Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Sci Total Environ ; 927: 172410, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38608884

RESUMO

There is little evidence of the long-term consequences of maintaining sanitary hot water at high temperatures on the persistence of Legionella in the plumbing system. The aims of this study were to describe the persistence and genotypic variability of L. pneumophila in a hospital building with two entirely independent hot water distribution systems, and to estimate the thermotolerance of the genotypic variants by studying the quantity of VBNC L. pneumophila. Eighty isolates from 55 water samples obtained between the years 2012-2017 were analyzed. All isolates correspond to L. pneumophila serogroup 6. The isolates were discriminated in four restriction patterns by pulsed-field gel electrophoresis. In one installation, pattern A + Aa predominated, accounting for 75.8 % of samples, while the other installation exhibited pattern B as the most frequent (81.8 % of samples; p < 0.001). The mean temperature of the isolates was: 52.6 °C (pattern A + Aa) and 55.0 °C (pattern B), being significantly different. Nine strains were selected as representative among patterns to study their thermotolerance by flow-cytometry after 24 h of thermic treatment. VBNC bacteria were detected in all samples. After thermic treatment at 50 °C, 52.0 % of bacteria had an intact membrane, and after 55 °C this percentage decreased to 23.1 %. Each pattern exhibited varying levels of thermotolerance. These findings indicate that the same hospital building can be colonized with different predominant types of Legionella if it has independent hot water installations. Maintaining a minimum temperature of 50 °C at distal points of the system would allow the survival of replicative L. pneumophila. However, the presence of Legionella in hospital water networks is underestimated if culture is considered as the standard method for Legionella detection, because VBNC do not grow on culture plates. This phenomenon can carry implications for the Legionella risk management plans in hospitals that adjust their control measures based on the microbiological surveillance of water.


Assuntos
Infecção Hospitalar , Hospitais , Legionella pneumophila , Doença dos Legionários , Viabilidade Microbiana , Abastecimento de Água , Infecção Hospitalar/microbiologia , Temperatura Alta , Legionella pneumophila/classificação , Legionella pneumophila/citologia , Legionella pneumophila/genética , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/isolamento & purificação , Termotolerância , Fatores de Tempo , Doença dos Legionários/microbiologia , Doença dos Legionários/transmissão , Contagem de Colônia Microbiana , Humanos
2.
mBio ; 12(5): e0218021, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34634944

RESUMO

Legionella pneumophila, the causative agent of Legionnaires' disease, is a facultative intracellular pathogen that survives inside phagocytic host cells by establishing a protected replication niche, termed the "Legionella-containing vacuole" (LCV). To form an LCV and subvert pivotal host pathways, L. pneumophila employs a type IV secretion system (T4SS), which translocates more than 300 different effector proteins into the host cell. The L. pneumophila T4SS complex has been shown to span the bacterial cell envelope at the bacterial poles. However, the interactions between the T4SS and the LCV membrane are not understood. Using cryo-focused ion beam milling, cryo-electron tomography, and confocal laser scanning fluorescence microscopy, we show that up to half of the intravacuolar L. pneumophila bacteria tether their cell pole to the LCV membrane. Tethering coincides with the presence and function of T4SSs and likely promotes the establishment of distinct contact sites between T4SSs and the LCV membrane. Contact sites are characterized by indentations in the limiting LCV membrane and localize juxtaposed to T4SS machineries. The data are in agreement with the notion that effector translocation occurs by close membrane contact rather than by an extended pilus. Our findings provide novel insights into the interactions of the L. pneumophila T4SS with the LCV membrane in situ. IMPORTANCE Legionnaires' disease is a life-threatening pneumonia, which is characterized by high fever, coughing, shortness of breath, muscle pain, and headache. The disease is caused by the amoeba-resistant bacterium L. pneumophila found in various soil and aquatic environments and is transmitted to humans via the inhalation of small bacteria-containing droplets. An essential virulence factor of L. pneumophila is a so-called "type IV secretion system" (T4SS), which, by injecting a plethora of "effector proteins" into the host cell, determines pathogen-host interactions and the formation of a distinct intracellular compartment, the "Legionella-containing vacuole" (LCV). It is unknown how the T4SS makes contact to the LCV membrane to deliver the effectors. In this study, we identify indentations in the host cell membrane in close proximity to functional T4SSs localizing at the bacterial poles. Our work reveals first insights into the architecture of Legionella-LCV contact sites.


