Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Adv Exp Med Biol ; 1386: 325-345, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36258078

RESUMO

Bloodstream infections (BSI) with Pseudomonas aeruginosa account for 8.5% of all BSIs, their mortality rate, at about 40%, is the highest among causative agents. For this reason and due to its intrinsic and acquired resistance to antibiotics, P. aeruginosa represents a threat to public health systems. From the primary site of infection, often the urinary and respiratory tracts, P. aeruginosa uses its arsenal of virulence factors to cross both epithelial and endothelial barriers, ultimately reaching the bloodstream. In this chapter, we review the main steps involved in invasion and migration of P. aeruginosa into blood vessels, and the molecular mechanisms governing bacterial survival in blood. We also review the lifestyle of P. aeruginosa "on" and "in" host cells. In the context of genomic and phenotypic diversity of laboratory strains and clinical isolates, we underline the need for more standardized and robust methods applied to host-pathogen interaction studies, using several representative strains from distinct phylogenetic groups before drawing general conclusions. Finally, our literature survey reveals a need for further studies to complete our comprehension of the complex interplay between P. aeruginosa and the immune system in the blood, specifically in relation to the complement system cascade(s) and the Membrane Attack Complex (MAC), which play crucial roles in counteracting P. aeruginosa BSI.


Assuntos
Bacteriemia , Infecções por Pseudomonas , Humanos , Pseudomonas aeruginosa/genética , Infecções por Pseudomonas/microbiologia , Bacteriemia/microbiologia , Complexo de Ataque à Membrana do Sistema Complemento , Filogenia , Fatores de Virulência/genética , Antibacterianos/uso terapêutico
2.
PLoS Pathog ; 13(10): e1006630, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28968459

RESUMO

Guanylate binding proteins (GBPs) are interferon-inducible proteins involved in the cell-intrinsic immunity against numerous intracellular pathogens. The molecular mechanisms underlying the potent antibacterial activity of GBPs are still unclear. GBPs have been functionally linked to the NLRP3, the AIM2 and the caspase-11 inflammasomes. Two opposing models are currently proposed to explain the GBPs-inflammasome link: i) GBPs would target intracellular bacteria or bacteria-containing vacuoles to increase cytosolic PAMPs release ii) GBPs would directly facilitate inflammasome complex assembly. Using Francisella novicida infection, we investigated the functional interactions between GBPs and the inflammasome. GBPs, induced in a type I IFN-dependent manner, are required for the F. novicida-mediated AIM2-inflammasome pathway. Here, we demonstrate that GBPs action is not restricted to the AIM2 inflammasome, but controls in a hierarchical manner the activation of different inflammasomes complexes and apoptotic caspases. IFN-γ induces a quantitative switch in GBPs levels and redirects pyroptotic and apoptotic pathways under the control of GBPs. Furthermore, upon IFN-γ priming, F. novicida-infected macrophages restrict cytosolic bacterial replication in a GBP-dependent and inflammasome-independent manner. Finally, in a mouse model of tularemia, we demonstrate that the inflammasome and the GBPs are two key immune pathways functioning largely independently to control F. novicida infection. Altogether, our results indicate that GBPs are the master effectors of IFN-γ-mediated responses against F. novicida to control antibacterial immune responses in inflammasome-dependent and independent manners.


Assuntos
Francisella tularensis/imunologia , Proteínas de Ligação ao GTP/imunologia , Inflamassomos/imunologia , Interferon gama/imunologia , Tularemia/imunologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Imunofluorescência , Francisella , Técnicas de Silenciamento de Genes , Infecções por Bactérias Gram-Negativas/imunologia , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
PLoS Pathog ; 12(1): e1005377, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26727615

