Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 579
Filtrar
1.
Biomolecules ; 11(12)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34944446

RESUMO

Legionella pneumophila is a Gram-negative intracellular pathogen that causes Legionnaires' disease in elderly or immunocompromised individuals. This bacterium relies on the Dot/Icm (Defective in organelle trafficking/Intracellular multiplication) Type IV Secretion System (T4SS) and a large (>330) set of effector proteins to colonize the host cell. The structural variability of these effectors allows them to disrupt many host processes. Herein, we report the crystal structure of MavL to 2.65 Å resolution. MavL adopts an ADP-ribosyltransferase (ART) fold and contains the distinctive ligand-binding cleft of ART proteins. Indeed, MavL binds ADP-ribose with Kd of 13 µM. Structural overlay of MavL with poly-(ADP-ribose) glycohydrolases (PARGs) revealed a pair of aspartate residues in MavL that align with the catalytic glutamates in PARGs. MavL also aligns with ADP-ribose "reader" proteins (proteins that recognize ADP-ribose). Since no glycohydrolase activity was observed when incubated in the presence of ADP-ribosylated PARP1, MavL may play a role as a signaling protein that binds ADP-ribose. An interaction between MavL and the mammalian ubiquitin-conjugating enzyme UBE2Q1 was revealed by yeast two-hybrid and co-immunoprecipitation experiments. This work provides structural and molecular insights to guide biochemical studies aimed at elucidating the function of MavL. Our findings support the notion that ubiquitination and ADP-ribosylation are global modifications exploited by L. pneumophila.


Assuntos
Legionella pneumophila/crescimento & desenvolvimento , Sistemas de Secreção Tipo IV/química , Sistemas de Secreção Tipo IV/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Adenosina Difosfato Ribose/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Células HEK293 , Células HeLa , Humanos , Legionella pneumophila/enzimologia , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Células THP-1 , Ubiquitinação
2.
Cell Rep ; 37(5): 109894, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34731604

RESUMO

Legionella pneumophila grows intracellularly within a replication vacuole via action of Icm/Dot-secreted proteins. One such protein, SdhA, maintains the integrity of the vacuolar membrane, thereby preventing cytoplasmic degradation of bacteria. We show here that SdhA binds and blocks the action of OCRL (OculoCerebroRenal syndrome of Lowe), an inositol 5-phosphatase pivotal for controlling endosomal dynamics. OCRL depletion results in enhanced vacuole integrity and intracellular growth of a sdhA mutant, consistent with OCRL participating in vacuole disruption. Overexpressed SdhA alters OCRL function, enlarging endosomes, driving endosomal accumulation of phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2), and interfering with endosomal trafficking. SdhA interrupts Rab guanosine triphosphatase (GTPase)-OCRL interactions by binding to the OCRL ASPM-SPD2-Hydin (ASH) domain, without directly altering OCRL 5-phosphatase activity. The Legionella vacuole encompassing the sdhA mutant accumulates OCRL and endosomal antigen EEA1 (Early Endosome Antigen 1), consistent with SdhA blocking accumulation of OCRL-containing endosomal vesicles. Therefore, SdhA hijacking of OCRL is associated with blocking trafficking events that disrupt the pathogen vacuole.


Assuntos
Proteínas de Bactérias/metabolismo , Endossomos/enzimologia , Flavoproteínas/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/enzimologia , Macrófagos/enzimologia , Monoéster Fosfórico Hidrolases/metabolismo , Vacúolos/enzimologia , Animais , Proteínas de Bactérias/genética , Células COS , Chlorocebus aethiops , Endocitose , Endossomos/genética , Endossomos/microbiologia , Evolução Molecular , Feminino , Flavoproteínas/genética , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Legionella pneumophila/genética , Legionella pneumophila/crescimento & desenvolvimento , Doença dos Legionários/microbiologia , Macrófagos/microbiologia , Camundongos , Mutação , Fosfatidilinositol 4,5-Difosfato/metabolismo , Monoéster Fosfórico Hidrolases/genética , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Células U937 , Vacúolos/genética , Vacúolos/microbiologia , Proteínas rab de Ligação ao GTP/metabolismo
3.
Microbiol Spectr ; 9(1): e0040421, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34378969