Assuntos
Proteínas de Bactérias/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/microbiologia , Sistemas de Secreção Tipo IV/metabolismo , Vacúolos/microbiologia , Proteínas de Bactérias/genética , Polaridade Celular , Humanos , Legionella pneumophila/citologia , Legionella pneumophila/genética , Transporte Proteico , Sistemas de Secreção Tipo IV/genética
3.
EMBO J ; 38(14): e100957, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31304634

RESUMO

The self-assembly of cellular macromolecular machines such as the bacterial flagellar motor requires the spatio-temporal synchronization of gene expression with proper protein localization and association of dozens of protein components. In Salmonella and Escherichia coli, a sequential, outward assembly mechanism has been proposed for the flagellar motor starting from the inner membrane, with the addition of each new component stabilizing the previous one. However, very little is known about flagellar disassembly. Here, using electron cryo-tomography and sub-tomogram averaging of intact Legionella pneumophila, Pseudomonas aeruginosa, and Shewanella oneidensis cells, we study flagellar motor disassembly and assembly in situ. We first show that motor disassembly results in stable outer membrane-embedded sub-complexes. These sub-complexes consist of the periplasmic embellished P- and L-rings, and bend the membrane inward while it remains apparently sealed. Additionally, we also observe various intermediates of the assembly process including an inner-membrane sub-complex consisting of the C-ring, MS-ring, and export apparatus. Finally, we show that the L-ring is responsible for reshaping the outer membrane, a crucial step in the flagellar assembly process.


Assuntos
Bactérias/citologia , Proteínas de Bactérias/metabolismo , Flagelos/ultraestrutura , Bactérias/metabolismo , Bactérias/ultraestrutura , Membrana Externa Bacteriana/metabolismo , Tomografia com Microscopia Eletrônica , Escherichia coli/citologia , Escherichia coli/metabolismo , Escherichia coli/ultraestrutura , Flagelos/metabolismo , Legionella pneumophila/citologia , Legionella pneumophila/metabolismo , Legionella pneumophila/ultraestrutura , Pseudomonas aeruginosa/citologia , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/ultraestrutura , Shewanella/citologia , Shewanella/metabolismo , Shewanella/ultraestrutura
4.
Nature ; 572(7769): 387-391, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31330531

RESUMO

The bacterial pathogen Legionella pneumophila creates an intracellular niche permissive for its replication by extensively modulating host-cell functions using hundreds of effector proteins delivered by its Dot/Icm secretion system1. Among these, members of the SidE family (SidEs) regulate several cellular processes through a unique phosphoribosyl ubiquitination mechanism that bypasses the canonical ubiquitination machinery2-4. The activity of SidEs is regulated by another Dot/Icm effector known as SidJ5; however, the mechanism of this regulation is not completely understood6,7. Here we demonstrate that SidJ inhibits the activity of SidEs by inducing the covalent attachment of glutamate moieties to SdeA-a member of the SidE family-at E860, one of the catalytic residues that is required for the mono-ADP-ribosyltransferase activity involved in ubiquitin activation2. This inhibition by SidJ is spatially restricted in host cells because its activity requires the eukaryote-specific protein calmodulin (CaM). We solved a structure of SidJ-CaM in complex with AMP and found that the ATP used in this reaction is cleaved at the α-phosphate position by SidJ, which-in the absence of glutamate or modifiable SdeA-undergoes self-AMPylation. Our results reveal a mechanism of regulation in bacterial pathogenicity in which a glutamylation reaction that inhibits the activity of virulence factors is activated by host-factor-dependent acyl-adenylation.


Assuntos
Calmodulina/metabolismo , Ácido Glutâmico/metabolismo , Legionella pneumophila/enzimologia , Legionella pneumophila/metabolismo , Ubiquitinação , ADP-Ribosilação , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Catálise , Domínio Catalítico , Coenzimas/metabolismo , Células HEK293 , Humanos , Legionella pneumophila/citologia , Modelos Moleculares , Ubiquitina/química , Ubiquitina/metabolismo
5.
Nat Microbiol ; 4(7): 1173-1182, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31011165

RESUMO

Legionella pneumophila survives and replicates inside host cells by secreting ~300 effectors through the defective in organelle trafficking (Dot)/intracellular multiplication (Icm) type IVB secretion system (T4BSS). Here, we used complementary electron cryotomography and immunofluorescence microscopy to investigate the molecular architecture and biogenesis of the Dot/Icm secretion apparatus. Electron cryotomography mapped the location of the core and accessory components of the Legionella core transmembrane subcomplex, revealing a well-ordered central channel that opens into a large, windowed secretion chamber with an unusual 13-fold symmetry. Immunofluorescence microscopy deciphered an early-stage assembly process that begins with the targeting of Dot/Icm components to the bacterial poles. Polar targeting of this T4BSS is mediated by two Dot/Icm proteins, DotU and IcmF, that, interestingly, are homologues of the T6SS membrane complex components TssL and TssM, suggesting that the Dot/Icm T4BSS is a hybrid system. Together, these results revealed that the Dot/Icm complex assembles in an 'axial-to-peripheral' pattern.


