism , Cytoskeletal Proteins/metabolism , Endocytosis , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Bacterial Secretion Systems , Models, Biological , Protein Transport
6.
mBio ; 3(1)2012.
Article in English | MEDLINE | ID: mdl-22251971

ABSTRACT

UNLABELLED: Rho GTPases are important regulators of many cellular processes. Subversion of Rho GTPases is a common infection strategy employed by many important human pathogens. Enteropathogenic Escherichia coli and enterohemorrhagic Escherichia coli (EPEC and EHEC) translocate the effector EspH, which inactivates mammalian Rho guanine exchange factors (GEFs), as well as Map, EspT, and EspM2, which, by mimicking mammalian RhoGEFs, activate Rho GTPases. In this study we found that EspH induces focal adhesion disassembly, triggers cell detachment, activates caspase-3, and induces cytotoxicity. EspH-induced cell detachment and caspase-3 activation can be offset by EspT, EspM2, and the Salmonella Cdc42/Rac1 GEF effector SopE, which remain active in the presence of EspH. EPEC and EHEC therefore use a novel strategy of controlling Rho GTPase activity by translocating one effector to inactivate mammalian RhoGEFs, replacing them with bacterial RhoGEFs. This study also expands the functional range of bacterial RhoGEFs to include cell adhesion and survival. IMPORTANCE: Many human pathogens use a type III secretion system to translocate effectors that can functionally be divided into signaling, disabling, and countervirulence effectors. Among the signaling effectors are those that activate Rho GTPases, which play a central role in coordinating actin dynamics. However, many pathogens also translocate effectors with antagonistic or counteractive functions. For example, Salmonella translocates SopE and SptP, which sequentially turn Rac1 and Cdc42 on and off. In this paper, we show that enteropathogenic E. coli translocates EspH, which inactivates mammalian RhoGEFs and triggers cytotoxicity and at the same time translocates the bacterial RhoGEFs EspM2 and EspT, which are insensitive to EspH, and so neutralizes EspH-induced focal adhesion disassembly, cell detachment, and caspase-3 activation. Our data point to an intriguing infection strategy in which EPEC and EHEC override cellular Rho GTPase signaling by disabling mammalian RhoGEFs and replacing them with with bacterial RhoGEFs that promote cell adhesion and survival.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/pathogenicity , Host-Pathogen Interactions , rho GTP-Binding Proteins/antagonists & inhibitors , Caspase 3/metabolism , Cell Adhesion , Cell Death , HeLa Cells , Humans , Protein Interaction Mapping , Virulence Factors/metabolism
7.
Cell Microbiol ; 13(12): 1881-93, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21848814

ABSTRACT

Enteropathogenic Escherichia coli (EPEC), enterohaemorrhagic E. coli (EHEC) and Citrobacter rodentium colonize their respective hosts while forming attaching and effacing lesions. Their infection strategy relies on translocation of a battery of type III secretion system effectors, including Map, EspM and EspT, which belong to the WxxxE/SopE family of guanine nucleotide exchange factors. Using the C. rodentium mouse model we found that EspT triggers expression of KC and TNFα in vivo. Indeed, a growing body of evidence suggests that, in addition to subversion of actin dynamics, the SopE and the WxxxE effectors activate signalling pathways involved in immune responses. In this study we found that EspT induces expression of the pro-inflammatory mediators cyclooxygenase-2 (COX-2) an enzyme involved in production of prostaglandin E(2) (PGE2), interleukin (Il)-8 and Il-1ß in U937 human macrophages by activating the nuclear factor kappa-B (NF-κB), the extracellular signal-regulated kinases 1 and 2 (Erk1/2) and c-Jun N-terminal kinase (JNK) pathways. Since EspT modulates the activation of Cdc42 and Rac1, which mediates bacterial invasion into epithelial cells, we investigated the involvement of these Rho GTPases and bacterial invasion on pro-inflammatory responses and found that (i) Rac1, but not Cdc42, is involved in EspT-induced Il-8 and Il-1ß secretion and (ii) cytochalasin D inhibits EspT-induced EPEC invasion into U937 but not Il-8 or Il-1ß secretion. These results suggest that while EPEC translocates a number of effectors (i.e. NleC, NleD, NleE, NleH) that inhibit inflammation, a subset of strains, which encode EspT, employ an infection strategy that also involves upregulation of immune mediators.


