Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microbiol Spectr ; 11(4): e0521722, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37255442

RESUMO

Peptidoglycan is an essential component of the bacterial cell envelope that sustains the turgor pressure of the cytoplasm, determines cell shape, and acts as a scaffold for the anchoring of envelope polymers such as lipoproteins. The final cross-linking step of peptidoglycan polymerization is performed by classical d,d-transpeptidases belonging to the penicillin-binding protein (PBP) family and by l,d-transpeptidases (LDTs), which are dispensable for growth in most bacterial species and whose physiological functions remain elusive. In this study, we investigated the contribution of LDTs to cell envelope synthesis in Pseudomonas aeruginosa grown in planktonic and biofilm conditions. We first assigned a function to each of the three P. aeruginosa LDTs by gene inactivation in P. aeruginosa, heterospecific gene expression in Escherichia coli, and, for one of them, direct determination of its enzymatic activity. We found that the three P. aeruginosa LDTs catalyze peptidoglycan cross-linking (LdtPae1), the anchoring of lipoprotein OprI to the peptidoglycan (LdtPae2), and the hydrolysis of the resulting peptidoglycan-OprI amide bond (LdtPae3). Construction of a phylogram revealed that LDTs performing each of these three functions in various species cannot be assigned to distinct evolutionary lineages, in contrast to what has been observed with PBPs. We showed that biofilm, but not planktonic bacteria, displayed an increase proportion of peptidoglycan cross-links formed by LdtPae1 and a greater extent of OprI anchoring to peptidoglycan, which is controlled by LdtPae2 and LdtPae3. Consistently, deletion of each of the ldt genes impaired biofilm formation and potentiated the bactericidal activity of EDTA. These results indicate that LDTs contribute to the stabilization of the bacterial cell envelope and to the adaptation of peptidoglycan metabolism to growth in biofilm. IMPORTANCE Active-site cysteine LDTs form a functionally heterologous family of enzymes that contribute to the biogenesis of the bacterial cell envelope through formation of peptidoglycan cross-links and through the dynamic anchoring of lipoproteins to peptidoglycan. Here, we report the role of three P. aeruginosa LDTs that had not been previously characterized. We show that these enzymes contribute to resistance to the bactericidal activity of EDTA and to the adaptation of cell envelope polymers to conditions that prevail in biofilms. These results indicate that LDTs should be considered putative targets in the development of drug-EDTA associations for the control of biofilm-related infections.


Assuntos
Peptidil Transferases , Peptidil Transferases/genética , Peptidil Transferases/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Peptidoglicano/metabolismo , Ácido Edético , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/metabolismo , Escherichia coli/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
2.
iScience ; 26(4): 106286, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-36942053

RESUMO

Animal models for studying human pathogens are crucially lacking. We describe the implantation in mice of engineered human mature microvasculature consisting of endothelial and perivascular cells embedded in collagen hydrogel that allows investigation of pathogen interactions with the endothelium, including in vivo functional studies. Using Neisseria meningitidis as a paradigm of human-restricted infection, we demonstrated the strength and opportunities associated with the use of this approach.

3.
ChemMedChem ; 18(9): e202300077, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36779293

RESUMO

Ruthenium(II) alkyne azide cycloaddition (RuAAC) is an attractive reaction to access 1,5-triazole derivatives and is applicable to internal alkynes. Here, we explore RuAAC to introduce molecular diversity on the diazabicyclooctane (DBO) scaffold of ß-lactamase inhibitors. The methodology presented is fully regioselective and enabled synthesis of a series of 1,5-triazole DBOs and trisubstituted analogues. Molecular modelling and biological evaluation revealed that the DBO substituents provided putative stabilizing interactions in the active site of broad-spectrum ß-lactamase KPC-2 and promising activity against a hyperpermeable strain of Escherichia coli producing KPC-2.


Assuntos
Rutênio , Inibidores de beta-Lactamases , Inibidores de beta-Lactamases/química , Rutênio/farmacologia , Rutênio/química , Reação de Cicloadição , Azidas , Triazóis/química , Catálise , Alcinos
4.
Antimicrob Agents Chemother ; 66(9): e0235721, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35943263

RESUMO

Treatment of multidrug-resistant tuberculosis with combinations of carbapenems and ß-lactamase inhibitors carries risks for dysbiosis and for the development of resistances in the intestinal microbiota. Using Escherichia coli producing carbapenemase KPC-2 as a model, we show that carbapenems can be modified to obtain drugs that are inactive against E. coli but retain antitubercular activity. Furthermore, functionalization of the diazabicyclooctanes scaffold provided drugs that did not effectively inactivate KPC-2 but retained activity against Mycobacterium tuberculosis targets.