RESUMO

To achieve systemic infection, bacterial pathogens must overcome the critical and challenging step of transmigration across epithelial barriers. This is particularly true for opportunistic pathogens such as Pseudomonas aeruginosa, an agent which causes nosocomial infections. Despite extensive study, details on the mechanisms used by this bacterium to transmigrate across epithelial tissues, as well as the entry sites it uses, remain speculative. Here, using real-time microscopy and a model epithelial barrier, we show that P. aeruginosa employs a paracellular transmigration route, taking advantage of altered cell-cell junctions at sites of cell division or when senescent cells are expelled from the cell layer. Once a bacterium transmigrates, it is followed by a cohort of bacteria using the same entry point. The basal compartment is then invaded radially from the initial penetration site. Effective transmigration and propagation require type 4 pili, the type 3 secretion system (T3SS) and a flagellum, although flagellum-deficient bacteria can occasionally invade the basal compartment from wounded areas. In the basal compartment, the bacteria inject the T3SS toxins into host cells, disrupting the cytoskeleton and focal contacts to allow their progression under the cells. Thus, P. aeruginosa exploits intrinsic host cell processes to breach the epithelium and invade the subcellular compartment.


Assuntos
Células Epiteliais/metabolismo , Infecções por Pseudomonas/virologia , Pseudomonas aeruginosa/patogenicidade , Animais , Divisão Celular/fisiologia , Linhagem Celular , Senescência Celular/fisiologia , Cães , Humanos , Imuno-Histoquímica , Junções Intercelulares/metabolismo , Células Madin Darby de Rim Canino , Microscopia Confocal , Transfecção
4.
Int Immunol ; 29(8): 377-384, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28992059

RESUMO

While NLRC4-dependent sensing of intracellular Gram-negative pathogens such as Salmonella enterica serovar typhimurium is a beneficial host response, NLRC4-dependent sensing of the Pseudomonas aeruginosa type 3 secretion system (T3SS) has been shown to be involved in pathogenicity. In mice, different pathogen-associated microbial patterns are sensed by the combination of the NLRC4-inflammasome with different neuronal apoptosis inhibitory proteins (NAIPs). NAIP2 is involved in sensing PscI, an inner-rod protein of the P. aeruginosa T3SS. Surprisingly, only a single human NAIP (hNAIP) has been found. Moreover, there is no description of hNAIP-NLRC4 inflammasome recognition of T3SS inner-rod proteins in humans. Here, we show that the P. aeruginosa T3SS inner-rod protein PscI and needle protein PscF are both sensed by the hNAIP-NLRC4 inflammasome in human macrophages and PBMCs from healthy donors, allowing caspase-1 and IL-1ß maturation and resulting in a robust inflammatory response. TLR4 and TLR2 are involved in redundantly sensing these two T3SS components.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Inflamassomos/metabolismo , Macrófagos/imunologia , Proteína Inibidora de Apoptose Neuronal/metabolismo , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Sistemas de Secreção Tipo III/metabolismo , Animais , Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Humanos , Imunidade Inata , Peptídeos e Proteínas de Sinalização Intercelular , Interleucina-1beta/metabolismo , Macrófagos/microbiologia , Camundongos , Moléculas com Motivos Associados a Patógenos/imunologia , Células THP-1 , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Sistemas de Secreção Tipo III/imunologia
5.
Environ Microbiol ; 19(10): 4045-4064, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28654176

RESUMO

Pathogenic bacteria secrete protein toxins that provoke apoptosis or necrosis of eukaryotic cells. Here, we developed a live-imaging method, based on incorporation of a DNA-intercalating dye into membrane-damaged host cells, to study the kinetics of primary bone marrow-derived macrophages (BMDMs) mortality induced by opportunistic pathogen Pseudomonas aeruginosa expressing either Type III Secretion System (T3SS) toxins or the pore-forming toxin, Exolysin (ExlA). We found that ExlA promotes the activation of Caspase-1 and maturation of interleukin-1ß. BMDMs deficient for Caspase-1 and Caspase-11 were resistant to ExlA-induced death. Furthermore, by using KO BMDMs, we determined that the upstream NLRP3/ASC complex leads to the Caspase-1 activation. We also demonstrated that Pseudomonas putida and Pseudomonas protegens and the Drosophila pathogen Pseudomonas entomophila, which naturally express ExlA-like toxins, are cytotoxic toward macrophages and provoke the same type of pro-inflammatory death as does ExlA+ P. aeruginosa. These results demonstrate that ExlA-like toxins of two-partner secretion systems from diverse Pseudomonas species activate the NLRP3 inflammasome and provoke inflammatory pyroptotic death of macrophages.


