Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 12: 1064010, 2022.
Article in English | MEDLINE | ID: mdl-36519135

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that can cause critical cellular damage and subvert the immune response to promote its survival. Among the numerous virulence factors of P. aeruginosa, the type III secretion system (T3SS) is involved in host cell pathogenicity. Using a needle-like structure, T3SS detects eukaryotic cells and injects toxins directly into their cytosol, thus highlighting its ability to interfere with the host immune response. In this mini-review, we discuss how the T3SS and bacterial effectors secreted by this pathway not only activate the immune response but can also manipulate it to promote the establishment of P. aeruginosa infections.


Subject(s)
Pseudomonas Infections , Type III Secretion Systems , Humans , Type III Secretion Systems/metabolism , Pseudomonas aeruginosa , Bacterial Proteins/metabolism , Virulence Factors/metabolism , Immunity , Pseudomonas Infections/microbiology
2.
Front Immunol ; 13: 931027, 2022.
Article in English | MEDLINE | ID: mdl-35860265

ABSTRACT

The opportunistic pathogen Pseudomonas aeruginosa is one of the most common agents of respiratory infections and has been associated with high morbidity and mortality rates. The ability of P. aeruginosa to cause severe respiratory infections results from the coordinated action of a variety of virulence factors that promote bacterial persistence in the lungs. Several of these P. aeruginosa virulence mechanisms are mediated by bacterial lipids, mainly lipopolysaccharide, rhamnolipid, and outer membrane vesicles. Other mechanisms arise from the activity of P. aeruginosa enzymes, particularly ExoU, phospholipase C, and lipoxygenase A, which modulate host lipid signaling pathways. Moreover, host phospholipases, such as cPLA2α and sPLA2, are also activated during the infectious process and play important roles in P. aeruginosa pathogenesis. These mechanisms affect key points of the P. aeruginosa-host interaction, such as: i) biofilm formation that contributes to bacterial colonization and survival, ii) invasion of tissue barriers that allows bacterial dissemination, iii) modulation of inflammatory responses, and iv) escape from host defenses. In this mini-review, we present the lipid-based mechanism that interferes with the establishment of P. aeruginosa in the lungs and discuss how bacterial and host lipids can impact the outcome of P. aeruginosa respiratory infections.


Subject(s)
Pseudomonas Infections , Respiratory Tract Infections , Humans , Lipids , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Respiratory Tract Infections/complications , Virulence Factors
3.
Braz J Microbiol ; 53(2): 583-594, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35169995

ABSTRACT

Corynebacterium diphtheriae, the leading causing agent of diphtheria, has been increasingly related to invasive diseases, including sepsis, endocarditis, pneumonia, and osteomyelitis. Oxidative stress defense is required not only for successful growth and survival under environmental conditions but also in the regulation of virulence mechanisms of human pathogenic species, by promoting mucosal colonization, survival, dissemination, and defense against the innate immune system. OxyR, functioning as a negative and/or positive transcriptional regulator, has been included among the major bacterial coordinators of antioxidant response. OxyR was first reported as a repressor of catalase expression in C. diphtheriae. However, the involvement of OxyR in C. diphtheriae pathogenesis remains unclear. Accordingly, this work aimed to investigate the role of OxyR in mechanisms of host-pathogen interaction of C. diphtheriae through the disruption of the OxyR of the diphtheria toxin (DT)-producing C. diphtheriae CDC-E8392 strain. The effects of OxyR gene disruption were analyzed through interaction assays with human epithelial cell lines (HEp-2 and pneumocytes A549) and by the induction of experimental infections in Caenorhabditis elegans nematodes and Swiss Webster mice. The OxyR disruption exerted influence on NO production and mechanism accountable for the expression of the aggregative-adherence pattern (AA) expressed by CDC-E8392 strain on human epithelial HEp-2 cells. Moreover, invasive potential and intracytoplasmic survival within HEp-2 cells, as well as the arthritogenic potential in mice, were found affected by the OxyR disruption. In conclusion, data suggest that OxyR is implicated in mechanisms of host-pathogen interaction of C. diphtheriae.