Assuntos
Carbapenêmicos , Mycobacterium tuberculosis , Antibacterianos/farmacologia , Proteínas de Bactérias/farmacologia , Carbapenêmicos/farmacologia , Escherichia coli , Meropeném/farmacologia , Testes de Sensibilidade Microbiana , Inibidores de beta-Lactamases/farmacologia , beta-Lactamases/farmacologia
6.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34725157

RESUMO

Neisseria meningitidis utilizes type IV pili (T4P) to adhere to and colonize host endothelial cells, a process at the heart of meningococcal invasive diseases leading to meningitis and sepsis. T4P are polymers of an antigenically variable major pilin building block, PilE, plus several core minor pilins that initiate pilus assembly and are thought to be located at the pilus tip. Adhesion of N. meningitidis to human endothelial cells requires both PilE and a conserved noncore minor pilin PilV, but the localization of PilV and its precise role in this process remains to be clarified. Here, we show that both PilE and PilV promote adhesion to endothelial vessels in vivo. The substantial adhesion defect observed for pilV mutants suggests it is the main adhesin. Consistent with this observation, superresolution microscopy showed the abundant distribution of PilV throughout the pilus. We determined the crystal structure of PilV and modeled it within the pilus filament. The small size of PilV causes it to be recessed relative to adjacent PilE subunits, which are dominated by a prominent hypervariable loop. Nonetheless, we identified a conserved surface-exposed adhesive loop on PilV by alanine scanning mutagenesis. Critically, antibodies directed against PilV inhibit N. meningitidis colonization of human skin grafts. These findings explain how N. meningitidis T4P undergo antigenic variation to evade the humoral immune response while maintaining their adhesive function and establish the potential of this highly conserved minor pilin as a vaccine and therapeutic target for the prevention and treatment of N. meningitidis infections.


Assuntos
Aderência Bacteriana , Proteínas de Bactérias/fisiologia , Fímbrias Bacterianas/fisiologia , Neisseria meningitidis/fisiologia , Animais , Anticorpos/uso terapêutico , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Feminino , Fímbrias Bacterianas/química , Fímbrias Bacterianas/ultraestrutura , Humanos , Infecções Meningocócicas/tratamento farmacológico , Camundongos SCID
7.
PLoS Pathog ; 17(2): e1009299, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33592056

RESUMO

Neisseria meningitidis (the meningococcus) remains a major cause of bacterial meningitis and fatal sepsis. This commensal bacterium of the human nasopharynx can cause invasive diseases when it leaves its niche and reaches the bloodstream. Blood-borne meningococci have the ability to adhere to human endothelial cells and rapidly colonize microvessels. This crucial step enables dissemination into tissues and promotes deregulated inflammation and coagulation, leading to extensive necrotic purpura in the most severe cases. Adhesion to blood vessels relies on type IV pili (TFP). These long filamentous structures are highly dynamic as they can rapidly elongate and retract by the antagonistic action of two ATPases, PilF and PilT. However, the consequences of TFP dynamics on the pathophysiology and the outcome of meningococcal sepsis in vivo have been poorly studied. Here, we show that human graft microvessels are replicative niches for meningococci, that seed the bloodstream and promote sustained bacteremia and lethality in a humanized mouse model. Intriguingly, although pilus-retraction deficient N. meningitidis strain (ΔpilT) efficiently colonizes human graft tissue, this mutant did not promote sustained bacteremia nor induce mouse lethality. This effect was not due to a decreased inflammatory response, nor defects in bacterial clearance by the innate immune system. Rather, TFP-retraction was necessary to promote the release of TFP-dependent contacts between bacteria and, in turn, the detachment from colonized microvessels. The resulting sustained bacteremia was directly correlated with lethality. Altogether, these results demonstrate that pilus retraction plays a key role in the occurrence and outcome of meningococcal sepsis by supporting sustained bacteremia. These findings open new perspectives on the role of circulating bacteria in the pathological alterations leading to lethal sepsis.