Subject(s)
Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , JNK Mitogen-Activated Protein Kinases/immunology , NF-kappa B/immunology , Animals , Cyclooxygenase 2/immunology , Cytochalasin D/pharmacology , Dinoprostone/immunology , Disease Models, Animal , Enteropathogenic Escherichia coli/drug effects , Enteropathogenic Escherichia coli/immunology , Fluorescent Antibody Technique , Humans , Inflammation/immunology , Interleukin-1beta/immunology , Interleukin-8/immunology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Protein Transport , U937 Cells , rac1 GTP-Binding Protein/immunology
8.
Biochimie ; 92(6): 583-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20144678

ABSTRACT

Type-IIA secreted phospholipase A(2) (sPLA(2)-IIA) has been proposed to play a role in the development of inflammatory diseases. It has been shown to release arachidonic acid, the precursor of proinflammatory eicosanoids, to hydrolyze phospholipids of pulmonary surfactant, and to bind to specific receptors located on cell surface membranes. However, the most established biological role of sPLA(2)-IIA is related to its potent bactericidal property in particular toward Gram-positive bacteria. This enzyme is present in animal and human biological fluids at concentrations sufficient to kill bacteria. Human recombinant sPLA(2)-IIA is able to kill Gram-positive bacteria at concentrations as low as 1.1 ng/ml. This remarkable property is due to the unique preference of sPLA(2)-IIA for anionic phospholipids such as phosphatidylglycerol, the main phospholipid component of bacterial membranes. Much higher concentrations of sPLA(2)-IIA are required for its action on host cell membranes and surfactant both of which are mainly composed by phosphatidylcholine, a poor substrate for sPLA(2)-IIA. Transgenic mice over-expressing human sPLA(2)-IIA are resistant to infection by Staphylococcus aureus, Escherichia coli, and Bacillus anthracis, the etiological agent of anthrax. Conversely, certain bacteria, such as B. anthracis, E. coli and Bordetella pertussis are able to inhibit sPLA(2)-IIA expression by host cells, thus highlighting a mechanism by which these bacteria can subvert the host immune system. Intranasal instillation of recombinant sPLA(2)-IIA protects mice from mortality caused by pulmonary anthrax. Interestingly, this protective effect was obtained even with B. anthracis strains that down-regulate the expression of endogenous sPLA(2)-IIA, indicating that instilled sPLA(2)-IIA can overcome the subversive action of B. anthracis. We conclude that sPLA(2)-IIA is an efficient endogenous antibiotic of the host and can play a role in host defense against pathogenic bacteria. It can be used as a therapeutic agent in adjunct with current therapy to treat bacteria resistant to multiple antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Group II Phospholipases A2/pharmacology , Animals , Anthrax/immunology , Antigens, Bacterial/immunology , Arachidonic Acid/metabolism , Bacillus anthracis/immunology , Bacillus anthracis/pathogenicity , Bacterial Toxins/antagonists & inhibitors , Drug Resistance, Microbial/immunology , Group II Phospholipases A2/metabolism , Host-Pathogen Interactions , Humans , Mice
9.
Infect Immun ; 78(4): 1417-25, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20123714

ABSTRACT

Subversion of Rho family small GTPases, which control actin dynamics, is a common infection strategy used by bacterial pathogens. In particular, Salmonella enterica serovar Typhimurium, Shigella flexneri, enteropathogenic Escherichia coli (EPEC), and enterohemorrhagic Escherichia coli (EHEC) translocate type III secretion system (T3SS) effector proteins to modulate the Rho GTPases RhoA, Cdc42, and Rac1, which trigger formation of stress fibers, filopodia, and lamellipodia/ruffles, respectively. The Salmonella effector SopE is a guanine nucleotide exchange factor (GEF) that activates Rac1 and Cdc42, which induce "the trigger mechanism of cell entry." Based on a conserved Trp-xxx-Glu motif, the T3SS effector proteins IpgB1 and IpgB2 of Shigella, SifA and SifB of Salmonella, and Map of EPEC and EHEC were grouped together into a WxxxE family; recent studies identified the T3SS EPEC and EHEC effectors EspM and EspT as new family members. Recent structural and functional studies have shown that representatives of the WxxxE effectors share with SopE a 3-D fold and GEF activity. In this minireview, we summarize contemporary findings related to the SopE and WxxxE GEFs in the context of their role in subverting general host cell signaling pathways and infection.