RESUMO

Legionella pneumophila, the causative agent of Legionnaires' disease, is mostly found in man-made water systems and is one of the most closely monitored waterborne pathogens. With the aim of finding natural ways to control waterborne pathogens and thus further reduce the impact of disinfection by-products on human health, some studies have demonstrated the ability of bacteria to kill Legionella through the production of secondary metabolites or antimicrobial compounds. Here, we describe an unexpected growth inhibition of L. pneumophila when exposed to a physically separated strain of Pseudomonas fluorescens, designated as MFE01. Most of the members of the Legionellaceae family are sensitive to the volatile substances emitted by MFE01, unlike other bacteria tested. Using headspace solid-phase microextraction GC-MS strategy, a volatilome comparison revealed that emission of 1-undecene, 2-undecanone, and 2-tridecanone were mainly reduced in a Tn5-transposon mutant unable to inhibit at distance the growth of L. pneumophila strain Lens. We showed that 1-undecene was mainly responsible for the inhibition at distance in vitro, and led to cell lysis in small amounts, as determined by gas chromatography-mass spectrometry (GC-MS). Collectively, our results provide new insights into the mode of action of bacterial volatiles and highlight them as potent anti-Legionella agents to focus research on novel strategies to fight legionellosis. IMPORTANCE Microbial volatile compounds are molecules whose activities are increasingly attracting the attention of researchers. Indeed, they can act as key compounds in long-distance intrakingdom and interkingdom communication, but also as antimicrobials in competition and predation. In fact, most studies to date have focused on their antifungal activities and only a few have reported on their antibacterial properties. Here, we describe that 1-undecene, naturally produced by P. fluorescens, is a volatile with potent activity against bacteria of the genus Legionella. In small amounts, it is capable of inducing cell lysis even when the producing strain is physically separated from the target. This is the first time that such activity is described. This molecule could therefore constitute an efficient compound to counter bacterial pathogens whose treatment may fail, particularly in pulmonary diseases. Indeed, inhalation of these volatiles should be considered as a possible route of therapy in addition to antibiotic treatment.


Assuntos
Antibacterianos/farmacologia , Legionella pneumophila/efeitos dos fármacos , Pseudomonas fluorescens/metabolismo , Humanos , Legionella pneumophila/crescimento & desenvolvimento , Doença dos Legionários/terapia , Pseudomonas fluorescens/genética , Compostos Orgânicos Voláteis/análise , Compostos Orgânicos Voláteis/farmacologia
4.
Mol Microbiol ; 116(2): 624-647, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34018265

RESUMO

Legionella pneumophila possesses a unique intracellular lifecycle featuring distinct morphological stages that include replicative forms and transmissive cyst forms. Expression of genes associated with virulence traits and cyst morphogenesis is concomitant, and governed by a complex stringent response based-regulatory network and the stationary phase sigma factor RpoS. In Pseudomonas spp., rpoS expression is controlled by the autorepressor PsrA, and orthologs of PsrA and RpoS are required for cyst formation in Azotobacter. Here we report that the L. pneumophila psrA ortholog, expressed as a leaderless monocistronic transcript, is also an autorepressor, but is not a regulator of rpoS expression. Further, the binding site sequence recognized by L. pneumophila PsrA is different from that of Pseudomonas PsrA, suggesting a repertoire of target genes unique to L. pneumophila. While PsrA was dispensable for growth in human U937-derived macrophages, lack of PsrA affected bacterial intracellular growth in Acanthamoeba castellanii protozoa, but also increased the quantity of poly-3-hydroxybutyrate (PHB) inclusions in matured transmissive cysts. Interestingly, overexpression of PsrA increased the size and bacterial load of the replicative vacuole in both host cell types. Taken together, we report that PsrA is a host-specific requirement for optimal temporal progression of L. pneumophila intracellular lifecycle in A. castellanii.


Assuntos
Acanthamoeba castellanii/microbiologia , Regulação Bacteriana da Expressão Gênica/genética , Legionella pneumophila/crescimento & desenvolvimento , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Proteínas de Bactérias/genética , Humanos , Hidroxibutiratos/metabolismo , Legionella pneumophila/genética , Macrófagos/microbiologia , Poliésteres/metabolismo , Regiões Promotoras Genéticas/genética , Fator sigma/genética , Transcrição Gênica/genética
5.
Science ; 372(6545): 935-941, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33927055

RESUMO

During infection, intracellular bacterial pathogens translocate a variety of effectors into host cells that modify host membrane trafficking for their benefit. We found a self-organizing system consisting of a bacterial phosphoinositide kinase and its opposing phosphatase that formed spatiotemporal patterns, including traveling waves, to remodel host cellular membranes. The Legionella effector MavQ, a phosphatidylinositol (PI) 3-kinase, was targeted to the endoplasmic reticulum (ER). MavQ and the Legionella PI 3-phosphatase SidP, even in the absence of other bacterial components, drove rapid PI 3-phosphate turnover on the ER and spontaneously formed traveling waves that spread along ER subdomains inducing vesicle and tubule budding. Thus, bacteria can exploit a self-organizing membrane-targeting mechanism to hijack host cellular structures for survival.