Assuntos
Legionella pneumophila/química , Sistemas de Secreção Tipo IV/metabolismo , Sistemas de Secreção Tipo IV/ultraestrutura , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Membrana Celular/química , Membrana Celular/ultraestrutura , Polaridade Celular , Tomografia com Microscopia Eletrônica , Legionella pneumophila/citologia , Legionella pneumophila/genética , Legionella pneumophila/ultraestrutura , Microscopia de Fluorescência , Mutação , Periplasma/química , Periplasma/ultraestrutura , Multimerização Proteica , Sistemas de Secreção Tipo IV/química
6.
Autophagy ; 13(8): 1467-1469, 2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28598235

RESUMO

RavZ, an effector protein of pathogenic Legionella pneumophila, inhibits host macroautophagy/autophagy by deconjugation of lipidated LC3 proteins from phosphatidylethanolamine (PE) on the autophagosome membrane. The mechanism for how RavZ specifically recognizes and deconjugates the lipidated LC3s is not clear. To understand the structure-function relationship of LC3-deconjugation by RavZ, we prepared semisynthetic LC3 proteins modified with different fragments of PE or 1-hexadecanol (C16). We find that RavZ activity is strictly dependent on the conjugated PE structure and RavZ extracts LC3-PE from the membrane before deconjugation. Structural and biophysical analysis of RavZ-LC3 interactions suggest that RavZ initially recognizes LC3-PE on the membrane via its N-terminal LC3-interacting region (LIR) motif. RavZ specifically targets to autophagosome membranes by interaction with phosphatidylinositol 3-phosphate (PtdIns3P) via its C-terminal domain and association with membranes via the hydrophobic α3 helix. The α3 helix is involved in extraction of the PE moiety and docking of the fatty acid chains into the lipid-binding site of RavZ, which is related in structure to that of the phospholipid transfer protein Sec14. The LIR interaction and lipid binding facilitate subsequent proteolytic cleavage of LC3-PE. The findings reveal a novel mode of host-pathogen interaction.


Assuntos
Autofagia , Legionella pneumophila/citologia , Autofagossomos/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Legionella pneumophila/metabolismo , Modelos Biológicos
7.
Artigo em Inglês | MEDLINE | ID: mdl-27734007

RESUMO

The water-borne pathogen Legionella pneumophila (Lp) strongly expresses the lpg1659 gene in water. This gene encodes a hypothetical protein predicted to be a membrane protein using in silico analysis. While no conserved domains were identified in Lpg1659, similar proteins are found in many Legionella species and other aquatic bacteria. RT-qPCR showed that lpg1659 is positively regulated by the alternative sigma factor RpoS, which is essential for Lp to survive in water. These observations suggest an important role of this novel protein in the survival of Lp in water. Deletion of lpg1659 did not affect cell morphology, membrane integrity or tolerance to high temperature. Moreover, lpg1659 was dispensable for growth of Lp in rich medium, and during infection of the amoeba Acanthamoeba castellanii and of THP-1 human macrophages. However, deletion of lpg1659 resulted in an early loss of culturability in water, while over-expression of this gene promoted the culturability of Lp. Therefore, these results suggest that lpg1659 is required for Lp to maintain culturability, and possibly long-term survival, in water. Since the loss of culturability observed in the absence of Lpg1659 was complemented by the addition of trace metals into water, this membrane protein is likely a transporter for acquiring essential trace metal for maintaining culturability in water and potentially in other metal-deprived conditions. Given its role in the survival of Lp in water, Lpg1659 was named LasM for Legionella aquatic survival membrane protein.


Assuntos
Regulação Bacteriana da Expressão Gênica , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Regiões Promotoras Genéticas , Microbiologia da Água , Acanthamoeba castellanii/microbiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular/microbiologia , Deleção de Genes , Humanos , Legionella pneumophila/citologia , Legionella pneumophila/metabolismo , Doença dos Legionários/microbiologia , Macrófagos/microbiologia , Mutação , Fator sigma/genética , Fator sigma/metabolismo , Sobrevida , Análise de Sobrevida , Temperatura , Termotolerância , Oligoelementos/metabolismo , Água
8.
BMC Microbiol ; 16(1): 174, 2016 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-27484084