Assuntos
Toxinas Bacterianas/toxicidade , Caspase 1/metabolismo , Morte Celular , Macrófagos/microbiologia , Pseudomonas/patogenicidade , Animais , Apoptose , Proteínas de Bactérias/toxicidade , Células da Medula Óssea , Inflamassomos , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pseudomonas/metabolismo
6.
Mol Microbiol ; 96(2): 419-36, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25614137

RESUMO

The export of bacterial toxins across the bacterial envelope requires the assembly of complex, membrane-embedded protein architectures. Pseudomonas aeruginosa employs type III secretion (T3S) injectisome to translocate exotoxins directly into the cytoplasm of a target eukaryotic cell. This multi-protein channel crosses two bacterial membranes and extends further as a needle through which the proteins travel. We show in this work that PscI, proposed to form the T3S system (T3SS) inner rod, possesses intrinsic properties to polymerize into flexible and regularly twisted fibrils and activates IL-1ß production in mouse bone marrow macrophages in vitro. We also found that point mutations within C-terminal amphipathic helix of PscI alter needle assembly in vitro and T3SS function in cell infection assays, suggesting that this region is essential for an efficient needle assembly. The overexpression of PscF partially compensates for the absence of the inner rod in PscI-deficient mutant by forming a secretion-proficient injectisome. All together, we propose that the polymerized PscI in P. aeruginosa optimizes the injectisome function by anchoring the needle within the envelope-embedded complex of the T3S secretome and - contrary to its counterpart in Salmonella - is not involved in substrate switching.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Sistemas de Secreção Tipo III/metabolismo , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Polimerização , Transporte Proteico , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética , Sistemas de Secreção Tipo III/química , Sistemas de Secreção Tipo III/genética
7.
PLoS Pathog ; 10(3): e1003939, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24626230

RESUMO

Infection of the vascular system by Pseudomonas aeruginosa (Pa) occurs during bacterial dissemination in the body or in blood-borne infections. Type 3 secretion system (T3SS) toxins from Pa induce a massive retraction when injected into endothelial cells. Here, we addressed the role of type 2 secretion system (T2SS) effectors in this process. Mutants with an inactive T2SS were much less effective than wild-type strains at inducing cell retraction. Furthermore, secretomes from wild-types were sufficient to trigger cell-cell junction opening when applied to cells, while T2SS-inactivated mutants had minimal activity. Intoxication was associated with decreased levels of vascular endothelial (VE)-cadherin, a homophilic adhesive protein located at endothelial cell-cell junctions. During the process, the protein was cleaved in the middle of its extracellular domain (positions 335 and 349). VE-cadherin attrition was T3SS-independent but T2SS-dependent. Interestingly, the epithelial (E)-cadherin was unaffected by T2SS effectors, indicating that this mechanism is specific to endothelial cells. We showed that one of the T2SS effectors, the protease LasB, directly affected VE-cadherin proteolysis, hence promoting cell-cell junction disruption. Furthermore, mouse infection with Pa to induce acute pneumonia lead to significant decreases in lung VE-cadherin levels, whereas the decrease was minimal with T2SS-inactivated or LasB-deleted mutant strains. We conclude that the T2SS plays a pivotal role during Pa infection of the vascular system by breaching the endothelial barrier, and propose a model in which the T2SS and the T3SS cooperate to intoxicate endothelial cells.