Assuntos
Proteínas de Bactérias/metabolismo , Retículo Endoplasmático/metabolismo , Membranas Intracelulares/metabolismo , Legionella pneumophila/fisiologia , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animais , Proteínas de Bactérias/química , Células COS , Chlorocebus aethiops , Retículo Endoplasmático/ultraestrutura , Retroalimentação Fisiológica , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Membranas Intracelulares/ultraestrutura , Legionella pneumophila/enzimologia , Legionella pneumophila/genética , Legionella pneumophila/crescimento & desenvolvimento , Camundongos , Mutação , Fosfatidilinositol 3-Quinase/química , Fosfatos de Fosfatidilinositol/química , Monoéster Fosfórico Hidrolases/metabolismo , Domínios Proteicos , Células RAW 264.7
6.
Pathog Dis ; 79(4)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33734371

RESUMO

The human pulmonary environment is complex, containing a matrix of cells, including fibroblasts, epithelial cells, interstitial macrophages, alveolar macrophages and neutrophils. When confronted with foreign material or invading pathogens, these cells mount a robust response. Nevertheless, many bacterial pathogens with an intracellular lifecycle stage exploit this environment for replication and survival. These include, but are not limited to, Coxiella burnetii, Legionella pneumophila, Yersinia pestis, Mycobacterium tuberculosis and Staphylococcus aureus. Currently, few human disease-relevant model systems exist for studying host-pathogen interactions during these bacterial infections in the lung. Here, we present two novel infection platforms, human alveolar macrophages (hAMs) and human precision-cut lung slices (hPCLS), along with an up-to-date synopsis of research using said models. Additionally, alternative uses for these systems in the absence of pathogen involvement are presented, such as tissue banking and further characterization of the human lung environment. Overall, hAMs and hPCLS allow novel human disease-relevant investigations that other models, such as cell lines and animal models, cannot completely provide.


Assuntos
Infecções Bacterianas/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Pneumopatias/microbiologia , Pulmão/microbiologia , Macrófagos Alveolares/microbiologia , Modelos Biológicos , Infecções Bacterianas/imunologia , Infecções Bacterianas/patologia , Coxiella burnetii/crescimento & desenvolvimento , Coxiella burnetii/imunologia , Coxiella burnetii/patogenicidade , Humanos , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/imunologia , Legionella pneumophila/patogenicidade , Pulmão/imunologia , Pulmão/patologia , Pneumopatias/imunologia , Pneumopatias/patologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/patologia , Microtomia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Cultura Primária de Células , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Bancos de Tecidos , Técnicas de Cultura de Tecidos , Yersinia pestis/crescimento & desenvolvimento , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
7.
Cell Microbiol ; 23(5): e13318, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33583106

RESUMO

Dictyostelium discoideum Sey1 is the single ortholog of mammalian atlastin 1-3 (ATL1-3), which are large homodimeric GTPases mediating homotypic fusion of endoplasmic reticulum (ER) tubules. In this study, we generated a D. discoideum mutant strain lacking the sey1 gene and found that amoebae deleted for sey1 are enlarged, but grow and develop similarly to the parental strain. The ∆sey1 mutant amoebae showed an altered ER architecture, and the tubular ER network was partially disrupted without any major consequences for other organelles or the architecture of the secretory and endocytic pathways. Macropinocytic and phagocytic functions were preserved; however, the mutant amoebae exhibited cumulative defects in lysosomal enzymes exocytosis, intracellular proteolysis, and cell motility, resulting in impaired growth on bacterial lawns. Moreover, ∆sey1 mutant cells showed a constitutive activation of the unfolded protein response pathway (UPR), but they still readily adapted to moderate levels of ER stress, while unable to cope with prolonged stress. In D. discoideum ∆sey1 the formation of the ER-associated compartment harbouring the bacterial pathogen Legionella pneumophila was also impaired. In the mutant amoebae, the ER was less efficiently recruited to the "Legionella-containing vacuole" (LCV), the expansion of the pathogen vacuole was inhibited at early stages of infection and intracellular bacterial growth was reduced. In summary, our study establishes a role of D. discoideum Sey1 in ER architecture, proteolysis, cell motility and intracellular replication of L. pneumophila.