RESUMO

BACKGROUND: The opportunistic bacterial pathogen Legionella pneumophila uses substrate effectors of Dot/Icm type IVB secretion system (T4BSS) to accomplish survival and replication in amoebae cells and mammalian alveolar macrophages. During the conversion between its highly resistant, infectious dormant form and vigorously growing, uninfectious replicative form, L. pneumophila utilizes a complicated regulatory network in which proteolysis may play a significant role. As a highly conserved core protease, ClpP is involved in various cellular processes as well as virulence in bacteria, and has been proved to be required for the expression of transmission traits and cell division of L. pneumophila. RESULTS: The clpP-deficient L. pneumophila strain failed to replicate and was digested in the first 3 h post-infection in mammalian cells J774A.1. Further investigation demonstrates that the clpP deficient mutant strain was unable to escape the endosome-lysosomal pathway in host cells. We also found that the clpP deficient mutant strain still expresses T4BSS components, induces contact-dependent cytotoxicity and translocate effector proteins RalF and LegK2, indicating that its T4BSS was overall functional. Interestingly, we further found that the translocation of several effector proteins is significantly reduced without ClpP. CONCLUSIONS: The data indicate that ClpP plays an important role in regulating the virulence and effector translocation of Legionella pneumophila.


Assuntos
Proteínas de Bactérias/genética , Endopeptidase Clp/genética , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Animais , Proteínas de Bactérias/metabolismo , Translocação Bacteriana/efeitos dos fármacos , Linhagem Celular , Endocitose/fisiologia , Endopeptidase Clp/deficiência , Endopeptidase Clp/metabolismo , Endossomos/metabolismo , Endossomos/microbiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Legionella pneumophila/citologia , Legionella pneumophila/enzimologia , Lisossomos/metabolismo , Lisossomos/microbiologia , Macrófagos/microbiologia , Camundongos , Mutação , Fagocitose , Deleção de Sequência , Virulência
9.
Nature ; 533(7601): 120-4, 2016 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-27049943

RESUMO

Signalling by ubiquitination regulates virtually every cellular process in eukaryotes. Covalent attachment of ubiquitin to a substrate is catalysed by the E1, E2 and E3 three-enzyme cascade, which links the carboxy terminus of ubiquitin to the ε-amino group of, in most cases, a lysine of the substrate via an isopeptide bond. Given the essential roles of ubiquitination in the regulation of the immune system, it is not surprising that the ubiquitination network is a common target for diverse infectious agents. For example, many bacterial pathogens exploit ubiquitin signalling using virulence factors that function as E3 ligases, deubiquitinases or as enzymes that directly attack ubiquitin. The bacterial pathogen Legionella pneumophila utilizes approximately 300 effectors that modulate diverse host processes to create a permissive niche for its replication in phagocytes. Here we demonstrate that members of the SidE effector family of L. pneumophila ubiquitinate multiple Rab small GTPases associated with the endoplasmic reticulum. Moreover, we show that these proteins are capable of catalysing ubiquitination without the need for the E1 and E2 enzymes. A putative mono-ADP-ribosyltransferase motif critical for the ubiquitination activity is also essential for the role of the SidE family in intracellular bacterial replication in a protozoan host. The E1/E2-independent ubiquitination catalysed by these enzymes is energized by nicotinamide adenine dinucleotide, which activates ubiquitin by the formation of ADP-ribosylated ubiquitin. These results establish that ubiquitination can be catalysed by a single enzyme, the activity of which does not require ATP.


Assuntos
Proteínas de Bactérias/metabolismo , Legionella pneumophila/química , Ubiquitinação , ADP Ribose Transferases/química , ADP Ribose Transferases/metabolismo , Adenosina Difosfato Ribose/metabolismo , Trifosfato de Adenosina , Motivos de Aminoácidos , Sequência de Aminoácidos , Carga Bacteriana , Biocatálise , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/metabolismo , Legionella pneumophila/citologia , Legionella pneumophila/enzimologia , Legionella pneumophila/patogenicidade , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , NAD/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo , Enzimas Ativadoras de Ubiquitina , Enzimas de Conjugação de Ubiquitina , Fatores de Virulência/metabolismo , Proteínas rab de Ligação ao GTP/química , Proteínas rab de Ligação ao GTP/metabolismo
10.
J Colloid Interface Sci ; 459: 175-182, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26291573

RESUMO

Magnetic nanoparticles are the next tool in medical diagnoses and treatment in many different biomedical applications, including magnetic hyperthermia as alternative treatment for cancer and bacterial infections, as well as the disruption of biofilms. The colloidal stability of the magnetic nanoparticles in a biological environment is crucial for efficient delivery. A surface that can be easily modifiable can also improve the delivery and imaging properties of the magnetic nanoparticle by adding targeting and imaging moieties, providing a platform for additional modification. The strategy presented in this work includes multiple nitroDOPA anchors for robust binding to the surface tied to the same polymer backbone as multiple poly(ethylene oxide) chains for steric stability. This approach provides biocompatibility and enhanced stability in fetal bovine serum (FBS) and phosphate buffer saline (PBS). As a proof of concept, these polymer-particles complexes were then modified with a near infrared dye and utilized in characterizing the integration of magnetic nanoparticles in biofilms. The work presented in this manuscript describes the synthesis and characterization of a nontoxic platform for the labeling of near IR-dyes for bioimaging.