Subject(s)
Bacterial Proteins/metabolism , Enterobacteriaceae/pathogenicity , Guanine Nucleotide Exchange Factors/metabolism , Virulence Factors/metabolism , Actins/metabolism , Amino Acid Sequence , Escherichia coli Proteins/metabolism , Glycoproteins/metabolism , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Multimerization , Protein Structure, Tertiary , rac1 GTP-Binding Protein/metabolism
10.
Biochem Pharmacol ; 79(8): 1149-55, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-19962969

ABSTRACT

Bacillus anthracis, the etiological agent of anthrax, produces lethal toxin (LT) that displays a metallo-proteolytic activity toward the N-terminus of the MAPK-kinases. We have previously shown that secreted type-IIA phospholipase A(2) (sPLA(2)-IIA) exhibits potent anthracidal activity. In vitro expression of sPLA(2)-IIA in guinea pig alveolar macrophages (AMs), the major source of this enzyme in lung tissues, is inhibited by LT. Here, we examined the mechanisms involved in sPLA(2)-IIA inhibition by LT. We first showed that chemical inhibitors of p38 and ERK MAPKs reduced sPLA(2)-IIA expression in AMs indicating that these kinases play a role in sPLA(2)-IIA expression. LT inhibited IL-1beta-induced p38 phosphorylation as well as sPLA(2)-IIA promoter activity in CHO cells. Inhibition of sPLA(2)-IIA promoter activity was mimicked by co-transfection with dominant negative construct of p38 (DN-p38) and reversed by the active form of p38-MAPK (AC-p38). Both LT and DN-p38 decreased IL-1beta-induced NF-kappaB luciferase activity. This contrasted with the effect of AC-p38, which enhanced this activity. However, neither LT nor specific p-38 inhibitor interfered with LPS-induced IkappaBalpha degradation or NF-kappaB nuclear translocation in AMs. Subcutaneous administration of LT to guinea pig before LPS challenge reduced sPLA(2)-IIA levels in broncho-alveolar lavages and ears. We conclude that sPLA(2)-IIA expression is induced via a sequential MAPK-NF-kappaB activation and that LT inhibits this expression likely by interfering with the transactivation of NF-kappaB in the nucleus. This inhibition, which is operating both in vitro and in vivo, may represent a mechanism by which B. anthracis subvert host defense.


Subject(s)
Antigens, Bacterial/pharmacology , Bacterial Toxins/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Group II Phospholipases A2/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Cricetulus , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/physiology , Group II Phospholipases A2/biosynthesis , Guinea Pigs , Male , p38 Mitogen-Activated Protein Kinases/physiology
11.
PLoS Pathog ; 5(12): e1000683, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20011125

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) strains are defined as extracellular pathogens which nucleate actin rich pedestal-like membrane extensions on intestinal enterocytes to which they intimately adhere. EPEC infection is mediated by type III secretion system effectors, which modulate host cell signaling. Recently we have shown that the WxxxE effector EspT activates Rac1 and Cdc42 leading to formation of membrane ruffles and lamellipodia. Here we report that EspT-induced membrane ruffles facilitate EPEC invasion into non-phagocytic cells in a process involving Rac1 and Wave2. Internalized EPEC resides within a vacuole and Tir is localized to the vacuolar membrane, resulting in actin polymerization and formation of intracellular pedestals. To the best of our knowledge this is the first time a pathogen has been shown to induce formation of actin comets across a vacuole membrane. Moreover, our data breaks the dogma of EPEC as an extracellular pathogen and defines a new category of invasive EPEC.


Subject(s)
Actins/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Signal Transduction/physiology , 3T3 Cells , Animals , Caco-2 Cells , Cell Membrane , Enteropathogenic Escherichia coli/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Mice , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Pseudopodia/metabolism , Transfection , Vacuoles/microbiology , Virulence , Wiskott-Aldrich Syndrome Protein Family/metabolism , rac1 GTP-Binding Protein/metabolism
12.
PLoS Pathog ; 5(4): e1000359, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19343203

ABSTRACT

Lethal toxin (LT) is a critical virulence factor of Bacillus anthracis, the etiological agent of anthrax, whose pulmonary form is fatal in the absence of treatment. Inflammatory response is a key process of host defense against invading pathogens. We report here that intranasal instillation of a B. anthracis strain bearing inactive LT stimulates cytokine production and polymorphonuclear (PMN) neutrophils recruitment in lungs. These responses are repressed by a prior instillation of an LT preparation. In contrast, instillation of a B. anthracis strain expressing active LT represses lung inflammation. The inhibitory effects of LT on cytokine production are also observed in vitro using mouse and human pulmonary epithelial cells. These effects are associated with an alteration of ERK and p38-MAPK phosphorylation, but not JNK phosphorylation. We demonstrate that although NF-kappaB is essential for IL-8 expression, LT downregulates this expression without interfering with NF-kappaB activation in epithelial cells. Histone modifications are known to induce chromatin remodelling, thereby enhancing NF-kappaB binding on promoters of a subset of genes involved in immune response. We show that LT selectively prevents histone H3 phosphorylation at Ser 10 and recruitment of the p65 subunit of NF-kappaB at the IL-8 and KC promoters. Our results suggest that B. anthracis represses the immune response, in part by altering chromatin accessibility of IL-8 promoter to NF-kappaB in epithelial cells. This epigenetic reprogramming, in addition to previously reported effects of LT, may represent an efficient strategy used by B. anthracis for invading the host.