Assuntos
Antígenos CD/metabolismo , Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos/fisiologia , Caderinas/metabolismo , Metaloendopeptidases/metabolismo , Infecções por Pseudomonas/metabolismo , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Imunofluorescência , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C
8.
Infect Immun ; 83(5): 1789-98, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25690097

RESUMO

Pseudomonas aeruginosa is responsible for high-morbidity infections of cystic fibrosis patients and is a major agent of nosocomial infections. One of its most potent virulence factors is a type III secretion system (T3SS) that injects toxins directly into the host cell cytoplasm. ExsB, a lipoprotein localized in the bacterial outer membrane, is one of the components of this machinery, of which the function remained elusive until now. The localization of the exsB gene within the exsCEBA regulatory gene operon suggested an implication in the T3SS regulation, while its similarity with yscW from Yersinia spp. argued in favor of a role in machinery assembly. The present work shows that ExsB is necessary for full in vivo virulence of P. aeruginosa. Furthermore, the requirement of ExsB for optimal T3SS assembly and activity is demonstrated using eukaryotic cell infection and in vitro assays. In particular, ExsB promotes the assembly of the T3SS secretin in the bacterial outer membrane, highlighting the molecular role of ExsB as a pilotin. This involvement in the regulation of the T3S apparatus assembly may explain the localization of the ExsB-encoding gene within the regulatory gene operon.


Assuntos
Proteínas de Bactérias/metabolismo , Sistemas de Secreção Bacterianos , Lipoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Multimerização Proteica , Pseudomonas aeruginosa/fisiologia , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/microbiologia , Humanos , Lipoproteínas/genética , Masculino , Camundongos Endogâmicos BALB C , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/genética , Análise de Sobrevida , Fatores de Virulência/genética
9.
Biophys J ; 104(7): 1445-55, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23561521

RESUMO

Pseudomonas aeruginosa is a major agent of hospital-acquired infections, and a pathogen of immunocompromised, cystic fibrosis and burn patients. It uses a type III secretion system for the injection of toxins directly into host cells, through a translocon assembled in the host cell membrane. The hydrophobic translocator subunits of this system, PopB and PopD, have membrane permeabilizing activity based on previous dye leakage experiments, but little is known about the mechanism of assembly and the pore properties of this translocon. Using electrophysiology, we have observed that an equimolar mixture of PopB and PopD induces current fluctuations in planar lipid bilayers, with a unitary conductance of 57 pS in 1 M KCl and numerous larger conductance levels. The activity depends on voltage magnitude and polarity, and increases with protein concentration and the duration of the voltage step. PopB alone is sufficient for producing current fluctuations. PopD rarely displays any transitions, but accelerates PopB onset of activity. The effects of pH, ionic strength, and lipid composition have also been explored. Our data provide new, to our knowledge, insights into the behavior of PopB and PopD by highlighting similarities with secreted pore-forming peptides, and by suggesting that PopB/PopD may form channels via the toroidal pore model. We believe that the events we report here represent the initial steps of insertion and assembly of these translocators in the membrane.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Fenômenos Eletrofisiológicos , Pseudomonas aeruginosa/metabolismo , Animais , Antígenos de Bactérias/química , Antígenos de Bactérias/toxicidade , Proteínas de Bactérias/química , Proteínas de Bactérias/toxicidade , Guanidina/farmacologia , Concentração de Íons de Hidrogênio , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Macrófagos/efeitos dos fármacos , Membranas Artificiais , Camundongos , Concentração Osmolar , Porosidade , Redobramento de Proteína/efeitos dos fármacos
10.
BMC Biochem ; 12: 46, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21861921

RESUMO

BACKGROUND: Pathogens depend on peptidase activities to accomplish many physiological processes, including interaction with their hosts, highlighting parasitic peptidases as potential drug targets. In this study, a major leucyl aminopeptidolytic activity was identified in Trypanosoma cruzi, the aetiological agent of Chagas disease. RESULTS: The enzyme was isolated from epimastigote forms of the parasite by a two-step chromatographic procedure and associated with a single 330-kDa homohexameric protein as determined by sedimentation velocity and light scattering experiments. Peptide mass fingerprinting identified the enzyme as the predicted T. cruzi aminopeptidase EAN97960. Molecular and enzymatic analysis indicated that this leucyl aminopeptidase of T. cruzi (LAPTc) belongs to the peptidase family M17 or leucyl aminopeptidase family. LAPTc has a strong dependence on neutral pH, is mesophilic and retains its oligomeric form up to 80°C. Conversely, its recombinant form is thermophilic and requires alkaline pH. CONCLUSIONS: LAPTc is a 330-kDa homohexameric metalloaminopeptidase expressed by all T. cruzi forms and mediates the major parasite leucyl aminopeptidolytic activity. Since biosynthetic pathways for essential amino acids, including leucine, are lacking in T. cruzi, LAPTc could have a function in nutritional supply.