Assuntos
Biofilmes , Di-Hidroxifenilalanina/química , Corantes Fluorescentes/química , Legionella pneumophila , Nanopartículas/química , Polietilenoglicóis/química , Animais , Bovinos , Legionella pneumophila/citologia , Legionella pneumophila/fisiologia , Camundongos , Microscopia de Fluorescência
11.
Cell Microbiol ; 17(5): 607-20, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25363599

RESUMO

The formation and release of outer membrane vesicles (OMVs) is a phenomenon observed in many bacteria, including Legionella pneumophila. During infection, this human pathogen primarily invades alveolar macrophages and replicates within a unique membrane-bound compartment termed Legionella-containing vacuole. In the current study, we analysed the membrane architecture of L. pneumophila OMVs by small-angle X-ray scattering and biophysically characterized OMV membranes. We investigated the interaction of L. pneumophila OMVs with model membranes by Förster resonance energy transfer and Fourier transform infrared spectroscopy. These experiments demonstrated the incorporation of OMV membrane material into liposomes composed of different eukaryotic phospholipids, revealing an endogenous property of OMVs to fuse with eukaryotic membranes. Cellular co-incubation experiments showed a dose- and time-dependent binding of fluorophore-labelled OMVs to macrophages. Trypan blue quenching experiments disclosed a rapid internalization of OMVs into macrophages at 37 and 4 °C. Purified OMVs induced tumour necrosis factor-α production in human macrophages at concentrations starting at 300 ng ml(-1). Experiments on HEK293-TLR2 and TLR4/MD-2 cell lines demonstrated a dominance of TLR2-dependent signalling pathways. In summary, we demonstrate binding, internalization and biological activity of L. pneumophila OMVs on human macrophages. Our data support OMV membrane fusion as a mechanism for the remote delivery of virulence factors to host cells.


Assuntos
Membrana Celular/metabolismo , Exossomos/metabolismo , Interações Hospedeiro-Patógeno , Legionella pneumophila/fisiologia , Fatores de Virulência/metabolismo , Fenômenos Biofísicos , Células Cultivadas , Endocitose , Células Epiteliais/metabolismo , Exossomos/química , Transferência Ressonante de Energia de Fluorescência , Humanos , Legionella pneumophila/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Espalhamento a Baixo Ângulo , Espectroscopia de Infravermelho com Transformada de Fourier , Temperatura , Fator de Necrose Tumoral alfa/metabolismo
12.
Environ Microbiol ; 16(2): 382-95, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23968544

RESUMO

Legionella pneumophila is an intracellular bacterial parasite of freshwater protozoa and an accidental waterborne human pathogen. L. pneumophila is highly pleomorphic showing several forms that differentiate within its developmental cycle. In water, L. pneumophila produces viable but non-culturable cells (VBNCCs), which remain largely uncharacterized. We produced VBNCCs from two developmental forms of L. pneumophila [stationary phase forms (SPFs) and mature infectious forms (MIFs)] in two water microcosms [double-deionized (dd) and tap water] at 45°C. In contrast with SPFs, MIFs upheld a robust ultrastructure and high viability in the two water microcosms. In dd-water, MIFs and SPFs lost their culturability faster than in tap water and did not consume their poly-ß-hydroxybutyrate inclusions. Resuscitation in Acanthamoeba castellani was only possible for VBNCCs produced from SPFs in tap water. Addition of salts to dd-water prolonged L. pneumophila culturability to tap water levels, suggesting that L. pneumophila requires ions to maintain its readiness to resume growth. VBNCCs resisted detergent lysis and digestion in the ciliate Tetrahymena, except for VBNCCs produced from SPFs in dd-water. L. pneumophila VBNCCs thus show distinct traits according to its originating developmental form and the surrounding water microcosm.