Assuntos
Dictyostelium/fisiologia , Retículo Endoplasmático/ultraestrutura , GTP Fosfo-Hidrolases/metabolismo , Legionella pneumophila/fisiologia , Proteínas de Protozoários/metabolismo , Vacúolos/microbiologia , Dictyostelium/crescimento & desenvolvimento , Dictyostelium/microbiologia , Dictyostelium/ultraestrutura , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Retículo Endoplasmático Rugoso/microbiologia , Retículo Endoplasmático Rugoso/fisiologia , GTP Fosfo-Hidrolases/genética , Homeostase , Interações Hospedeiro-Patógeno , Legionella pneumophila/crescimento & desenvolvimento , Movimento , Muramidase/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas de Protozoários/genética , Vacúolos/fisiologia
8.
Commun Biol ; 4(1): 157, 2021 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-33542442

RESUMO

Catalytically inactive dCas9 imposes transcriptional gene repression by sterically precluding RNA polymerase activity at a given gene to which it was directed by CRISPR (cr)RNAs. This gene silencing technology, known as CRISPR interference (CRISPRi), has been employed in various bacterial species to interrogate genes, mostly individually or in pairs. Here, we developed a multiplex CRISPRi platform in the pathogen Legionella pneumophila capable of silencing up to ten genes simultaneously. Constraints on precursor-crRNA expression were overcome by combining a strong promoter with a boxA element upstream of a CRISPR array. Using crRNAs directed against virulence protein-encoding genes, we demonstrated that CRISPRi is fully functional not only during growth in axenic media, but also during macrophage infection, and that gene depletion by CRISPRi recapitulated the growth defect of deletion strains. By altering the position of crRNA-encoding spacers within the CRISPR array, our platform achieved the gradual depletion of targets that was mirrored by the severity in phenotypes. Multiplex CRISPRi thus holds great promise for probing large sets of genes in bulk in order to decipher virulence strategies of L. pneumophila and other bacterial pathogens.


Assuntos
Proteínas de Bactérias/genética , Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Inativação Gênica , Legionella pneumophila/genética , Fatores de Virulência/genética , Proteínas de Bactérias/metabolismo , Proteína 9 Associada à CRISPR/metabolismo , Regulação Bacteriana da Expressão Gênica , Humanos , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidade , Estudo de Prova de Conceito , Células U937 , Virulência/genética , Fatores de Virulência/metabolismo
9.
Cell Microbiol ; 23(5): e13313, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33491325

RESUMO

ProA is a secreted zinc metalloprotease of Legionella pneumophila causing lung damage in animal models of Legionnaires' disease. Here we demonstrate that ProA promotes infection of human lung tissue explants (HLTEs) and dissect the contribution to cell type specific replication and extracellular virulence mechanisms. For the first time, we reveal that co-incubation of HLTEs with purified ProA causes a significant increase of the alveolar septal thickness. This destruction of connective tissue fibres was further substantiated by collagen IV degradation assays. The moderate attenuation of a proA-negative mutant in A549 epithelial cells and THP-1 macrophages suggests that effects of ProA in tissue mainly result from extracellular activity. Correspondingly, ProA contributes to dissemination and serum resistance of the pathogen, which further expands the versatile substrate spectrum of this thermolysin-like protease. The crystal structure of ProA at 1.48 Å resolution showed high congruence to pseudolysin of Pseudomonas aeruginosa, but revealed deviations in flexible loops, the substrate binding pocket S1 ' and the repertoire of cofactors, by which ProA can be distinguished from respective homologues. In sum, this work specified virulence features of ProA at different organisational levels by zooming in from histopathological effects in human lung tissue to atomic details of the protease substrate determination.


Assuntos
Proteínas de Bactérias/metabolismo , Colágeno Tipo IV/metabolismo , Legionella pneumophila/enzimologia , Legionella pneumophila/patogenicidade , Pulmão/microbiologia , Metaloendopeptidases/metabolismo , Alvéolos Pulmonares/patologia , Fatores de Virulência/metabolismo , Células A549 , Proteínas de Bactérias/química , Atividade Bactericida do Sangue , Humanos , Legionella pneumophila/crescimento & desenvolvimento , Pulmão/patologia , Metaloendopeptidases/química , Proteólise , Alvéolos Pulmonares/metabolismo , Células THP-1 , Virulência , Fatores de Virulência/química
10.
Chemistry ; 27(7): 2506-2512, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33075184

RESUMO

Legionnaires' disease is caused by infection with the intracellularly replicating Gram-negative bacterium Legionella pneumophila. This pathogen uses an unconventional way of ubiquitinating host proteins by generating a phosphoribosyl linkage between substrate proteins and ubiquitin by making use of an ADPribosylated ubiquitin (UbADPr ) intermediate. The family of SidE effector enzymes that catalyze this reaction is counteracted by Legionella hydrolases, which are called Dups. This unusual ubiquitination process is important for Legionella proliferation and understanding these processes on a molecular level might prove invaluable in finding new treatments. Herein, a modular approach is used for the synthesis of triazole-linked UbADPr , and analogues thereof, and their affinity towards the hydrolase DupA is determined and hydrolysis rates are compared to natively linked UbADPr . The inhibitory effects of modified Ub on the canonical eukaryotic E1-enzyme Uba1 are investigated and rationalized in the context of a high-resolution crystal structure reported herein. Finally, it is shown that synthetic UbADPr analogues can be used to effectively pull-down overexpressed DupA from cell lysate.