Assuntos
Bacteriemia/microbiologia , Modelos Animais de Doenças , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/fisiologia , Infecções Meningocócicas/microbiologia , Neisseria meningitidis/patogenicidade , Sepse/microbiologia , Animais , Bacteriemia/metabolismo , Bacteriemia/patologia , Aderência Bacteriana , Células Endoteliais , Feminino , Proteínas de Fímbrias/genética , Humanos , Infecções Meningocócicas/metabolismo , Infecções Meningocócicas/patologia , Camundongos , Camundongos SCID , Sepse/metabolismo , Sepse/patologia , Transplante de Pele
8.
J Clin Microbiol ; 59(3)2021 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-33328176

RESUMO

Staphylococcus epidermidis is a pathogen emerging worldwide as a leading cause of health care-associated infections. A standardized high-resolution typing method to document transmission and dissemination of multidrug-resistant S. epidermidis strains is needed. Our aim was to provide a core genome multilocus sequence typing (cgMLST) scheme for S. epidermidis to improve the international surveillance of S. epidermidis We defined a cgMLST scheme based on 699 core genes and used it to investigate the population structure of the species and the genetic relatedness of isolates recovered from infants hospitalized in several wards of a French hospital. Our results show the long-lasting endemic persistence of S. epidermidis clones within and across wards of hospitals and demonstrate the ability of our cgMLST approach to identify and track these clones. We made the scheme publicly available through the Institut Pasteur BIGSdb server (http://bigsdb.pasteur.fr/epidermidis/). This tool should enable international harmonization of the epidemiological surveillance of multidrug-resistant S. epidermidis clones. By comparing gene distribution among infection and commensal isolates, we also confirmed the association of the mecA locus with infection isolates and of the fdh gene with commensal isolates. (This study has been registered at ClinicalTrials.gov under registration no. NCT03374371.).


Assuntos
Infecções Estafilocócicas , Staphylococcus epidermidis , Células Clonais , Genoma Bacteriano/genética , Hospitais , Humanos , Tipagem de Sequências Multilocus , Infecções Estafilocócicas/epidemiologia , Staphylococcus epidermidis/genética
9.
mSphere ; 4(6)2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31801841

RESUMO

Neisseria meningitidis is an inhabitant of the nasopharynx, from which it is transmitted from person to person or disseminates in blood and becomes a harmful pathogen. In this work, we addressed colonization of the nasopharyngeal niche by focusing on the interplay between meningococci and the airway mucus that lines the mucosa of the host. Using Calu-3 cells grown in air interface culture (cells grown with the apical domain facing air), we studied meningococcal colonization of the mucus and the host response. Our results suggested that N. meningitidis behaved like commensal bacteria in mucus, without interacting with human cells or actively transmigrating through the cell layer. As a result, type IV pili do not play a role in this model, and meningococci did not trigger a strong innate immune response from the Calu-3 cells. Finally, we have shown that this model is suitable for studying interaction of N. meningitidis with other bacteria living in the nasopharynx and that Streptococcus mitis, but not Moraxella catarrhalis, can promote meningococcal growth in this model.IMPORTANCEN. meningitidis is transmitted from person to person by aerosol droplets produced by breathing, talking, or coughing or by direct contact with a contaminated fluid. The natural reservoir of N. meningitidis is the human nasopharynx mucosa, located at the back of the nose and above the oropharynx. The means by which meningococci cross the nasopharyngeal wall is still under debate, due to the lack of a convenient and relevant model mimicking the nasopharyngeal niche. Here, we took advantage of Calu-3 cells grown in air interface culture to study how meningococci colonize the nasopharyngeal niche. We report that the airway mucus is both a niche for meningococcal growth and a protective barrier against N. meningitidis infection. As such, N. meningitidis behaves like commensal bacteria and is unlikely to induce infection without an external trigger.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Fatores Imunológicos/metabolismo , Muco/metabolismo , Nasofaringe/imunologia , Nasofaringe/microbiologia , Neisseria meningitidis/imunologia , Linhagem Celular , Humanos , Modelos Teóricos , Mucosite/imunologia , Mucosite/microbiologia
10.
J Infect Dis ; 220(12): 1967-1976, 2019 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-31420648

RESUMO

Staphylococcus aureus is a leading cause of both acute and chronic infections in humans. The importance of the pentose phosphate pathway (PPP) during S. aureus infection is currently largely unexplored. In the current study, we focused on one key PPP enzyme, transketolase (TKT). We showed that inactivation of the unique gene encoding TKT activity in S. aureus USA300 (∆tkt) led to drastic metabolomic changes. Using time-lapse video imaging and mice infection, we observed a major defect of the ∆tkt strain compared with wild-type strain in early intracellular proliferation and in the ability to colonize kidneys. Transcriptional activity of the 2 master regulators sigma B and RpiRc was drastically reduced in the ∆tkt mutant during host cells invasion. The concomitant increased RNAIII transcription suggests that TKT-or a functional PPP-strongly influences the ability of S. aureus to proliferate within host cells by modulating key transcriptional regulators.