Assuntos
Água Doce/química , Legionella pneumophila/citologia , Viabilidade Microbiana , Microbiologia da Água , Água Potável/química , Concentração de Íons de Hidrogênio , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/ultraestrutura , Microscopia Eletrônica de Transmissão , Sais/química , Temperatura
13.
Water Res ; 47(17): 6606-17, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24064547

RESUMO

Legionella pneumophila is the agent responsible for legionellosis. Numerous bacteria, including L. pneumophila, can enter into a viable but not culturable (VBNC) state under unfavorable environmental conditions. In this state, cells are unable to form colonies on standard medium but are still alive. Here we show that VBNC L. pneumophila cells, obtained by monochloramine treatment, were still able to synthesize proteins, some of which are involved in virulence. Protein synthesis was measured using (35)S-labeling and the proteomes of VBNC and culturable cells then compared. This analysis allowed the identification of nine proteins that were accumulated in the VBNC state. Among them, four were involved in virulence, i.e., the macrophage infectivity potentiator protein, the hypothetical protein lpl2247, the ClpP protease proteolytic subunit and the 27 kDa outer membrane protein. Others, i.e., the enoyl reductase, the electron transfer flavoprotein (alpha and beta subunits), the 50S ribosomal proteins (L1 and L25) are involved in metabolic and energy production pathways. However, resuscitation experiments performed with Acanthamoeba castellanii failed, suggesting that the accumulation of virulence factors by VBNC cells is not sufficient to maintain their virulence.


Assuntos
Proteínas de Bactérias/biossíntese , Legionella pneumophila/citologia , Legionella pneumophila/patogenicidade , Viabilidade Microbiana , Fatores de Virulência/biossíntese , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cloraminas/farmacologia , Esterases/metabolismo , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/enzimologia , Viabilidade Microbiana/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteoma/metabolismo
14.
BMC Infect Dis ; 13: 371, 2013 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-23937571

RESUMO

BACKGROUND: Legionella pneumophila (Lp) flagellin activates signaling pathways in murine macrophages that control Lp replication. Nucleotide-binding oligomerization domain (NOD) containing-like receptor (NLR) family, caspase recruitment domain (CARD) containing 4 (NLRC4) and Toll-like Receptor (TLR5) both recognize Lp flagellin in vitro, but whether these two receptors play redundant or separate functional roles in vivo is unknown. METHODS: The immune response of Nlrc4-/-, Nlrc4-/-/Tlr5-/-, and wild type C57Bl/6 mice was analyzed after in vivo infection with aerosolized Lp. RESULTS: Lp clearance from the lungs was delayed in Nlrc4-/- mice over seven days in comparison to wild type controls. Nlrc4-/-/Tlr5-/- mice had no additional defect. In contrast to TLR5, NLRC4 did not regulate recruitment of neutrophils to the lung. Although there were no differences among the mouse strains in the lung transcriptome at 4 hours, Nlrc4-/- and Nlrc4-/-Tlr5-/- mice had increased lung inflammation at 72 hours in comparison to WT. Nlrc4-/-/Tlr5-/- mice also had altered cytokine production at both 4 and 24 hours post infection when compared to wild-type (WT) and Nlrc4-/- mice. Lp replication in murine alveolar macrophages was NLRC4-dependent and TLR5-independent. CONCLUSION: These studies reveal that NLRC4 and TLR5 mediate different roles in the inflammatory response to Lp flagellin in an aerosolized infection model and NLRC4 regulates replication in both lungs and alveolar macrophages.


Assuntos
Proteínas Reguladoras de Apoptose/farmacologia , Proteínas de Ligação ao Cálcio/farmacologia , Legionella pneumophila/citologia , Doença dos Legionários/microbiologia , Animais , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/metabolismo , Feminino , Flagelina/metabolismo , Interações Hospedeiro-Patógeno , Legionella pneumophila/imunologia , Legionella pneumophila/metabolismo , Doença dos Legionários/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais , Receptores Toll-Like/genética
15.
Cell Microbiol ; 14(10): 1632-55, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22727141

RESUMO

Legionella, the aetiological agent responsible for Legionellosis, is an opportunistic pathogen that infects humans upon the inhalation of contaminated aerosolized water droplets. Legionella is pleomorphic and its different morphotypes exhibit varying degrees of virulence. While the filamentous forms of Legionella pneumophila (Lp) have been reported in patient samples since the first description of legionellosis, their role in disease has not been studied. Our results show that both E-cadherin and ß1 integrin receptors mediate filamentous Lp (FLp) attachment to lung epithelial cells (LECs). The activation of these receptors induces the formation of actin enriched membrane surface structures that we designated 'hooks' and 'membrane wraps'. These structures entrap the filaments on the cell surface leading to their gradual internalization through a zipper mechanism of phagocytosis dependent on actomyosin activity. The supply of E-cadherin receptors from the recycling pathway and ß1 integrins released from focal adhesion turnover are required to sustain this process. Intracellular FLp inhabits a vacuolar compartment where filaments differentiate into short rods and replicate to produce infective progeny. Here we are reporting a first description of the invasion mechanism used by FLp to invade LECs. Therefore, filamentous morphotype of Lp can induce its own uptake by LECs and has the potential ability to cause disease.