Subject(s)
Corynebacterium diphtheriae , Diphtheria , Endocarditis , Animals , Corynebacterium diphtheriae/genetics , Diphtheria/microbiology , Endocarditis/microbiology , Host-Pathogen Interactions , Mice , Virulence
4.
Mem Inst Oswaldo Cruz ; 113(6): e140421, 2018.
Article in English | MEDLINE | ID: mdl-29641644

ABSTRACT

BACKGROUND Streptococcus agalactiae can causes sepsis, pneumonia, and meningitis in neonates, the elderly, and immunocompromised patients. Although the virulence properties of S. agalactiae have been partially elucidated, the molecular mechanisms related to reactive oxygen species (ROS) generation in infected human endothelial cells need further investigation. OBJECTIVES This study aimed to evaluate the influence of oxidative stress in human umbilical vein endothelial cells (HUVECs) during S. agalactiae infection. METHODS ROS production during S. agalactiae-HUVEC infection was detected using the probe CM-H2DCFDA. Microfilaments labelled with phalloidin-FITC and p47phox-Alexa 546 conjugated were analysed by immunofluorescence. mRNA levels of p47phox (NADPH oxidase subunit) were assessed using Real Time qRT-PCR. The adherence and intracellular viability of S. agalactiae in HUVECs with or without pre-treatment of DPI, apocynin (NADPH oxidase inhibitors), and LY294002 (PI3K inhibitor) were evaluated by penicillin/gentamicin exclusion. Phosphorylation of p47phox and Akt activation by S. agalactiae were evaluated by immunoblotting analysis. FINDINGS Data showed increased ROS production 15 min after HUVEC infection. Real-Time qRT-PCR and western blotting performed in HUVEC infected with S. agalactiae detected alterations in mRNA levels and activation of p47phox. Pre-treatment of endothelial cells with NADPH oxidase (DPI and apocynin) and PI3K/Akt pathway (LY294002) inhibitors reduced ROS production, bacterial intracellular viability, and generation of actin stress fibres in HUVECs infected with S. agalactiae. CONCLUSIONS ROS generation via the NADPH oxidase pathway contributes to invasion of S. agalactiae in human endothelial cells accompanied by cytoskeletal reorganisation through the PI3K/Akt pathway, which provides novel evidence for the involvement of oxidative stress in S. agalactiae pathogenesis.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , NADPH Oxidases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Streptococcal Infections/metabolism , Streptococcus agalactiae/metabolism , Analysis of Variance , Bacterial Adhesion , Blotting, Western , Human Umbilical Vein Endothelial Cells/microbiology , Humans , NADPH Oxidases/analysis , Oxidative Stress/physiology , Reactive Oxygen Species/analysis , Real-Time Polymerase Chain Reaction , Reference Values , Signal Transduction/physiology , Time Factors
5.
Mem. Inst. Oswaldo Cruz ; 113(6): e140421, 2018. graf
Article in English | LILACS | ID: biblio-894933