Assuntos
ADP-Ribosilação , Legionella pneumophila/enzimologia , Doença dos Legionários/microbiologia , Ubiquitina/química , Ubiquitina/metabolismo , Proteínas de Bactérias/metabolismo , Células HEK293 , Humanos , Hidrolases/metabolismo , Legionella pneumophila/crescimento & desenvolvimento , Enzimas Ativadoras de Ubiquitina/metabolismo , Ubiquitinação
11.
Biocontrol Sci ; 25(3): 179-182, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32938848

RESUMO

Testing for Legionella spp. in public bath water samples is regulated in Japan. In this study, we used a total of 132 public bath water samples to compare the performance of Legiolert® and the conventional plate culture method for the enumeration of Legionella pneumophila. When Legiolert and plate culturing were performed at the same detection limit, L. pneumophila was detected in 26.5% of 132 samples by Legiolert, while 12.9% contained Legionella spp. (11.4% contained L. pneumophila) based on the plate culture method. Moreover, results of 83.3% of the total samples were consistent between the two methods, meaning that they were both positive or both negative. In this study, we demonstrated that Legiolert is a simpler and more effective method of monitoring for L. pneumophila in bath water samples.


Assuntos
Técnicas Bacteriológicas , Legionella pneumophila/crescimento & desenvolvimento , Microbiologia da Água , Carga Bacteriana/métodos , Legionella pneumophila/isolamento & purificação
12.
FEMS Microbiol Lett ; 367(18)2020 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-32860684

RESUMO

Free-living amoebae are known to act as replication niches for the pathogenic bacterium Legionella pneumophila in freshwater environments. However, we previously reported that some strains of the Willaertia magna species are more resistant to L. pneumophila infection and differ in their ability to support its growth. From this observation, we hypothesize that L. pneumophila growth in environment could be partly dependent on the composition of amoebic populations and on the possible interactions between different amoebic species. We tested this hypothesis by studying the growth of L. pneumophila and of a permissive free-living amoeba, Vermamoeba vermiformis (formerly named Hartmannella vermiformis), in co-culture with or without other free-living amoebae (Acanthamoeba castellanii and W. magna). We demonstrate the occurrence of inter-amoebic phagocytosis with A. castellanii and W. magna being able to ingest V. vermiformis infected or not infected with L. pneumophila. We also found that L. pneumophila growth is strongly impacted by the permissiveness of each interactive amoeba demonstrating that L. pneumophila proliferation and spread are controlled, at least in part, by inter-amoebic interactions.


Assuntos
Amébidos/microbiologia , Legionella pneumophila/crescimento & desenvolvimento , Fagocitose , Amébidos/classificação , Amébidos/crescimento & desenvolvimento , Técnicas de Cocultura , Interações entre Hospedeiro e Microrganismos , Doença dos Legionários/transmissão , Microbiologia da Água
13.
Int J Mol Sci ; 21(15)2020 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-32731616

RESUMO

Legionella pneumophila is an environmental bacterium, an opportunistic premise plumbing pathogen that causes the Legionnaires' disease. L. pneumophila presents a serious health hazard in building water systems, due to its high resistance to standard water disinfection methods. Our aim was to study the use of photodynamic inactivation (PDI) against Legionella. We investigated and compared the photobactericidal potential of five cationic dyes. We tested toluidine blue (TBO) and methylene blue (MB), and three 3-N-methylpyridylporphyrins, one tetra-cationic and two tri-cationic, one with a short (CH3) and the other with a long (C17H35) alkyl chain, against L. pneumophila in tap water and after irradiation with violet light. All tested dyes demonstrated a certain dark toxicity against L. pneumophila; porphyrins with lower minimal effective concentration (MEC) values than TBO and MB. Nanomolar MEC values, significantly lower than with TBO and MB, were obtained with all three porphyrins in PDI experiments, with amphiphilic porphyrin demonstrating the highest PDI activity. All tested dyes showed increasing PDI with longer irradiation (0-108 J/cm2), especially the two hydrophilic porphyrins. All three porphyrins caused significant changes in cell membrane permeability after irradiation and L. pneumophila, co-cultivated with Acanthamoeba castellanii after treatment with all three porphyrins and irradiation, did not recover in amoeba. We believe our results indicate the considerable potential of cationic porphyrins as effective anti-Legionella agents.