Assuntos
Leucil Aminopeptidase/química , Leucil Aminopeptidase/metabolismo , Multimerização Proteica , Trypanosoma cruzi/enzimologia , Sequência de Aminoácidos , Citoplasma/metabolismo , Descoberta de Drogas , Hidrólise , Leucil Aminopeptidase/classificação , Leucil Aminopeptidase/isolamento & purificação , Dados de Sequência Molecular , Filogenia , Estrutura Quaternária de Proteína , Transporte Proteico , Alinhamento de Sequência , Trypanosoma cruzi/citologia , Trypanosoma cruzi/efeitos dos fármacos
11.
Metabolites ; 11(2)2021 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-33494144

RESUMO

Pseudomonas aeruginosa (P.a) is one of the most critical antibiotic resistant bacteria in the world and is the most prevalent pathogen in cystic fibrosis (CF), causing chronic lung infections that are considered one of the major causes of mortality in CF patients. Although several studies have contributed to understanding P.a within-host adaptive evolution at a genomic level, it is still difficult to establish direct relationships between the observed mutations, expression of clinically relevant phenotypes, and clinical outcomes. Here, we performed a comparative untargeted LC/HRMS-based metabolomics analysis of sequential isolates from chronically infected CF patients to obtain a functional view of P.a adaptation. Metabolic profiles were integrated with expression of bacterial phenotypes and clinical measurements following multiscale analysis methods. Our results highlighted significant associations between P.a "metabotypes", expression of antibiotic resistance and virulence phenotypes, and frequency of clinical exacerbations, thus identifying promising biomarkers and therapeutic targets for difficult-to-treat P.a infections.

12.
J Mol Biol ; 432(24): 166690, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-33289667

RESUMO

Pseudomonas aeruginosa is an opportunistic bacterium of which the main virulence factor is the Type III Secretion System. The ATPase of this machinery, PscN (SctN), is thought to be localized at the base of the secretion apparatus and to participate in the recognition, chaperone dissociation and unfolding of exported T3SS proteins. In this work, a protein-protein interaction ELISA revealed the interaction of PscN with a wide range of exported T3SS proteins including the needle, translocator, gate-keeper and effector. These interactions were further confirmed by Microscale Thermophoresis that also indicated a preferential interaction of PscN with secreted proteins or protein-chaperone complex rather than with chaperones alone, in line with the release of the chaperones in the bacterial cytoplasm after the dissociation from their exported proteins. Moreover, we suggest a new role of the gate-keeper complex and the ATPase in the regulation of early substrates recognition by the T3SS. This finding sheds a new light on the mechanism of secretion switching from early to middle substrates in P. aeruginosa.


Assuntos
Adenosina Trifosfatases/genética , Chaperonas Moleculares/genética , Pseudomonas aeruginosa/genética , Sistemas de Secreção Tipo III/genética , Adenosina Trifosfatases/química , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Chaperonas Moleculares/química , Mapas de Interação de Proteínas/genética , Pseudomonas aeruginosa/patogenicidade , Especificidade por Substrato , Sistemas de Secreção Tipo III/química , Fatores de Virulência/química , Fatores de Virulência/genética
13.
ACS Infect Dis ; 5(11): 1843-1854, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31525902