Assuntos
Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Estresse Fisiológico , Transcetolase/metabolismo , Animais , Carbono/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Regulação Bacteriana da Expressão Gênica , Inativação Gênica , Genes Bacterianos , Humanos , Rim/metabolismo , Rim/microbiologia , Metabolômica/métodos , Camundongos , Mutação , Fenótipo , Transdução de Sinais , Staphylococcus aureus/enzimologia , Estresse Fisiológico/genética , Transcetolase/genética
11.
Nat Microbiol ; 4(6): 972-984, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30911127

RESUMO

Bacterial virulence factors are attractive targets for the development of therapeutics. Type IV pili, which are associated with a remarkable array of properties including motility, the interaction between bacteria and attachment to biotic and abiotic surfaces, represent particularly appealing virulence factor targets. Type IV pili are present in numerous bacterial species and are critical for their pathogenesis. In this study, we report that trifluoperazine and related phenothiazines block functions associated with Type IV pili in different bacterial pathogens, by affecting piliation within minutes. Using Neisseria meningitidis as a paradigm of Gram-negative bacterial pathogens that require Type IV pili for pathogenesis, we show that piliation is sensitive to altered activity of the Na+ pumping NADH-ubiquinone oxidoreductase (Na+-NQR) complex and that these compounds probably altered the establishment of the sodium gradient. In vivo, these compounds exert a strong protective effect. They reduce meningococcal colonization of the human vessels and prevent subsequent vascular dysfunctions, intravascular coagulation and overwhelming inflammation, the hallmarks of invasive meningococcal infections. Finally, they reduce lethality. This work provides a proof of concept that compounds with activity against bacterial Type IV pili could beneficially participate in the treatment of infections caused by Type IV pilus-expressing bacteria.


Assuntos
Fímbrias Bacterianas/efeitos dos fármacos , Fímbrias Bacterianas/fisiologia , Infecções Meningocócicas/prevenção & controle , Neisseria meningitidis/efeitos dos fármacos , Fatores de Virulência , Animais , Antibacterianos/farmacologia , Vasos Sanguíneos/lesões , Vasos Sanguíneos/microbiologia , Vasos Sanguíneos/patologia , Combinação de Medicamentos , Complexo I de Transporte de Elétrons , Feminino , Fímbrias Bacterianas/genética , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Bactérias Gram-Negativas , Humanos , Camundongos , Neisseria meningitidis/genética , Neisseria meningitidis/crescimento & desenvolvimento , Fenotiazinas/farmacologia , Pele/patologia , Transplante de Pele , ATPase Trocadora de Sódio-Potássio , Trifluoperazina/farmacologia
13.
PLoS Pathog ; 14(4): e1006981, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29630665

RESUMO

Purpura fulminans is a deadly complication of Neisseria meningitidis infections due to extensive thrombosis of microvessels. Although a Disseminated Intra-vascular Coagulation syndrome (DIC) is frequently observed during Gram negative sepsis, it is rarely associated with extensive thrombosis like those observed during meningococcemia, suggesting that the meningococcus induces a specific dysregulation of coagulation. Another specific feature of N. meningitidis pathogenesis is its ability to colonize microvessels endothelial cells via type IV pili. Importantly, endothelial cells are key in controlling the coagulation cascade through the activation of the potent anticoagulant Protein C (PC) thanks to two endothelial cell receptors among which the Endothelial Protein C Receptor (EPCR). Considering that congenital or acquired deficiencies of PC are associated with purpura fulminans, we hypothesized that a defect in the activation of PC following meningococcal adhesion to microvessels is responsible for the thrombotic events observed during meningococcemia. Here we showed that the adhesion of N. meningitidis on endothelial cells results in a rapid and intense decrease of EPCR expression by inducing its cleavage in a process know as shedding. Using siRNA experiments and CRISPR/Cas9 genome edition we identified ADAM10 (A Disintegrin And Metalloproteinase-10) as the protease responsible for this shedding. Surprisingly, ADAM17, the only EPCR sheddase described so far, was not involved in this process. Finally, we showed that this ADAM10-mediated shedding of EPCR induced by the meningococcal interaction with endothelial cells was responsible for an impaired activation of Protein C. This work unveils for the first time a direct link between meningococcal adhesion to endothelial cells and a severe dysregulation of coagulation, and potentially identifies new therapeutic targets for meningococcal purpura fulminans.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Endotélio Vascular/patologia , Proteínas de Membrana/metabolismo , Infecções Meningocócicas/complicações , Microvasos/patologia , Proteína C/metabolismo , Púrpura Fulminante/etiologia , Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/genética , Aderência Bacteriana , Coagulação Sanguínea/fisiologia , Células Cultivadas , Receptor de Proteína C Endotelial/genética , Endotélio Vascular/metabolismo , Endotélio Vascular/microbiologia , Humanos , Proteínas de Membrana/genética , Infecções Meningocócicas/microbiologia , Microvasos/metabolismo , Microvasos/microbiologia , Neisseria meningitidis/fisiologia , Proteína C/genética , Púrpura Fulminante/metabolismo , Púrpura Fulminante/patologia
14.
Virulence ; 8(8): 1808-1819, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29099305