Assuntos
Antibacterianos , Legionella pneumophila/crescimento & desenvolvimento , Doença dos Legionários/tratamento farmacológico , Fotoquimioterapia , Porfirinas , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Doença dos Legionários/metabolismo , Porfirinas/síntese química , Porfirinas/química , Porfirinas/farmacologia
14.
Int J Infect Dis ; 97: 374-379, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32534142

RESUMO

OBJECTIVES: To report atypical pathogens from clinical trial data comparing delafloxacin to moxifloxacin in the treatment of adults with community-acquired bacterial pneumonia (CABP). METHODS: Multiple diagnostic methods were employed to diagnose atypical infections including culture, serology, and urinary antigen. RESULTS: The microbiological intent-to-treat (MITT) population included 520 patients; 30% had an atypical bacterial pathogen identified (156/520). Overall, 13.1% (68/520) had a monomicrobial atypical infection and 2.3% (12/520) had polymicrobial all-atypical infections. Among patients with polymicrobial infections, Streptococcus pneumoniae was the most frequently occurring co-infecting organism and Chlamydia pneumoniae was the most frequently occurring co-infecting atypical organism. For Mycoplasma pneumoniae and Legionella pneumophila, serology yielded the highest number of diagnoses. Delafloxacin and moxifloxacin had similar in vitro activity against M. pneumoniae and delafloxacin had greater activity against L. pneumophila. Two macrolide-resistant M. pneumoniae isolates were recovered. No fluoroquinolone-resistant M. pneumoniae were isolated. The rates of microbiological success (documented or presumed eradication) at test-of-cure were similar between the delafloxacin and moxifloxacin groups. There was no evidence of a correlation between minimum inhibitory concentration (MIC) and outcome; a high proportion of favorable outcomes was observed across all delafloxacin baseline MICs. CONCLUSIONS: Delafloxacin may be considered a treatment option as monotherapy for CABP in adults, where broad-spectrum coverage including atypical activity is desirable.


Assuntos
Antibacterianos/administração & dosagem , Infecções Comunitárias Adquiridas/tratamento farmacológico , Fluoroquinolonas/administração & dosagem , Moxifloxacina/administração & dosagem , Pneumonia Bacteriana/tratamento farmacológico , Adulto , Infecções Comunitárias Adquiridas/microbiologia , Feminino , Humanos , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/isolamento & purificação , Macrolídeos/administração & dosagem , Masculino , Testes de Sensibilidade Microbiana , Mycoplasma pneumoniae/efeitos dos fármacos , Mycoplasma pneumoniae/crescimento & desenvolvimento , Mycoplasma pneumoniae/isolamento & purificação , Pneumonia Bacteriana/microbiologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/isolamento & purificação , Adulto Jovem
15.
FEMS Microbiol Lett ; 367(7)2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32188994

RESUMO

Polyphenols derived from a variety of plants have demonstrated antimicrobial activity against diverse microbial pathogens. Legionella pneumophila is an intracellular bacterial pathogen that opportunistically causes a severe inflammatory pneumonia in humans, called Legionnaires' Disease, via replication within macrophages. Previous studies demonstrated that tea polyphenols attenuate L. pneumophila intracellular replication within mouse macrophages via increased tumor necrosis factor (TNF) production. Sorghum bicolor is a sustainable cereal crop that thrives in arid environments and is well-suited to continued production in warming climates. Sorghum polyphenols have anticancer and antioxidant properties, but their antimicrobial activity has not been evaluated. Here, we investigated the impact of sorghum polyphenols on L. pneumophila intracellular replication within RAW 264.7 mouse macrophages. Sorghum high-polyphenol extract (HPE) attenuated L. pneumophila intracellular replication in a dose-dependent manner but did not impair either bacterial replication in rich media or macrophage viability. Moreover, HPE treatment enhanced both TNF and IL-6 secretion from L. pneumophila infected macrophages. Thus, polyphenols derived from sorghum enhance macrophage restriction of L. pneumophila, likely via increased pro-inflammatory cytokine production. This work reveals commonalities between plant polyphenol-mediated antimicrobial activity and provides a foundation for future evaluation of sorghum as an antimicrobial agent.