ABSTRACT

BACKGROUND Streptococcus agalactiae can causes sepsis, pneumonia, and meningitis in neonates, the elderly, and immunocompromised patients. Although the virulence properties of S. agalactiae have been partially elucidated, the molecular mechanisms related to reactive oxygen species (ROS) generation in infected human endothelial cells need further investigation. OBJECTIVES This study aimed to evaluate the influence of oxidative stress in human umbilical vein endothelial cells (HUVECs) during S. agalactiae infection. METHODS ROS production during S. agalactiae-HUVEC infection was detected using the probe CM-H2DCFDA. Microfilaments labelled with phalloidin-FITC and p47phox-Alexa 546 conjugated were analysed by immunofluorescence. mRNA levels of p47phox (NADPH oxidase subunit) were assessed using Real Time qRT-PCR. The adherence and intracellular viability of S. agalactiae in HUVECs with or without pre-treatment of DPI, apocynin (NADPH oxidase inhibitors), and LY294002 (PI3K inhibitor) were evaluated by penicillin/gentamicin exclusion. Phosphorylation of p47phox and Akt activation by S. agalactiae were evaluated by immunoblotting analysis. FINDINGS Data showed increased ROS production 15 min after HUVEC infection. Real-Time qRT-PCR and western blotting performed in HUVEC infected with S. agalactiae detected alterations in mRNA levels and activation of p47phox. Pre-treatment of endothelial cells with NADPH oxidase (DPI and apocynin) and PI3K/Akt pathway (LY294002) inhibitors reduced ROS production, bacterial intracellular viability, and generation of actin stress fibres in HUVECs infected with S. agalactiae. CONCLUSIONS ROS generation via the NADPH oxidase pathway contributes to invasion of S. agalactiae in human endothelial cells accompanied by cytoskeletal reorganisation through the PI3K/Akt pathway, which provides novel evidence for the involvement of oxidative stress in S. agalactiae pathogenesis.


Subject(s)
Humans , Reactive Oxygen Species/analysis , NADPH Oxidases/analysis , NADPH Oxidases/metabolism , Human Umbilical Vein Endothelial Cells/microbiology , Signal Transduction/physiology , Real-Time Polymerase Chain Reaction
6.
Front Immunol ; 8: 1127, 2017.
Article in English | MEDLINE | ID: mdl-28959260

ABSTRACT

Leishmania parasites infect macrophages, causing a wide spectrum of human diseases, from cutaneous to visceral forms. In search of novel therapeutic targets, we performed comprehensive in vitro and ex vivo mapping of the signaling pathways upstream and downstream of antioxidant transcription factor [nuclear factor erythroid 2-related factor 2 (Nrf2)] in cutaneous leishmaniasis (CL), by combining functional assays in human and murine macrophages with a systems biology analysis of in situ (skin biopsies) CL patient samples. First, we show the PKR pathway controls the expression and activation of Nrf2 in Leishmania amazonensis infection in vitro. Nrf2 activation also required PI3K/Akt signaling and autophagy mechanisms. Nrf2- or PKR/Akt-deficient macrophages exhibited increased levels of ROS/RNS and reduced expression of Sod1 Nrf2-dependent gene and reduced parasite load. L. amazonensis counteracted the Nrf2 inhibitor Keap1 through the upregulation of p62 via PKR. This Nrf2/Keap1 observation was confirmed in situ in skin biopsies from Leishmania-infected patients. Next, we explored the ex vivo transcriptome in CL patients, as compared to healthy controls. We found the antioxidant response element/Nrf2 signaling pathway was significantly upregulated in CL, including downstream target p62. In silico enrichment analysis confirmed upstream signaling by interferon and PI3K/Akt, and validated our in vitro findings. Our integrated in vitro, ex vivo, and in silico approach establish Nrf2 as a central player in human cutaneous leishmaniasis and reveal Nrf2/PKR crosstalk and PI3K/Akt pathways as potential therapeutic targets.

7.
Sci Rep ; 5: 16777, 2015 Nov 26.
Article in English | MEDLINE | ID: mdl-26608746

ABSTRACT

HIV-1 co-infection with human parasitic diseases is a growing public health problem worldwide. Leishmania parasites infect and replicate inside macrophages, thereby subverting host signaling pathways, including the response mediated by PKR. The HIV-1 Tat protein interacts with PKR and plays a pivotal role in HIV-1 replication. This study shows that Tat increases both the expression and activation of PKR in Leishmania-infected macrophages. Importantly, the positive effect of Tat addition on parasite growth was dependent on PKR signaling, as demonstrated in PKR-deficient macrophages or macrophages treated with the PKR inhibitor. The effect of HIV-1 Tat on parasite growth was prevented when the supernatant of HIV-1-infected macrophages was treated with neutralizing anti-HIV-1 Tat prior to Leishmania infection. The addition of HIV-1 Tat to Leishmania-infected macrophages led to inhibition of iNOS expression, modulation of NF-kB activation and enhancement of IL-10 expression. Accordingly, the expression of a Tat construct containing mutations in the basic region (49-57aa), which is responsible for the interaction with PKR, favored neither parasite growth nor IL-10 expression in infected macrophages. In summary, we show that Tat enhances Leishmania growth through PKR signaling.