RESUMO

Neisseria meningitidis is the causative agent of cerebrospinal meningitis and that of a rapidly progressing fatal septic shock known as purpura fulminans. Meningococcemia is characterized by bacterial adhesion to human endothelial cells of the microvessels. Host specificity has hampered studies on the role of blood vessels colonization in N. meningitidis associated pathogenesis. In this work, using a humanized model of SCID mice allowing the study of bacterial adhesion to human cells in an in vivo context we demonstrate that meningococcal colonization of human blood vessels is a prerequisite to the establishment of sepsis and lethality. To identify the molecular pathways involved in bacterial virulence, we performed transposon insertion site sequencing (Tn-seq) in vivo. Our results demonstrate that 36% of the genes that are important for growth in the blood of mice are dispensable when bacteria colonize human blood vessels, suggesting that human endothelial cells lining the blood vessels are feeding niches for N. meningitidis in vivo. Altogether, our work proposes a new paradigm for meningococcal virulence in which colonization of blood vessels is associated with metabolic adaptation and sustained bacteremia responsible for sepsis and subsequent lethality.


Assuntos
Bacteriemia/microbiologia , Infecções Meningocócicas/sangue , Infecções Meningocócicas/microbiologia , Microvasos/microbiologia , Neisseria meningitidis/fisiologia , Animais , Bacteriemia/sangue , Aderência Bacteriana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neisseria meningitidis/genética
15.
PLoS Pathog ; 13(7): e1006495, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28704569

RESUMO

Neisseria meningitidis is a commensal of human nasopharynx. In some circumstances, this bacteria can invade the bloodstream and, after crossing the blood brain barrier, the meninges. A filamentous phage, designated MDAΦ for Meningococcal Disease Associated, has been associated with invasive disease. In this work we show that the prophage is not associated with a higher virulence during the bloodstream phase of the disease. However, looking at the interaction of N. meningitidis with epithelial cells, a step essential for colonization of the nasopharynx, we demonstrate that the presence of the prophage, via the production of viruses, increases colonization of encapsulated meningococci onto monolayers of epithelial cells. The analysis of the biomass covering the epithelial cells revealed that meningococci are bound to the apical surface of host cells by few layers of heavily piliated bacteria, whereas, in the upper layers, bacteria are non-piliated but surrounded by phage particles which (i) form bundles of filaments, and/or (ii) are in some places associated with bacteria. The latter are likely to correspond to growing bacteriophages during their extrusion through the outer membrane. These data suggest that, as the biomass increases, the loss of piliation in the upper layers of the biomass does not allow type IV pilus bacterial aggregation, but is compensated by a large production of phage particles that promote bacterial aggregation via the formation of bundles of phage filaments linked to the bacterial cell walls. We propose that MDAΦ by increasing bacterial colonization in the mucosa at the site-of-entry, increase the occurrence of diseases.


Assuntos
Inovirus/fisiologia , Infecções Meningocócicas/microbiologia , Neisseria meningitidis/patogenicidade , Neisseria meningitidis/virologia , Animais , Aderência Bacteriana , Células Epiteliais/microbiologia , Feminino , Fímbrias Bacterianas/fisiologia , Humanos , Camundongos , Camundongos SCID , Nasofaringe/microbiologia , Neisseria meningitidis/crescimento & desenvolvimento , Neisseria meningitidis/fisiologia , Prófagos/fisiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...