Assuntos
Células Epiteliais/microbiologia , Legionella pneumophila/patogenicidade , Fagocitose , Actomiosina/metabolismo , Aderência Bacteriana , Caderinas/metabolismo , Linhagem Celular , Humanos , Integrina beta1/metabolismo , Legionella pneumophila/citologia , Ligação Proteica
16.
Future Microbiol ; 7(3): 369-81, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22393890

RESUMO

Eukaryotic proteins are tightly regulated by post-translational modifications, leading to a very subtle degree of regulation in time and space. Pathogen-mediated post-translational modifications are key strategies to modulate host factors by targeting central signaling pathways in the host cell. Legionella pneumophila, an intracellular pathogen that coevolved with protozoan hosts, encodes a large arsenal of secreted effectors conferring the ability to evade host cellular defenses and to manipulate them to promote invasion and intracellular replication. Conservation of many signaling pathways of protozoa in human macrophages confers the ability of L. pneumophila to infect humans, causing a severe pneumonia called legionnaires' disease. Most of the secreted proteins are delivered by the Dot/Icm type IV secretion system and several of these have been shown to act on different cellular pathways critical for infection. Moreover, multiple effectors target a single host function to orchestrate bacterial survival. In this review, we focus on those effectors in the repertoire of L. pneumophila proteins that target key cellular pathways by specific post-translational modifications.


Assuntos
Legionella pneumophila/citologia , Legionella pneumophila/metabolismo , Doença dos Legionários/metabolismo , Doença dos Legionários/microbiologia , Viabilidade Microbiana , Proteínas/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Legionella pneumophila/genética , Processamento de Proteína Pós-Traducional
17.
Appl Environ Microbiol ; 77(17): 6225-32, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21742913

RESUMO

A new method was developed for the rapid and sensitive detection of viable Legionella pneumophila. The method combines specific immunofluorescence (IF) staining using monoclonal antibodies with a bacterial viability marker (ChemChrome V6 cellular esterase activity marker) by means of solid-phase cytometry (SPC). IF methods were applied to the detection and enumeration of both the total and viable L. pneumophila cells in water samples. The sensitivity of the IF methods coupled to SPC was 34 cells liter(-1), and the reproducibility was good, with the coefficient of variation generally falling below 30%. IF methods were applied to the enumeration of total and viable L. pneumophila cells in 46 domestic hot water samples as well as in cooling tower water and natural water samples, such as thermal spring water and freshwater samples. Comparison with standard plate counts showed that (i) the total direct counts were always higher than the plate counts and (ii) the viable counts were higher than or close to the plate counts. With domestic hot waters, when the IF assay was combined with the viability test, SPC detected up to 3.4 × 10(3) viable but nonculturable L. pneumophila cells per liter. These direct IF methods could be a powerful tool for high-frequency monitoring of domestic hot waters or for investigating the occurrence of viable L. pneumophila in both man-made water systems and environmental water samples.


Assuntos
Carga Bacteriana/métodos , Legionella pneumophila/isolamento & purificação , Legionella pneumophila/fisiologia , Microbiologia da Água , Imunofluorescência/métodos , Legionella pneumophila/citologia , Viabilidade Microbiana , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Coloração e Rotulagem/métodos
18.
Clin Exp Immunol ; 162(2): 289-97, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20819093

RESUMO

CCL20 is a chemokine that attracts immature dendritic cells. We show that monocytes, cells characteristic of the innate immune response, infected with Mycobacterium tuberculosis express the CCL20 gene at a much higher level than the same cells infected with non-tuberculous mycobacteria. Interferon (IFN)-γ, a fundamental cytokine in the immune response to tuberculosis, strongly inhibits both the transcription and the translation of CCL20. We have also confirmed that dendritic cells are a suitable host for mycobacteria proliferation, although CCL20 does not seem to influence their intracellular multiplication rate. The chemokine, however, down-regulates the characteristic production of reactive oxygen species (ROS) induced by M. tuberculosis in monocytes, which may affect the activity of the cells. Apoptosis mediated by the mycobacteria, possibly ROS-dependent, was also inhibited by CCL20.