Subject(s)
HIV-1/metabolism , Leishmania/growth & development , RNA, Double-Stranded/metabolism , eIF-2 Kinase/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Cell Line , Enzyme Activation , Humans , Interleukin-10/metabolism , Intracellular Space/parasitology , Leishmania/metabolism , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Leishmaniasis/pathology , Macrophages/enzymology , Macrophages/parasitology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Protein Structure, Tertiary , Signal Transduction , tat Gene Products, Human Immunodeficiency Virus/chemistry
8.
Open Biol ; 5(9): 150118, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26400473

ABSTRACT

Leishmania amazonensis activates the NF-κB transcriptional repressor homodimer (p50/p50) and promotes nitric oxide synthase (iNOS) downregulation. We investigated the role of PI3K/Akt in p50/p50 NF-κB activation and the effect on iNOS expression in L. amazonensis infection. The increased occupancy of p50/p50 on the iNOS promoter of infected macrophages was observed and we demonstrated that both p50/p50 NF-κB induction and iNOS downregulation in infected macrophages depended on PI3K/Akt activation. Importantly, the intracellular growth of the parasite was also impaired during PI3K/Akt signalling inhibition and in macrophages knocked-down for Akt 1 expression. It was also observed that the increased nuclear levels of p50/p50 in L. amazonensis-infected macrophages were associated with reduced phosphorylation of 907 Ser p105, the precursor of p50. Corroborating these data, we demonstrated the increased levels of phospho-9 Ser GSK3ß in infected macrophages, which is associated with GSK3ß inhibition and, consequently, its inability to phosphorylate p105. Remarkably, we found that the levels of pPTEN 370 Ser, a negative regulator of PI3K, increased due to L. amazonensis infection. Our data support the notion that PI3K/Akt activity is sustained during the parasite infection, leading to NF-κB 105 phosphorylation and further processing to originate p50/p50 homodimers and the consequent downregulation of iNOS expression.


Subject(s)
Leishmania/physiology , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Dimerization , Down-Regulation , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Leishmania/genetics , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Leishmaniasis/pathology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/chemistry , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/metabolism , Signal Transduction
9.
Pathog Dis ; 73(7)2015 Oct.
Article in English | MEDLINE | ID: mdl-26187894

ABSTRACT

Intravital microscopy was used to assess the involvement of ExoU, a Pseudomonas aeruginosa cytotoxin with phospholipase A2 activity, in dysfunction of cerebral microcirculation during experimental pneumosepsis. Cortical vessels from mice intratracheally infected with low density of the ExoU-producing PA103 P. aeruginosa strain exhibited increased leukocyte rolling and adhesion to venule endothelium, decreased capillar density and impaired arteriolar response to vasoactive acetylcholine. These phenomena were mediated by the platelet activating factor receptor (PAFR) pathway because they were reversed in mice treated with a PAFR antagonist prior to infection. Brains from PA103-infected animals exhibited a perivascular inflammatory infiltration that was not detected in animals infected with an exoU deficient mutant or in mice treated with the PAFR antagonist and infected with the wild type bacteria. No effect on brain capillary density was detected in mice infected with the PAO1 P. aeruginosa strain, which do not produce ExoU. Finally, after PA103 infection, mice with a targeted deletion of the PAFR gene exhibited higher brain capillary density and lower leukocyte adhesion to venule endothelium, as well as lower increase of systemic inflammatory cytokines, when compared to wild-type mice. Altogether, our results establish a role for PAFR in mediating ExoU-induced cerebral microvascular failure in a murine model of sepsis.