RESUMO

Pseudomonas aeruginosa (P. aeruginosa) is an opportunistic pathogen naturally resistant to many common antibiotics and acquires new resistance traits at an alarming pace. Targeting the bacterial virulence factors by an antivirulence strategy, therefore, represents a promising alternative approach besides antibiotic therapy. The Type III secretion system (T3SS) of P. aeruginosa is one of its main virulence factors. It consists of more than 20 proteins building a complex syringe-like machinery enabling the injection of toxin into host cells. Previous works showed that disrupting interactions between components of this machinery efficiently lowers the bacterial virulence. Using automated target-based screening of commercial and in-house libraries of small molecules, we identified compounds inhibiting the protein-protein interaction between PscE and PscG, the two cognate chaperones of the needle subunit PscF of P. aeruginosa T3SS. Two hits were selected and assembled using Split/Mix/Click chemistry to build larger hybrid analogues. Their efficacy and toxicity were evaluated using phenotypic analysis including automated microscopy and image analysis. Two nontoxic hybrid leads specifically inhibited the T3SS and reduced the ex vivo cytotoxicity of bacteria and their virulence in Galleria mellonella.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/patogenicidade , Sistemas de Secreção Tipo III/metabolismo , Animais , Antibacterianos/química , Proteínas de Bactérias/genética , Modelos Animais de Doenças , Humanos , Mariposas , Ligação Proteica/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Sistemas de Secreção Tipo III/genética , Virulência/efeitos dos fármacos
14.
Front Microbiol ; 10: 573, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001211

RESUMO

The type three secretion system (T3SS) is a macromolecular protein nano-syringe used by different bacterial pathogens to inject effectors into host cells. The extracellular part of the syringe is a needle-like filament formed by the polymerization of a 9-kDa protein whose structure and proper localization on the bacterial surface are key determinants for efficient toxin injection. Here, we combined in vivo, in vitro, and in silico approaches to characterize the Pseudomonas aeruginosa T3SS needle and its major component PscF. Using a combination of mutagenesis, phenotypic analyses, immunofluorescence, proteolysis, mass spectrometry, atomic force microscopy, electron microscopy, and molecular modeling, we propose a model of the P. aeruginosa needle that exposes the N-terminal region of each PscF monomer toward the outside of the filament, while the core of the fiber is formed by the C-terminal helix. Among mutations introduced into the needle protein PscF, D76A, and P47A/Q54A caused a defect in the assembly of the needle on the bacterial surface, although the double mutant was still cytotoxic on macrophages in a T3SS-dependent manner and formed filamentous structures in vitro. These results suggest that the T3SS needle of P. aeruginosa displays an architecture that is similar to that of other bacterial needles studied to date and highlight the fact that small, targeted perturbations in needle assembly can inhibit T3SS function. Therefore, the T3SS needle represents an excellent drug target for small molecules acting as virulence blockers that could disrupt pathogenesis of a broad range of bacteria.

15.
Sci Rep ; 8(1): 5, 2018 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-29311668

RESUMO

Pathogenic bacteria induce eukaryotic cell damage which range from discrete modifications of signalling pathways, to morphological alterations and even to cell death. Accurate quantitative detection of these events is necessary for studying host-pathogen interactions and for developing strategies to protect host organisms from bacterial infections. Investigation of morphological changes is cumbersome and not adapted to high-throughput and kinetics measurements. Here, we describe a simple and cost-effective method based on automated analysis of live cells with stained nuclei, which allows real-time quantification of bacteria-induced eukaryotic cell damage at single-cell resolution. We demonstrate that this automated high-throughput microscopy approach permits screening of libraries composed of interference-RNA, bacterial strains, antibodies and chemical compounds in ex vivo infection settings. The use of fluorescently-labelled bacteria enables the concomitant detection of changes in bacterial growth. Using this method named CLIQ-BID (Cell Live Imaging Quantification of Bacteria Induced Damage), we were able to distinguish the virulence profiles of different pathogenic bacterial species and clinical strains.