Assuntos
Legionella pneumophila/efeitos dos fármacos , Macrófagos/microbiologia , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Sorghum/química , Animais , Legionella pneumophila/crescimento & desenvolvimento , Camundongos , Células RAW 264.7
16.
Cell Microbiol ; 22(4): e13151, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32096265

RESUMO

Legionella pneumophila requires the Dot/Icm translocation system to replicate in a vacuolar compartment within host cells. Strains lacking the translocated substrate SdhA form a permeable vacuole during residence in the host cell, exposing bacteria to the host cytoplasm. In primary macrophages, mutants are defective for intracellular growth, with a pyroptotic cell death response mounted due to bacterial exposure to the cytosol. To understand how SdhA maintains vacuole integrity during intracellular growth, we performed high-throughput RNAi screens against host membrane trafficking genes to identify factors that antagonise vacuole integrity in the absence of SdhA. Depletion of host proteins involved in endocytic uptake and recycling resulted in enhanced intracellular growth and lower levels of permeable vacuoles surrounding the ΔsdhA mutant. Of interest were three different Rab GTPases involved in these processes: Rab11b, Rab8b and Rab5 isoforms, that when depleted resulted in enhanced vacuole integrity surrounding the sdhA mutant. Proteins regulated by these Rabs are responsible for interfering with proper vacuole membrane maintenance, as depletion of the downstream effectors EEA1, Rab11FIP1, or VAMP3 rescued vacuole integrity and intracellular growth of the sdhA mutant. To test the model that specific vesicular components associated with these effectors could act to destabilise the replication vacuole, EEA1 and Rab11FIP1 showed increased density about the sdhA mutant vacuole compared with the wild type (WT) vacuole. Depletion of Rab5 isoforms or Rab11b reduced this aberrant redistribution. These findings are consistent with SdhA interfering with both endocytic and recycling membrane trafficking events that act to destabilise vacuole integrity during infection.


Assuntos
Citosol/microbiologia , Endocitose , Interações Hospedeiro-Patógeno , Legionella pneumophila/crescimento & desenvolvimento , Vacúolos/microbiologia , Vacúolos/patologia , Animais , Proteínas de Bactérias/genética , Transporte Biológico , Feminino , Flavoproteínas/genética , Macrófagos/microbiologia , Camundongos , Transporte Proteico , Células RAW 264.7 , Interferência de RNA
17.
PLoS One ; 15(1): e0227574, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31940328

RESUMO

Legionella pneumophila can cause a potentially fatal form of humane pneumonia (Legionnaires' disease), which is most problematic in immunocompromised and in elderly people. Legionella species is present at low concentrations in soil, natural and artificial aquatic systems and is therefore constantly entering man-made water systems. The environment temperature for it's ideal growth range is between 32 and 42°C, thus hot water pipes represent ideal environment for spread of Legionella. The bacteria are dormant below 20°C and do not survive above 60°C. The primary method used to control the risk from Legionella is therefore water temperature control. There are several other effective treatments to prevent growth of Legionella in water systems, however current disinfection methods can be applied only intermittently thus allowing Legionella to grow in between treatments. Here we present an alternative disinfection method based on antibacterial coatings with Cu-TiO2 nanotubes deposited on preformed surfaces. In the experiment the microbiocidal efficiency of submicron coatings on polystyrene to the bacterium of the genus Legionella pneumophila with a potential use in a water supply system was tested. The treatment thus constantly prevents growth of Legionella pneumophila in presence of water at room temperature. Here we show that 24-hour illumination with low power UVA light source (15 W/m2 UVA illumination) of copper doped TiO2 nanotube coated surfaces is effective in preventing growth of Legionella pneumophila. Microbiocidal effects of Cu-TiO2 nanotube coatings were dependent on the flow of the medium and the intensity of UV-A light. It was determined that tested submicron coatings have microbiocidal effects specially in a non-flow or low-flow conditions, as in higher flow rates, probably to a greater possibility of Legionella pneumophila sedimentation on the coated polystyrene surfaces, meanwhile no significant differences among bacteria reduction was noted regarding to non or low flow of medium.


Assuntos
Cobre/química , Cobre/farmacologia , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/efeitos da radiação , Nanotubos/química , Titânio/química , Raios Ultravioleta , Antibacterianos/química , Antibacterianos/farmacologia , Catálise , Legionella pneumophila/crescimento & desenvolvimento , Processos Fotoquímicos , Propriedades de Superfície
18.
Nat Microbiol ; 5(4): 599-609, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31988381

RESUMO

Virulence mechanisms typically evolve through the continual interaction of a pathogen with its host. In contrast, it is poorly understood how environmentally acquired pathogens are able to cause disease without prior interaction with humans. Here, we provide experimental evidence for the model that Legionella pathogenesis in humans results from the cumulative selective pressures of multiple amoebal hosts in the environment. Using transposon sequencing, we identify Legionella pneumophila genes required for growth in four diverse amoebae, defining universal virulence factors commonly required in all host cell types and amoeba-specific auxiliary genes that determine host range. By comparing genes that promote growth in amoebae and macrophages, we show that adaptation of L. pneumophila to each amoeba causes the accumulation of distinct virulence genes that collectively allow replication in macrophages and, in some cases, leads to redundancy in this host cell type. In contrast, some bacterial proteins that promote replication in amoebae restrict growth in macrophages. Thus, amoebae-imposed selection is a double-edged sword, having both positive and negative impacts on disease. Comparing the genome composition and host range of multiple Legionella species, we demonstrate that their distinct evolutionary trajectories in the environment have led to the convergent evolution of compensatory virulence mechanisms.


Assuntos
Amoeba/microbiologia , Coevolução Biológica , Interações Hospedeiro-Patógeno/genética , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Macrófagos/microbiologia , Fatores de Virulência/genética , Adaptação Fisiológica , Amoeba/classificação , Animais , Proteínas de Bactérias/classificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Feminino , Especificidade de Hospedeiro , Humanos , Legionella pneumophila/classificação , Legionella pneumophila/crescimento & desenvolvimento , Camundongos , Filogenia , Cultura Primária de Células , Seleção Genética , Virulência , Fatores de Virulência/classificação , Fatores de Virulência/metabolismo
19.
J Leukoc Biol ; 107(2): 273-284, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31793076

RESUMO

Legionella pneumophila is an opportunistic human pathogen and causative agent of the acute pneumonia known as Legionnaire's disease. Upon inhalation, the bacteria replicate in alveolar macrophages (AM), within an intracellular vacuole termed the Legionella-containing vacuole. We recently found that, in vivo, IFNγ was required for optimal clearance of intracellular L. pneumophila by monocyte-derived cells (MC), but the cytokine did not appear to influence clearance by AM. Here, we report that during L. pneumophila lung infection, expression of the IFNγ receptor subunit 1 (IFNGR1) is down-regulated in AM and neutrophils, but not MC, offering a possible explanation for why AM are unable to effectively restrict L. pneumophila replication in vivo. To test this, we used mice that constitutively express IFNGR1 in AM and found that prevention of IFNGR1 down-regulation enhanced the ability of AM to restrict L. pneumophila intracellular replication. IFNGR1 down-regulation was independent of the type IV Dot/Icm secretion system of L. pneumophila indicating that bacterial effector proteins were not involved. In contrast to previous work, we found that signaling via type I IFN receptors was not required for IFNGR1 down-regulation in macrophages but rather that MyD88- or Trif- mediated NF-κB activation was required. This work has uncovered an alternative signaling pathway responsible for IFNGR1 down-regulation in macrophages during bacterial infection.


Assuntos
Legionella pneumophila/crescimento & desenvolvimento , Doença dos Legionários/microbiologia , Pulmão/microbiologia , Macrófagos Alveolares/microbiologia , NF-kappa B/metabolismo , Receptores de Interferon/antagonistas & inibidores , Animais , Regulação para Baixo , Interferon Tipo I/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/metabolismo , Pulmão/metabolismo , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Transgênicos , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Transdução de Sinais , Receptor de Interferon gama
20.
Mucosal Immunol ; 12(6): 1382-1390, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31462698

RESUMO

Legionella pneumophila (Lp) is a flagellated, intracellular bacterium that can cause Legionnaires' disease (LD). Lp activates multiple innate immune receptors, and TOLLIP dampens MyD88-dependent signaling and may influence susceptibility to LD. We evaluated the effect of TOLLIP on innate immunity, pneumonia severity, and LD susceptibility in mouse lungs and human populations. To accomplish this, we evaluated the effect of TOLLIP on lung-specific Lp control and immune response and associated a common functional TOLLIP variant with Lp-induced innate immune responses and LD susceptibility in humans. After aerosol Lp infection, Tollip-/- mice demonstrated significantly fewer bacterial colony-forming unit and increased cytokine responses from BAL fluid. Tollip-/- macrophages also suppressed intracellular Lp replication in a flagellin-independent manner. The presence of a previously characterized, functionally active SNP associated with decreased TOLLIP mRNA transcript in monocytes was associated with increased TNF and IL-6 secretion after Lp stimulation of PBMC ex vivo. This genotype was separately associated with decreased LD susceptibility (309 controls, 88 cases, p = 0.008, OR 0.36, 95% CI 0.16-0.76) in a candidate gene association study. These results suggest that TOLLIP decreases lung-specific TLR responses to increase LD susceptibility in human populations. Better understanding of TOLLIP may lead to novel immunomodulatory therapies.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Legionella pneumophila/patogenicidade , Doença dos Legionários/metabolismo , Pulmão/metabolismo , Adulto , Idoso , Animais , Carga Bacteriana , Estudos de Casos e Controles , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/imunologia , Doença dos Legionários/genética , Doença dos Legionários/imunologia , Doença dos Legionários/microbiologia , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...