Assuntos
Quimiocina CCL20/metabolismo , Monócitos/metabolismo , Monócitos/microbiologia , Mycobacterium tuberculosis/imunologia , Espécies Reativas de Oxigênio/metabolismo , Anticorpos/imunologia , Anticorpos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Quimiocina CCL2/imunologia , Quimiocina CCL2/metabolismo , Quimiocina CCL20/genética , Quimiocina CCL20/imunologia , Quimiocina CCL20/farmacologia , Quimiocinas CC/genética , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/imunologia , Contagem de Colônia Microbiana , Meios de Cultivo Condicionados/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Expressão Gênica/genética , Humanos , Interferon gama/farmacologia , Legionella pneumophila/citologia , Legionella pneumophila/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Mycobacterium avium/citologia , Mycobacterium avium/imunologia , Mycobacterium kansasii/citologia , Mycobacterium kansasii/imunologia , Mycobacterium tuberculosis/citologia
19.
J Exp Med ; 207(8): 1713-26, 2010 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-20660614

RESUMO

Farnesylation involves covalent linkage of eukaryotic proteins to a lipid moiety to anchor them into membranes, which is essential for the biological function of Ras and other proteins. A large cadre of bacterial effectors is injected into host cells by intravacuolar pathogens through elaborate type III-VII translocation machineries, and many of these effectors are incorporated into the pathogen-containing vacuolar membrane by unknown mechanisms. The Dot/Icm type IV secretion system of Legionella pneumophila injects into host cells the F-box effector Ankyrin B (AnkB), which functions as platforms for the docking of polyubiquitinated proteins to the Legionella-containing vacuole (LCV) to enable intravacuolar proliferation in macrophages and amoeba. We show that farnesylation of AnkB is indispensable for its anchoring to the cytosolic face of the LCV membrane, for its biological function within macrophages and Dictyostelium discoideum, and for intrapulmonary proliferation in mice. Remarkably, the protein farnesyltransferase, RCE-1 (Ras-converting enzyme-1), and isoprenyl cysteine carboxyl methyltransferase host farnesylation enzymes are recruited to the LCV in a Dot/Icm-dependent manner and are essential for the biological function of AnkB. In conclusion, this study shows novel localized recruitment of the host farnesylation machinery and its anchoring of an F-box effector to the LCV membrane, and this is essential for biological function in vitro and in vivo.


Assuntos
Anquirinas/metabolismo , Células Eucarióticas/metabolismo , Células Eucarióticas/microbiologia , Legionella pneumophila/fisiologia , Prenilação de Proteína/fisiologia , Animais , Anquirinas/química , Anquirinas/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Proliferação de Células , Dictyostelium/metabolismo , Dictyostelium/microbiologia , Endopeptidases/genética , Endopeptidases/metabolismo , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Farnesiltranstransferase/genética , Farnesiltranstransferase/metabolismo , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Membranas Intracelulares/metabolismo , Legionella pneumophila/citologia , Doença dos Legionários/microbiologia , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Camundongos Endogâmicos , Domínios e Motivos de Interação entre Proteínas/genética , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/metabolismo , Prenilação de Proteína/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/genética , Interferência de RNA , Transfecção , Células U937 , Proteínas Ubiquitinadas/metabolismo , Vacúolos/metabolismo , Vacúolos/microbiologia
20.
BMC Microbiol ; 10: 54, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20167127

RESUMO

BACKGROUND: Legionella pneumophila, the intracellular bacterial pathogen that causes Legionnaires' disease, exhibit characteristic transmission traits such as elevated stress tolerance, shortened length and virulence during the transition from the replication phase to the transmission phase. ClpP, the catalytic core of the Clp proteolytic complex, is widely involved in many cellular processes via the regulation of intracellular protein quality. RESULTS: In this study, we showed that ClpP was required for optimal growth of L. pneumophila at high temperatures and under several other stress conditions. We also observed that cells devoid of clpP exhibited cell elongation, incomplete cell division and compromised colony formation. Furthermore, we found that the clpP-deleted mutant was more resistant to sodium stress and failed to proliferate in the amoebae host Acanthamoeba castellanii. CONCLUSIONS: The data present in this study illustrate that the ClpP protease homologue plays an important role in the expression of transmission traits and cell division of L. pneumophila, and further suggest a putative role of ClpP in virulence regulation.


Assuntos
Endopeptidase Clp/fisiologia , Legionella pneumophila/fisiologia , Acanthamoeba castellanii/microbiologia , Sequência de Aminoácidos , Divisão Celular/genética , Endopeptidase Clp/genética , Temperatura Alta , Legionella pneumophila/citologia , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Dados de Sequência Molecular , Mutação , Alinhamento de Sequência , Cloreto de Sódio/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...