Assuntos
Fenômenos Fisiológicos Bacterianos , Células Eucarióticas/microbiologia , Células Eucarióticas/patologia , Imagem Molecular/métodos , Animais , Células Endoteliais , Células HeLa , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Células NIH 3T3
16.
FEBS J ; 274(14): 3601-3610, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17578515

RESUMO

Type III secretion systems of Gram-negative pathogenic bacteria allow the injection of effector proteins into the cytosol of host eukaryotic cells. Crossing of the eukaryotic plasma membrane is facilitated by a translocon, an oligomeric structure made up of two bacterial proteins inserted into the host membrane during infection. In Pseudomonas aeruginosa, a major human opportunistic pathogen, these proteins are PopB and PopD. Their interactions with their common chaperone PcrH in the cytosol of the bacteria are essential for the proper function of the injection system. The interaction region between PopD and PcrH was identified using limited proteolysis, revealing that the putative PopD transmembrane fragment is buried within the PopD/PcrH complex. In addition, structural features of PopD and PcrH, either individually or within the binary complex, were characterized using spectroscopic methods and 1D NMR. Whereas PcrH possesses the characteristics of a folded protein, PopD is in a molten globule state either alone or in the PopD/PcrH complex. The molten globule state is known to enable the membrane insertion of translocation/pore-forming domains of bacterial toxins. Therefore, within the bacterial cytoplasm, PopD is preserved in a state that is favorable to secretion and insertion into cell membranes.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Proteínas de Bactérias/genética , Dicroísmo Circular , Interações Hidrofóbicas e Hidrofílicas , Chaperonas Moleculares/genética , Conformação Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Dobramento de Proteína , Solventes
17.
Biochem J ; 396(3): 509-15, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16542158

RESUMO

Apyrase activity is present in the saliva of haematophagous arthropods. It is related to blood-feeding because of the apyrase ability to hydrolyse ADP, a key component of platelet aggregation. Five apyrases with apparent molecular masses of 88, 82, 79, 68 and 67 kDa were identified in the saliva of the vector of Chagas disease, Triatoma infestans. The large size observed during purification of these enzymes suggested oligomerization. In the present study, we confirmed, using gel-filtration and analytical ultracentrifugation, the presence of apyrase oligomers with molecular masses of 200 kDa in the saliva. Electrophoretic analyses showed that disulphide bonds were involved in homo-oligomerization. In addition, heterogeneity in disulphide bonds and in pI was detected, with the pI ranging from 4.9 to 5.4. The present study gives the first insights into the quaternary structure of soluble apyrases.


Assuntos
Apirase/biossíntese , Triatoma/enzimologia , Animais , Apirase/isolamento & purificação , Cromatografia em Gel , Reagentes de Ligações Cruzadas/química , Dissulfetos/química , Ditiotreitol/química , Eletroforese em Gel de Poliacrilamida , Estrutura Quaternária de Proteína , Saliva/enzimologia , Succinimidas/química , Ultracentrifugação
18.
mBio ; 8(1)2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28119472

RESUMO

Clinical strains of Pseudomonas aeruginosa lacking the type III secretion system genes employ a toxin, exolysin (ExlA), for host cell membrane disruption. Here, we demonstrated that ExlA export requires a predicted outer membrane protein, ExlB, showing that ExlA and ExlB define a new active two-partner secretion (TPS) system of P. aeruginosa In addition to the TPS signals, ExlA harbors several distinct domains, which include one hemagglutinin domain, five arginine-glycine-aspartic acid (RGD) motifs, and a C-terminal region lacking any identifiable sequence motifs. However, this C-terminal region is important for the toxic activity, since its deletion abolishes host cell lysis. Using lipid vesicles and eukaryotic cells, including red blood cells, we demonstrated that ExlA has a pore-forming activity which precedes cell membrane disruption of nucleated cells. Finally, we developed a high-throughput cell-based live-dead assay and used it to screen a transposon mutant library of an ExlA-producing P. aeruginosa clinical strain for bacterial factors required for ExlA-mediated toxicity. The screen resulted in the identification of proteins involved in the formation of type IV pili as being required for ExlA to exert its cytotoxic activity by promoting close contact between bacteria and the host cell. These findings represent the first example of cooperation between a pore-forming toxin of the TPS family and surface appendages in host cell intoxication. IMPORTANCE: The course and outcome of acute, toxigenic infections by Pseudomonas aeruginosa clinical isolates rely on the deployment of one of two virulence strategies: delivery of effectors by the well-known type III secretion system or the cytolytic activity of the recently identified two-partner secreted toxin, exolysin. Here, we characterize several features of the mammalian cell intoxication process mediated by exolysin. We found that exolysin requires the outer membrane protein ExlB for export into extracellular medium. Using in vitro recombinant protein and ex vivo assays, we demonstrated a pore-forming activity of exolysin. A cellular cytotoxicity screen of a transposon mutant library, made in an exolysin-producing clinical strain, identified type IV pili as bacterial appendages required for exolysin toxic function. This work deciphers molecular mechanisms underlying the activity of novel virulence factors used by P. aeruginosa clinical strains lacking the type III secretion system, including a requirement for the toxin-producing bacteria to be attached to the targeted cell to induce cytolysis, and defines new targets for developing antivirulence strategies.