Subject(s)
Bacterial Proteins/metabolism , Brain/pathology , Microcirculation/physiology , Platelet Activating Factor/metabolism , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/metabolism , Sepsis/pathology , Animals , Cell Adhesion , Cytokines/analysis , Female , Intravital Microscopy , Leukocytes/immunology , Mice , Platelet Membrane Glycoproteins/metabolism , Pseudomonas aeruginosa/growth & development , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
10.
Med Microbiol Immunol ; 204(6): 673-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25904542

ABSTRACT

ExoU is a potent proinflammatory toxin produced by Pseudomonas aeruginosa, a major agent of severe lung infection and sepsis. Because inflammation is usually associated with oxidative stress, we investigated the effect of ExoU on free radical production and antioxidant defense mechanisms during the course of P. aeruginosa infection. In an experimental model of acute pneumonia, ExoU accounted for increased lipid peroxidation in mice lungs as soon as 3 h after intratracheal instillation of PA103 P. aeruginosa strain. The contribution of airway cells to the generation of a redox imbalance was assessed by in vitro tests carried out with A549 airway epithelial cells. Cultures infected with the ExoU-producing PA103 P. aeruginosa strain produced significantly increased concentrations of lipid hydroperoxides, 8-isoprostane, reactive oxygen intermediates, peroxynitrite and nitric oxide (NO), when compared to cells infected with exoU-deficient mutants. Overproduction of NO by PA103-infected cells likely resulted from overexpression of both inducible and endothelial NO synthase isoforms. PA103 infection was also associated with a significantly increased activity of superoxide dismutase (SOD) and decreased levels of reduced glutathione (GSH), a major antioxidant compound. Our findings unveil another potential mechanism of tissue damage during infection by ExoU-producing P. aeruginosa strains.


Subject(s)
Bacterial Proteins/metabolism , Oxidation-Reduction , Oxidative Stress , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pseudomonas aeruginosa/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Sepsis , Animals , Antioxidants/metabolism , Catalase/metabolism , Cell Line , Disease Models, Animal , Female , Lipid Peroxidation , Mice , Superoxide Dismutase/metabolism
11.
Cell Microbiol ; 16(8): 1244-54, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24612488

ABSTRACT

ExoU is an important virulence factor in acute Pseudomonas aeruginosa infections. Here, we unveiled the mechanisms of ExoU-driven NF-κB activation by using human airway cells and mice infected with P. aeruginosa strains. Several approaches showed that PAFR was crucially implicated in the activation of the canonical NF-κB pathway. Confocal microscopy of lungs from infected mice revealed that PAFR-dependent NF-κB activation occurred mainly in respiratory epithelial cells, and reduced p65 nuclear translocation was detected in mice PAFR-/- or treated with the PAFR antagonist WEB 2086. Several evidences showed that ExoU-induced NF-κB activation regulated PAFR expression. First, ExoU increased p65 occupation of PAFR promoter, as assessed by ChIP. Second, luciferase assays in cultures transfected with different plasmid constructs revealed that ExoU promoted p65 binding to the three κB sites in PAFR promoter. Third, treatment of cell cultures with the NF-κB inhibitor Bay 11-7082, or transfection with IκBα negative-dominant, significantly decreased PAFR mRNA. Finally, reduction in PAFR expression was observed in mice treated with Bay 11-7082 or WEB 2086 prior to infection. Together, our data demonstrate that ExoU activates NF-κB by PAFR signalling, which in turns enhances PAFR expression, highlighting an important mechanism of amplification of response to this P. aeruginosa toxin.


Subject(s)
Bacterial Proteins/metabolism , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/genetics , Pseudomonas aeruginosa/pathogenicity , Receptors, G-Protein-Coupled/genetics , Transcription Factor RelA/metabolism , Animals , Azepines/pharmacology , Bacterial Toxins/metabolism , Cell Line , Enzyme Activation , Female , Gene Expression Regulation , Humans , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Platelet Activating Factor/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/biosynthesis , Promoter Regions, Genetic , Protein Binding , Pseudomonas Infections/pathology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/biosynthesis , Signal Transduction/genetics , Triazoles/pharmacology
12.
Mem Inst Oswaldo Cruz ; 107(6): 728-34, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22990960

ABSTRACT

An increased plasma concentration of von Willebrand factor (vWF) is detected in individuals with many infectious diseases and is accepted as a marker of endothelium activation and prothrombotic condition. To determine whether ExoU, a Pseudomonas aeruginosa cytotoxin with proinflammatory activity, enhances the release of vWF, microvascular endothelial cells were infected with the ExoU-producing PA103 P. aeruginosa strain or an exoU-deficient mutant. Significantly increased vWF concentrations were detected in conditioned medium and subendothelial extracellular matrix from cultures infected with the wild-type bacteria, as determined by enzyme-linked immunoassays. PA103-infected cells also released higher concentrations of procoagulant microparticles containing increased amounts of membrane-associated vWF, as determined by flow cytometric analyses of cell culture supernatants. Both flow cytometry and confocal microscopy showed that increased amounts of vWF were associated with cytoplasmic membranes from cells infected with the ExoU-producing bacteria. PA103-infected cultures exposed to platelet suspensions exhibited increased percentages of cells with platelet adhesion. Because no modulation of the vWF mRNA levels was detected by reverse transcription-polymerase chain reaction assays in PA103-infected cells, ExoU is likely to have induced the release of vWF from cytoplasmic stores rather than vWF gene transcription. Such release is likely to modify the thromboresistance of microvascular endothelial cells.


Subject(s)
Bacterial Proteins/metabolism , Endothelial Cells/microbiology , Endothelium, Vascular/microbiology , Pseudomonas aeruginosa/metabolism , von Willebrand Factor/metabolism , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Platelet Adhesiveness
13.
Mem. Inst. Oswaldo Cruz ; 107(6): 728-734, set. 2012. ilus
Article in English | LILACS | ID: lil-649486

ABSTRACT

An increased plasma concentration of von Willebrand factor (vWF) is detected in individuals with many infectious diseases and is accepted as a marker of endothelium activation and prothrombotic condition. To determine whether ExoU, a Pseudomonas aeruginosa cytotoxin with proinflammatory activity, enhances the release of vWF, microvascular endothelial cells were infected with the ExoU-producing PA103 P. aeruginosa strain or an exoU-deficient mutant. Significantly increased vWF concentrations were detected in conditioned medium and subendothelial extracellular matrix from cultures infected with the wild-type bacteria, as determined by enzyme-linked immunoassays. PA103-infected cells also released higher concentrations of procoagulant microparticles containing increased amounts of membrane-associated vWF, as determined by flow cytometric analyses of cell culture supernatants. Both flow cytometry and confocal microscopy showed that increased amounts of vWF were associated with cytoplasmic membranes from cells infected with the ExoU-producing bacteria. PA103-infected cultures exposed to platelet suspensions exhibited increased percentages of cells with platelet adhesion. Because no modulation of the vWF mRNA levels was detected by reverse transcription-polymerase chain reaction assays in PA103-infected cells, ExoU is likely to have induced the release of vWF from cytoplasmic stores rather than vWF gene transcription. Such release is likely to modify the thromboresistance of microvascular endothelial cells.


Subject(s)
Humans , Bacterial Proteins/metabolism , Endothelial Cells/microbiology , Endothelium, Vascular/microbiology , Pseudomonas aeruginosa/metabolism , von Willebrand Factor/metabolism , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Platelet Adhesiveness
14.
PLoS One ; 7(7): e41772, 2012.
Article in English | MEDLINE | ID: mdl-22848596

ABSTRACT

ExoU, a Pseudomonas aeruginosa cytotoxin injected into host cytosol by type III secretion system, exhibits a potent proinflammatory activity that leads to a marked recruitment of neutrophils to infected tissues. To evaluate the mechanisms that account for neutrophil infiltration, we investigated the effect of ExoU on IL-8 secretion and NF-κB activation. We demonstrate that ExoU increases IL-8 mRNA and protein levels in P. aeruginosa-infected epithelial and endothelial cell lines. Also, ExoU induces the nuclear translocation of p65/p50 NF-κB transactivator heterodimer as well as NF-κB-dependent transcriptional activity. ChIP assays clearly revealed that ExoU promotes p65 binding to NF-κB site in IL-8 promoter and the treatment of cultures with the NF-κB inhibitor Bay 11-7082 led to a significant reduction in IL-8 mRNA levels and protein secretion induced by ExoU. These results were corroborated in a murine model of pneumonia that revealed a significant reduction in KC secretion and neutrophil infiltration in bronchoalveolar lavage when mice were treated with Bay 11-7082 before infection with an ExoU-producing strain. In conclusion, our data demonstrate that ExoU activates NF-κB, stimulating IL-8 expression and secretion during P. aeruginosa infection, and unveils a new mechanism triggered by this important virulence factor to interfere in host signaling pathways.


Subject(s)
Bacterial Proteins/metabolism , Interleukin-8/metabolism , NF-kappa B/metabolism , Pseudomonas aeruginosa/physiology , Animals , Bacterial Proteins/biosynthesis , Bronchoalveolar Lavage Fluid/microbiology , Capillaries/cytology , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Gene Expression Regulation , Interleukin-8/genetics , Mice , Neutrophil Infiltration , Pseudomonas aeruginosa/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory System/immunology , Respiratory System/microbiology
15.
Microbes Infect ; 12(2): 154-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19931407

ABSTRACT

ExoU, a Pseudomonas aeruginosa cytotoxin injected via the type III secretion system into host cells, possesses eicosanoid-mediated proinflammatory properties due to its phospholipase A(2) (PLA(2)) activity. This report addressed the question whether ExoU may modulate the expression of adhesion molecules in host cells, therefore contributing to the recruitment of leukocyte into infected tissues. ExoU was shown to down-regulate membrane-bound ICAM-1 (mICAM-1) and up-regulate the release of soluble ICAM-1 (sICAM-1) from P. aeruginosa-infected endothelial cells. The modulation of ICAM-1 depended on the direct effect of the ExoU PLA(2) activity and involved the cyclooxygenase (COX) pathway. No differences in mICAM-1 and sICAM-1 mRNA levels were observed when cultures were infected with the ExoU-producing PA103 strain or the mutant PA103DeltaexoU, suggesting that ExoU may proteolytically cleave mICAM-1, producing sICAM-1 in a COX-dependent pathway.


Subject(s)
Bacterial Proteins/metabolism , Endothelial Cells/microbiology , Endothelium, Vascular/microbiology , Intercellular Adhesion Molecule-1/metabolism , Leukocidins/metabolism , Pseudomonas aeruginosa/pathogenicity , Cell Line , Cyclooxygenase 2/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression Regulation , Intercellular Adhesion Molecule-1/genetics , Lipoxygenase/metabolism , Pseudomonas aeruginosa/metabolism
16.
Braz. j. microbiol ; 34(supl.1): 25-26, Nov. 2003.
Article in English | LILACS | ID: lil-389975

ABSTRACT

Para determinar o papel de ExoU na citotoxicidade tardia de P. aeruginosa, células endoteliais (CE) foram expostas às cepas PA103, PA103DxoU e PA103::exsA por 1h e à gentamicina em meio de cultura. Após 24h, a viabilidade das CE infectadas com PA103 (33.7 ± 14.3%) foi inferior à de CE infectadas com PA103DexoU (77.7 ± 6.3%) e PA103::exsA (79.5 ± 23.3%). A citotoxicidade não dependeu da capacidade de interagir com as CE porque o percentual de células com bactérias associadas em culturas expostas a PA103 foi semelhante ao percentual em culturas expostas a PA103DexoU e inferior em culturas expostas a PA103::exsA. O tratamento das CE com citocalasina D reduziu a internalização de PA103, mas não interferiu em sua citotoxicidade.

17.
Article in English | LILACS-Express | LILACS, VETINDEX | ID: biblio-1469470

ABSTRACT

To ascertain the role of ExoU in late P. aeruginosa cytotoxicity, endothelial cells (EC) were exposed to wild type PA103, PA103deltaexoU and PA103::exsA for 1h and to gentamicin in culture medium. After 24h, the viability of PA103-infected cells (33.7 ± 14.3%) was significantly lower than the viability of PA103deltaexoU- (77.7 ± 6.3%) or PA103::exsA- (79.5 ± 23.3%) infected EC. P. aeruginosa cytotoxicity did not depend on the bacterial ability to interact with EC because the percentage of cells with associated PA103 (35.9 ± 15.8%) was similar to the percentage in PA103deltaexoU- (34.2 ± 16.0%) and lower than the percentage in PA103::exsA-infected cultures (82.9 ± 18.9%). Cell treatment with cytochalasin D reduced the PA103 internalization by EC but did not interfere with its ability to kill host cells.


Para determinar o papel de ExoU na citotoxicidade tardia de P. aeruginosa, células endoteliais (CE) foram expostas às cepas PA103, PA103deltaxoU e PA103::exsA por 1h e à gentamicina em meio de cultura. Após 24h, a viabilidade das CE infectadas com PA103 (33.7 ± 14.3%) foi inferior à de CE infectadas com PA103deltaexoU (77.7 ± 6.3%) e PA103::exsA (79.5 ± 23.3%). A citotoxicidade não dependeu da capacidade de interagir com as CE porque o percentual de células com bactérias associadas em culturas expostas a PA103 foi semelhante ao percentual em culturas expostas a PA103deltaexoU e inferior em culturas expostas a PA103::exsA. O tratamento das CE com citocalasina D reduziu a internalização de PA103, mas não interferiu em sua citotoxicidade.

18.
Microb Pathog ; 33(4): 153-66, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12385743

ABSTRACT

Pseudomonas aeruginosa, a common agent of septicemia, enters into human endothelial cellsin vitro but the effects of bacterial infection have not been addressed properly. In this study, human umbilical vein endothelial cells (HUVEC) were infected by the noninvasive PA103 and the invasive PAO1 P. aeruginosa strains and the viability of infected cells was assessed by the methyltiazole tetrazolium (MTT) assay. Both strains were cytotoxic within 3h of infection. To ascertain the role of proteins secreted by the type III secretion system (TTSS) in HUVEC killing, defective mutants of PAO1 and PA103 were constructed by plasmid insertion in exsA or pscC genes. ExsA is a transcriptional regulator that controls the expression of most TTSS related genes whereas pscC encodes a protein from the secretion machinery. Parental bacteria were significantly more cytotoxic to HUVEC than the mutants. Inactivation ofexsA reverted the inability of PA103 to enter into HUVEC but did not modify the invasiveness of PAO1. Cytofluorometric analysis of infected HUVEC labeled by DiOC(6)(3) showed that cell killing was associated with mitochondrial depolarization, an early event reported in apoptosis. However, infected cells did not show ultrastructural or DNA fragmentation features of apoptosis. Our results suggest that TTSS effectors mediate P. aeruginosa killing of HUVEC by a mechanism distinct from apoptosis.


Subject(s)
Bacterial Proteins/toxicity , Endothelium, Vascular/pathology , Pseudomonas aeruginosa/pathogenicity , Apoptosis , Cells, Cultured , Endothelium, Vascular/ultrastructure , Gentamicins/pharmacology , Humans , Membrane Potentials , Mitochondria/physiology , Vasculitis/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...