Subject(s)
Antigens, Bacterial/toxicity , Bacterial Toxins/toxicity , Gene Expression/drug effects , Histones/metabolism , Interleukin-8/metabolism , Lung/metabolism , Respiratory Mucosa/metabolism , Animals , Antigens, Bacterial/physiology , Chromatin/metabolism , Cytokines/genetics , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-8/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Pneumonia , Promoter Regions, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Microb Pathog ; 45(1): 30-7, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18486438

ABSTRACT

This report addressed the question whether ExoU stimulation of airway epithelial cells may contribute to the inflammatory response detected in the course of Pseudomonas aeruginosa respiratory infections. Infection with PA103 P. aeruginosa elicited a potent release of IL-6 and IL-8, as well as of arachidonic acid (AA) and PGE(2) that was reduced by the bacterial treatment with MAFP, a cPLA(2) inhibitor. Airway cells from the BEAS-2B line and in primary culture were shown to be enriched in lipid bodies (LBs), that are cytoplasmic domains implicated in AA transformation into eicosanoids. However, cells infected with PA103 and with a mutant deficient in exoU but complemented with a functional gene exhibited reduced contents of LBs, and this reduction was inhibited by MAFP. FACS analysis showed that the decrease in the LB content correlated with the presence of intracellular PGE(2). Also, in PA103-infected cells, PGE(2) was immunolocalized in LBs, suggesting that the reduction in the cell content of the organelles was due to consumption of their glycerolipids, resulting in local synthesis of the prostanoid. In conclusion, we showed the ExoU ability to induce airway epithelial cells to overproduce PGE(2) and we speculate that LB may represent intracellular loci involved in ExoU-induced eicosanoid synthesis.


Subject(s)
Bacterial Proteins/metabolism , Bronchi/metabolism , Epithelial Cells/metabolism , Inclusion Bodies/metabolism , Inflammation Mediators/metabolism , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Arachidonic Acid/metabolism , Arachidonic Acids/pharmacology , Bacterial Proteins/genetics , Bronchi/cytology , Bronchi/drug effects , Bronchi/microbiology , Cell Line , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Humans , Lipid Metabolism , Organophosphonates/pharmacology , Prostaglandins E/metabolism , Pseudomonas aeruginosa/drug effects
14.
PLoS Pathog ; 3(12): e187, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18069891

ABSTRACT

Bacillus anthracis, the etiological agent of anthrax, is a spore-forming gram-positive bacterium. Infection with this pathogen results in multisystem dysfunction and death. The pathogenicity of B. anthracis is due to the production of virulence factors, including edema toxin (ET). Recently, we established the protective role of type-IIA secreted phospholipase A2 (sPLA2-IIA) against B. anthracis. A component of innate immunity produced by alveolar macrophages (AMs), sPLA2-IIA is found in human and animal bronchoalveolar lavages at sufficient levels to kill B. anthracis. However, pulmonary anthrax is almost always fatal, suggesting the potential impairment of sPLA2-IIA synthesis and/or action by B. anthracis factors. We investigated the effect of purified ET and ET-deficient B. anthracis strains on sPLA2-IIA expression in primary guinea pig AMs. We report that ET inhibits sPLA2-IIA expression in AMs at the transcriptional level via a cAMP/protein kinase A-dependent process. Moreover, we show that live B. anthracis strains expressing functional ET inhibit sPLA2-IIA expression, whereas ET-deficient strains induced this expression. This stimulatory effect, mediated partly by the cell wall peptidoglycan, can be counterbalanced by ET. We conclude that B. anthracis down-regulates sPLA2-IIA expression in AMs through a process involving ET. Our study, therefore, describes a new molecular mechanism implemented by B. anthracis to escape innate host defense. These pioneering data will provide new molecular targets for future intervention against this deadly pathogen.


Subject(s)
Anthrax/prevention & control , Antigens, Bacterial/toxicity , Bacillus anthracis/enzymology , Bacterial Toxins/toxicity , Group II Phospholipases A2/metabolism , Macrophages, Alveolar/drug effects , Animals , Anthrax/enzymology , Anthrax/immunology , Bronchoalveolar Lavage , Cells, Cultured , Down-Regulation , Gene Expression Regulation, Bacterial , Gene Silencing , Group II Phospholipases A2/immunology , Guinea Pigs , Host-Pathogen Interactions , Immunity, Innate , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/microbiology , Male , RNA, Messenger/metabolism
15.
Microbes Infect ; 8(2): 450-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16293434

ABSTRACT

ExoU PLA2-like activity has been shown to account for membrane lysis and acute death of infected cells. Translocation of effector proteins by the type III secretion systems depends on close contact between microbial and host cells. Our finding that both the ExoU-producing PA103 Pseudomonas aeruginosa and its mutant obtained by deletion of exoU adhered poorly to endothelial cells (EC) led to the hypothesis that, in some cells, the amount of injected toxin may not be enough to induce cell lysis but cells would suffer from a long-term effect of ExoU intoxication. To address this question, cells were exposed to both bacteria for 1 h and then treated with gentamicin-containing medium, to eliminate infecting microorganisms. After 24 h, the percentage of viable EC in PA103-infected cultures was significantly lower than in cultures exposed to the mutant, as determined by the MTT assay. Cell death was not likely to depend on the ExoU lytic activity since cell labeling with propidium iodide was similar in cultures infected with both bacterial strains. Bacterial cytotoxicity was significantly reduced by MAFP, a specific inhibitor of cPLA2 and iPLA2. Since the PLA2 activity on membrane phospholipids generates free fatty acid, including arachidonic acid (AA), we next compared the bacterial ability to release AA from infected EC. PA103 was shown to induce a potent AA release that was inhibited by MAFP. AA oxidation by oxygenases generates eicosanoids, known to induce both cell death and proliferation. However neither inhibitors of cyclooxygenases (ibuprofen) nor lipoxygenases (NDGA) reduced the ExoU toxicity. Since non-enzymatic oxidation of AA generates reactive radicals, we next investigated the PA103 ability to induce oxidative stress in infected cells. FACS analysis of cell labeling with the C-11 fluor probe and with anti-4-hydroxynonel antibody revealed a significant peroxidation of cell membrane lipids. These results, together with our finding that PA103-infected EC death was significantly attenuated by alpha-tocopherol, led to the conclusion that AA-induced oxidative stress may be another mechanism of cell damage in the course of infection by ExoU-producing P. aeruginosa.


Subject(s)
Arachidonic Acid/metabolism , Bacterial Proteins/metabolism , Endothelial Cells/microbiology , Oxidative Stress , Pseudomonas aeruginosa/pathogenicity , Bacterial Proteins/genetics , Cell Death , Cell Line, Transformed , Dermis/blood supply , Dermis/cytology , Endothelial Cells/pathology , Endothelium, Vascular/cytology , Humans , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 289(5): L816-24, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15964894

ABSTRACT

Cystic fibrosis (CF) is characterized by an exacerbated inflammatory pulmonary response with excessive production of inflammatory mediators. We investigated here the impact of cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction on prostaglandin E2 (PGE2) production and type IIA secreted phospholipase A2 (sPLA2-IIA) expression. We show that both resting and LPS-stimulated human respiratory epithelial cell line bearing DeltaF508 mutation on CFTR (CF cells) released more PGE2 than control cell line. This was accompanied by enhanced expression and activity of cyclooxygenase-2 in CF cells. PGE2 release was attenuated after experimentally induced retrafficking of the DeltaF508-CFTR at the plasma membrane. sPLA2-IIA expression occurred at higher levels in CF cells than in control cells and was enhanced by LPS and PGE2. Suppression of PGE2 synthesis by aspirin led to an inhibition of LPS-induced sPLA2-IIA expression. Higher activation of NF-kappaB was observed in CF cells compared with control cells and was enhanced by LPS. However, addition of PGE2 or aspirin had no effect on NF-kappaB activation. LPS-induced sPLA2-IIA expression was reduced by an NF-kappaB inhibitor. We suggest that the lack of the CFTR in the plasma membrane results in a PGE2 overproduction and an enhanced sPLA2-IIA expression. This expression is upregulated by NF-kappaB and amplified by PGE2 via a unidentified signaling pathway.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dinoprostone/biosynthesis , Lung/metabolism , Phospholipases A/genetics , Base Sequence , Cell Line , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , DNA/genetics , Epithelial Cells/metabolism , Gene Expression , Group II Phospholipases A2 , Humans , Lung/pathology , Phospholipases A2 , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Deletion , Trachea/metabolism , Trachea/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...