Assuntos
Fímbrias Bacterianas/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Pseudomonas aeruginosa/fisiologia , Sistemas de Secreção Tipo II/metabolismo , Sobrevivência Celular , Elementos de DNA Transponíveis , Mutagênese Insercional , Pseudomonas aeruginosa/genética
19.
J Mol Biol ; 428(9 Pt A): 1790-803, 2016 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-27012424

RESUMO

The type V secretion system is a macromolecular machine employed by a number of bacteria to secrete virulence factors into the environment. The human pathogen Pseudomonas aeruginosa employs the newly described type Vd secretion system to secrete a soluble variant of PlpD, a lipase of the patatin-like family synthesized as a single macromolecule that also carries a polypeptide transport-associated domain and a 16-stranded ß-barrel. Here we report the crystal structure of the secreted form of PlpD in its biologically active state. PlpD displays a classical lipase α/ß hydrolase fold with a catalytic site located within a highly hydrophobic channel that entraps a lipidic molecule. The active site is covered by a flexible lid, as in other lipases, indicating that this region in PlpD must modify its conformation in order for catalysis at the water-lipid interface to occur. PlpD displays phospholipase A1 activity and is able to recognize a number of phosphatidylinositols and other phosphatidyl analogs. PlpD is the first example of an active phospholipase secreted through the type V secretion system, for which there are more than 200 homologs, revealing details of the lipid destruction arsenal expressed by P. aeruginosa in order to establish infection.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Fosfolipases/química , Fosfolipases/metabolismo , Pseudomonas aeruginosa/enzimologia , Domínio Catalítico , Cristalografia por Raios X , Modelos Moleculares , Fosfatidilinositóis/metabolismo , Conformação Proteica , Especificidade por Substrato , Sistemas de Secreção Tipo V/metabolismo
20.
Insect Biochem Mol Biol ; 34(10): 1051-8, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15475299

RESUMO

Apyrases are nucleoside triphosphate-diphosphohydrolases that remove Pi from ATP and ADP. The blood feeding reduviid Triatoma infestans, which transmits the Trypanosoma cruzi agent of Chagas disease to animals and man, presents in its salivary glands five apyrases with molecular masses of 88, 82, 79, 68 and 67 kDa. These triatomine apyrases have been associated with the prevention of ADP induced platelet aggregation in the host. Here we provide biochemical data showing that these apyrases are stored in the lumen of the salivary gland D1 pairs, and that about one half of the pool of the enzyme is consumed during feeding. After the feeding recovery of apyrases to maximal activity level takes days, thus suggesting de novo protein synthesis. This hypothesis is supported by quantitative RT-PCR analysis which shows an upregulation of the 79 kDa apyrase mRNA level after feeding.


Assuntos
Apirase/metabolismo , Triatoma/enzimologia , Animais , Apirase/química , Apirase/genética , Sequência de Bases , Sangue , Doença de Chagas/transmissão , DNA/genética , Ingestão de Alimentos , Expressão Gênica , Humanos , Insetos Vetores/enzimologia , Insetos Vetores/genética , Insetos Vetores/parasitologia , Cinética , Peso Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândulas Salivares/enzimologia , Triatoma/genética , Triatoma/parasitologia , Trypanosoma cruzi/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA