Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38559167

RESUMO

To promote intracellular survival and infection, Legionella spp. translocate hundreds of effector proteins into eukaryotic host cells using a type IV b protein secretion system (T4bSS). T4bSS are well known to translocate soluble as well as transmembrane domain-containing effector proteins (TMD-effectors) but the mechanisms of secretion are still poorly understood. Herein we investigated the secretion of hydrophobic TMD-effectors, of which about 80 were previously reported to be encoded by L. pneumophila. A proteomic analysis of fractionated membranes revealed that TMD-effectors are targeted to and inserted into the bacterial inner membranes of L. pneumophila independent of the presence of a functional T4bSS. While the T4bSS chaperones IcmS and IcmW were critical for secretion of all tested TMD-effectors, they did not influence inner membrane targeting of these proteins. As for soluble effector proteins, translocation of TMD-effectors into host cells depended on a C-terminal secretion signal and this signal needed to be presented towards the cytoplasmic side of the inner membrane. A different secretion behavior of TMD- and soluble effectors and the need for small periplasmic loops within TMD-effectors provided strong evidence that TMD-effectors are secreted in a two-step secretion process: Initially, an inner membrane intermediate is formed, that is extracted towards the cytoplasmic side, possibly by the help of the type IV coupling protein complex and subsequently secreted into eukaryotic host cells by the T4bSS core complex. Overall, our study highlights the amazing versatility of T4bSS to secrete soluble and TMD-effectors from different subcellular locations of the bacterial cell.

2.
Nat Commun ; 14(1): 7068, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37923743

RESUMO

Legionella pneumophila (LP) secretes more than 300 effectors into the host cytosol to facilitate intracellular replication. One of these effectors, SidH, 253 kDa in size with no sequence similarity to proteins of known function is toxic when overexpressed in host cells. SidH is regulated by the LP metaeffector LubX which targets SidH for degradation in a temporal manner during LP infection. The mechanism underlying the toxicity of SidH and its role in LP infection are unknown. Here, we determined the cryo-EM structure of SidH at 2.7 Å revealing a unique alpha helical arrangement with no overall similarity to known protein structures. Surprisingly, purified SidH came bound to a E. coli EF-Tu/t-RNA/GTP ternary complex which could be modeled into the cryo-EM density. Mutation of residues disrupting the SidH-tRNA interface and SidH-EF-Tu interface abolish the toxicity of overexpressed SidH in human cells, a phenotype confirmed in infection of Acanthamoeba castellani. We also present the cryo-EM structure of SidH in complex with a U-box domain containing ubiquitin ligase LubX delineating the mechanism of regulation of SidH. Our data provide the basis for the toxicity of SidH and into its regulation by the metaeffector LubX.


Assuntos
Legionella pneumophila , Doença dos Legionários , Humanos , Legionella pneumophila/metabolismo , Escherichia coli/metabolismo , Fator Tu de Elongação de Peptídeos/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas de Bactérias/metabolismo
3.
Genome Res ; 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-36109147

RESUMO

The unicellular amoeba Acanthamoeba castellanii is ubiquitous in aquatic environments, where it preys on bacteria. The organism also hosts bacterial endosymbionts, some of which are parasitic, including human pathogens such as Chlamydia and Legionella spp. Here we report complete, high-quality genome sequences for two extensively studied A. castellanii strains, Neff and C3. Combining long- and short-read data with Hi-C, we generated near chromosome-level assemblies for both strains with 90% of the genome contained in 29 scaffolds for the Neff strain and 31 for the C3 strain. Comparative genomics revealed strain-specific functional enrichment, most notably genes related to signal transduction in the C3 strain and to viral replication in Neff. Furthermore, we characterized the spatial organization of the A. castellanii genome and showed that it is reorganized during infection by Legionella pneumophila Infection-dependent chromatin loops were found to be enriched in genes for signal transduction and phosphorylation processes. In genomic regions where chromatin organization changed during Legionella infection, we found functional enrichment for genes associated with metabolism, organelle assembly, and cytoskeleton organization. Given Legionella infection is known to alter its host's cell cycle, to exploit the host's organelles, and to modulate the host's metabolism in its favor, these changes in chromatin organization may partly be related to mechanisms of host control during Legionella infection.

4.
Nat Commun ; 13(1): 762, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35140216

RESUMO

Legionella pneumophila is an intracellular bacterial pathogen that can cause a severe form of pneumonia in humans, a phenotype evolved through interactions with aquatic protozoa in the environment. Here, we show that L. pneumophila uses extracellular vesicles to translocate bacterial small RNAs (sRNAs) into host cells that act on host defence signalling pathways. The bacterial sRNA RsmY binds to the UTR of ddx58 (RIG-I encoding gene) and cRel, while tRNA-Phe binds ddx58 and irak1 collectively reducing expression of RIG-I, IRAK1 and cRel, with subsequent downregulation of IFN-ß. Thus, RsmY and tRNA-Phe are bacterial trans-kingdom regulatory RNAs downregulating selected sensor and regulator proteins of the host cell innate immune response. This miRNA-like regulation of the expression of key sensors and regulators of immunity is a feature of L. pneumophila host-pathogen communication and likely represents a general mechanism employed by bacteria that interact with eukaryotic hosts.


Assuntos
Eucariotos/imunologia , Interações Hospedeiro-Patógeno/imunologia , Legionella pneumophila/metabolismo , Doença dos Legionários/imunologia , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Bactérias/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , Eucariotos/genética , Vesículas Extracelulares , Humanos , Imunidade Inata , Quinases Associadas a Receptores de Interleucina-1 , Doença dos Legionários/microbiologia , Receptores Imunológicos , Transdução de Sinais
5.
Elife ; 102021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34882089

RESUMO

Legionella pneumophila, the causative agent of Legionnaires' disease, a severe pneumonia, injects via a type 4 secretion system (T4SS) more than 300 proteins into macrophages, its main host cell in humans. Certain of these proteins are implicated in reprogramming the metabolism of infected cells by reducing mitochondrial oxidative phosphorylation (OXPHOS) early after infection. Here. we show that despite reduced OXPHOS, the mitochondrial membrane potential (Δψm) is maintained during infection of primary human monocyte-derived macrophages (hMDMs). We reveal that L. pneumophila reverses the ATP-synthase activity of the mitochondrial FOF1-ATPase to ATP-hydrolase activity in a T4SS-dependent manner, which leads to a conservation of the Δψm, preserves mitochondrial polarization, and prevents macrophage cell death. Analyses of T4SS effectors known to target mitochondrial functions revealed that LpSpl is partially involved in conserving the Δψm, but not LncP and MitF. The inhibition of the L. pneumophila-induced 'reverse mode' of the FOF1-ATPase collapsed the Δψm and caused cell death in infected cells. Single-cell analyses suggested that bacterial replication occurs preferentially in hMDMs that conserved the Δψm and showed delayed cell death. This direct manipulation of the mode of activity of the FOF1-ATPase is a newly identified feature of L. pneumophila allowing to delay host cell death and thereby to preserve the bacterial replication niche during infection.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/genética , Legionella pneumophila/metabolismo , Mitocôndrias/metabolismo , ATPases Translocadoras de Prótons/deficiência , Trifosfato de Adenosina/genética , Proteínas de Bactérias/metabolismo , Legionella pneumophila/genética , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Sistemas de Secreção Tipo IV/metabolismo
6.
Environ Microbiol ; 21(3): 1035-1053, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30623561

RESUMO

The causative agent of Legionnaires' disease, Legionella pneumophila, colonizes amoebae and biofilms in the environment. The opportunistic pathogen employs the Lqs (Legionella quorum sensing) system and the signalling molecule LAI-1 (Legionella autoinducer-1) to regulate virulence, motility, natural competence and expression of a 133 kb genomic "fitness island", including a putative novel regulator. Here, we show that the regulator termed LvbR is an LqsS-regulated transcription factor that binds to the promoter of lpg1056/hnox1 (encoding an inhibitor of the diguanylate cyclase Lpg1057), and thus, regulates proteins involved in c-di-GMP metabolism. LvbR determines biofilm architecture, since L. pneumophila lacking lvbR accumulates less sessile biomass and forms homogeneous mat-like structures, while the parental strain develops more compact bacterial aggregates. Comparative transcriptomics of sessile and planktonic ΔlvbR or ΔlqsR mutant strains revealed concerted (virulence, fitness island, metabolism) and reciprocally (motility) regulated genes in biofilm and broth respectively. Moreover, ΔlvbR is hyper-competent for DNA uptake, defective for phagocyte infection, outcompeted by the parental strain in amoebae co-infections and impaired for cell migration inhibition. Taken together, our results indicate that L. pneumophila LvbR is a novel pleiotropic transcription factor, which links the Lqs and c-di-GMP regulatory networks to control biofilm architecture and pathogen-host cell interactions.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes , GMP Cíclico/análogos & derivados , Regulação Bacteriana da Expressão Gênica , Redes Reguladoras de Genes , Legionella pneumophila/genética , Fatores de Transcrição/metabolismo , 4-Butirolactona/análogos & derivados , Proteínas de Bactérias/genética , GMP Cíclico/metabolismo , Legionella pneumophila/patogenicidade , Doença dos Legionários/microbiologia , Percepção de Quorum , Virulência
7.
Artigo em Inglês | MEDLINE | ID: mdl-29404281

RESUMO

Legionella pneumophila is a gram-negative bacterium that inhabits freshwater ecosystems, where it is present in biofilm or as planktonic form. L. pneumophila is mainly found associated with protozoa, which serve as protection from hostile environments and as replication niche. If inhaled within aerosols, L. pneumophila is also able to infect and replicate in human alveolar macrophages, eventually causing the Legionnaires' disease. The transition between intracellular and extracellular environments triggers a differentiation program in which metabolic as well as morphogenetic changes occur. We here describe the current knowledge on how the different developmental states of this bacterium are regulated, with a particular emphasis on the stringent response activated during the transition from the replicative phase to the infectious phase and the metabolic features going in hand. We propose that the cellular differentiation of this intracellular pathogen is closely associated to key metabolic changes in the bacterium and the host cell, which together have a crucial role in the regulation of L. pneumophila virulence.


Assuntos
Interações Hospedeiro-Patógeno , Legionella pneumophila/fisiologia , Doença dos Legionários/microbiologia , Estágios do Ciclo de Vida , Animais , Metabolismo Energético , Meio Ambiente , Interações Hospedeiro-Patógeno/imunologia , Humanos , Doença dos Legionários/imunologia , Doença dos Legionários/metabolismo , Redes e Vias Metabólicas , Virulência
8.
Open Biol ; 7(11)2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29093212

RESUMO

Legionella pneumophila CsrA plays a crucial role in the life-stage-specific expression of virulence phenotypes and metabolic activity. However, its exact role is only partly known. To elucidate how CsrA impacts L. pneumophila metabolism we analysed the CsrA depended regulation of metabolic functions by comparative 13C-isotopologue profiling and oxygen consumption experiments of a L. pneumophila wild-type (wt) strain and its isogenic csrA- mutant. We show that a csrA- mutant has significantly lower respiration rates when serine, alanine, pyruvate, α-ketoglutarate or palmitate is the sole carbon source. By contrast, when grown in glucose or glycerol, no differences in respiration were detected. Isotopologue profiling uncovered that the transfer of label from [U-13C3]serine via pyruvate into the citrate cycle and gluconeogenesis was lower in the mutant as judged from the labelling patterns of protein-derived amino acids, cell-wall-derived diaminopimelate, sugars and amino sugars and 3-hydroxybutyrate derived from polyhydroxybutyrate (PHB). Similarly, the incorporation of [U-13C6]glucose via the glycolysis/Entner-Doudoroff (ED) pathway but not via the pentose phosphate pathway was repressed in the csrA- mutant. On the other hand, fluxes due to [U-13C3]glycerol utilization were increased in the csrA- mutant. In addition, we showed that exogenous [1,2,3,4-13C4]palmitic acid is efficiently used for PHB synthesis via 13C2-acetyl-CoA. Taken together, CsrA induces serine catabolism via the tricarboxylic acid cycle and glucose degradation via the ED pathway, but represses glycerol metabolism, fatty acid degradation and PHB biosynthesis, in particular during exponential growth. Thus, CsrA has a determining role in substrate usage and carbon partitioning during the L. pneumophila life cycle and regulates a switch from amino acid usage in replicative phase to glycerolipid usage during transmissive growth.


Assuntos
Aminoácidos/metabolismo , Proteínas de Bactérias/metabolismo , Glicolipídeos/metabolismo , Proteínas Repressoras/metabolismo , Respiração Celular , Ciclo do Ácido Cítrico , Glucose/metabolismo , Glicólise , Legionella pneumophila/metabolismo , Via de Pentose Fosfato
9.
PLoS Genet ; 13(2): e1006629, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28212376

RESUMO

The carbon storage regulator protein CsrA regulates cellular processes post-transcriptionally by binding to target-RNAs altering translation efficiency and/or their stability. Here we identified and analyzed the direct targets of CsrA in the human pathogen Legionella pneumophila. Genome wide transcriptome, proteome and RNA co-immunoprecipitation followed by deep sequencing of a wild type and a csrA mutant strain identified 479 RNAs with potential CsrA interaction sites located in the untranslated and/or coding regions of mRNAs or of known non-coding sRNAs. Further analyses revealed that CsrA exhibits a dual regulatory role in virulence as it affects the expression of the regulators FleQ, LqsR, LetE and RpoS but it also directly regulates the timely expression of over 40 Dot/Icm substrates. CsrA controls its own expression and the stringent response through a regulatory feedback loop as evidenced by its binding to RelA-mRNA and links it to quorum sensing and motility. CsrA is a central player in the carbon, amino acid, fatty acid metabolism and energy transfer and directly affects the biosynthesis of cofactors, vitamins and secondary metabolites. We describe the first L. pneumophila riboswitch, a thiamine pyrophosphate riboswitch whose regulatory impact is fine-tuned by CsrA, and identified a unique regulatory mode of CsrA, the active stabilization of RNA anti-terminator conformations inside a coding sequence preventing Rho-dependent termination of the gap operon through transcriptional polarity effects. This allows L. pneumophila to regulate the pentose phosphate pathway and the glycolysis combined or individually although they share genes in a single operon. Thus the L. pneumophila genome has evolved to acclimate at least five different modes of regulation by CsrA giving it a truly unique position in its life cycle.


Assuntos
Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano/genética , Legionella pneumophila/genética , Proteínas Repressoras/genética , Proteínas de Bactérias/metabolismo , Sequência de Bases , Northern Blotting , Evolução Molecular , Retroalimentação Fisiológica , Perfilação da Expressão Gênica/métodos , Glicólise/genética , Interações Hospedeiro-Patógeno , Humanos , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidade , Doença dos Legionários/microbiologia , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Óperon/genética , Via de Pentose Fosfato/genética , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Riboswitch/genética , Espectrometria de Massas em Tandem , Virulência/genética
10.
mBio ; 8(1)2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-28074027

RESUMO

Legionella pneumophila is an environmental bacterium that parasitizes protozoa, but it may also infect humans, thereby causing a severe pneumonia called Legionnaires' disease. To cycle between the environment and a eukaryotic host, L. pneumophila is regulating the expression of virulence factors in a life cycle-dependent manner: replicating bacteria do not express virulence factors, whereas transmissive bacteria are highly motile and infective. Here we show that Hfq is an important regulator in this network. Hfq is highly expressed in transmissive bacteria but is expressed at very low levels in replicating bacteria. A L. pneumophila hfq deletion mutant exhibits reduced abilities to infect and multiply in Acanthamoeba castellanii at environmental temperatures. The life cycle-dependent regulation of Hfq expression depends on a unique cis-encoded small RNA named Anti-hfq that is transcribed antisense of the hfq transcript and overlaps its 5' untranslated region. The Anti-hfq sRNA is highly expressed only in replicating L. pneumophila where it regulates hfq expression through binding to the complementary regions of the hfq transcripts. This results in reduced Hfq protein levels in exponentially growing cells. Both the small noncoding RNA (sRNA) and hfq mRNA are bound and stabilized by the Hfq protein, likely leading to the cleavage of the RNA duplex by the endoribonuclease RNase III. In contrast, after the switch to transmissive bacteria, the sRNA is not expressed, allowing now an efficient expression of the hfq gene and consequently Hfq. Our results place Hfq and its newly identified sRNA anti-hfq in the center of the regulatory network governing L. pneumophila differentiation from nonvirulent to virulent bacteria. IMPORTANCE: The abilities of L. pneumophila to replicate intracellularly and to cause disease depend on its capacity to adapt to different extra- and intracellular environmental conditions. Therefore, a timely and fine-tuned expression of virulence factors and adaptation traits is crucial. Yet, the regulatory circuits governing the life cycle of L. pneumophila from replicating to virulent bacteria are only partly uncovered. Here we show that the life cycle-dependent regulation of the RNA chaperone Hfq relies on a small regulatory RNA encoded antisense to the hfq-encoding gene through a base pairing mechanism. Furthermore, Hfq regulates its own expression in an autoregulatory loop. The discovery of this RNA regulatory mechanism in L. pneumophila is an important step forward in the understanding of how the switch from inoffensive, replicating to highly virulent, transmissive L. pneumophila is regulated.


Assuntos
Regulação Bacteriana da Expressão Gênica , Fator Proteico 1 do Hospedeiro/biossíntese , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/metabolismo , Pequeno RNA não Traduzido/metabolismo , Acanthamoeba castellanii/microbiologia , Deleção de Genes , Fator Proteico 1 do Hospedeiro/genética , Legionella pneumophila/genética , Pequeno RNA não Traduzido/genética , Virulência
11.
Mol Microbiol ; 99(4): 778-93, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26538361

RESUMO

The causative agent of Legionnaires' disease, Legionella pneumophila, employs the autoinducer compound LAI-1 (3-hydroxypentadecane-4-one) for cell-cell communication. LAI-1 is produced and detected by the Lqs (Legionella quorum sensing) system, comprising the autoinducer synthase LqsA, the sensor kinases LqsS and LqsT, as well as the response regulator LqsR. Lqs-regulated processes include pathogen-host interactions, production of extracellular filaments and natural competence for DNA uptake. Here we show that synthetic LAI-1 promotes the motility of L. pneumophila by signalling through LqsS/LqsT and LqsR. Upon addition of LAI-1, autophosphorylation of LqsS/LqsT by [γ-(32) P]-ATP was inhibited in a dose-dependent manner. In contrast, the Vibrio cholerae autoinducer CAI-1 (3-hydroxytridecane-4-one) promoted the phosphorylation of LqsS (but not LqsT). LAI-1 did neither affect the stability of phospho-LqsS or phospho-LqsT, nor the dephosphorylation by LqsR. Transcriptome analysis of L. pneumophila treated with LAI-1 revealed that the compound positively regulates a number of genes, including the non-coding RNAs rsmY and rsmZ, and negatively regulates the RNA-binding global regulator crsA. Accordingly, LAI-1 controls the switch from the replicative to the transmissive growth phase of L. pneumophila. In summary, the findings indicate that LAI-1 regulates motility and the biphasic life style of L. pneumophila through LqsS- and LqsT-dependent phosphorylation signalling.


Assuntos
Alcanos/metabolismo , Cetonas/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , Transdução de Sinais , Alcanos/farmacologia , Movimento Celular , Escherichia coli/genética , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Interações Hospedeiro-Patógeno , Cetonas/farmacologia , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/crescimento & desenvolvimento , Movimento , Fosforilação , Percepção de Quorum , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Vibrio cholerae/genética
12.
FEMS Microbiol Rev ; 39(3): 331-49, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26009640

RESUMO

Sequencing-based studies have illuminated increased transcriptional complexity within the genome structure of bacteria and have resulted in the identification of many small regulatory RNAs (sRNA) and a large amount of antisense transcription. It remains an open question whether these sRNAs all indeed play regulatory roles, but their identification led to an exponential increase in studies searching for their function. This allowed to show that sRNAs may modulate virulence gene expression, cellular differentiation, metabolic functions, adaptation to environmental conditions and pathogenesis. In this review we will provide mechanistic insights into how sRNAs bind mRNAs and/or proteins. Furthermore, the important roles of the RNA chaperone Hfq, the CsrA system and the CRISPR RNA will be discussed. We will then focus on sRNAs and 5(') untranslated region (UTR) elements of intracellular bacteria like Chlamydia, Listeria, Legionella, or Salmonella, and place emphasis on those that are expressed during replication in host cells and are implicated in virulence and metabolism. In addition, sRNAs that regulate motility, iron homeostasis, and differentiation or stress responses will be highlighted. Taken together sRNAs constitute key elements in many major regulatory networks governing the intracellular life and virulence of pathogenic bacteria.


Assuntos
Regiões 5' não Traduzidas/genética , Fenômenos Fisiológicos Bacterianos/genética , RNA Bacteriano/metabolismo , Virulência/genética , Bactérias/genética , Bactérias/metabolismo , Bactérias/patogenicidade , Regulação Bacteriana da Expressão Gênica , Ligação Proteica
13.
Environ Microbiol ; 17(4): 1338-50, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25141909

RESUMO

Legionella pneumophila is a pathogenic bacterium commonly found in water. Eventually, it could be transmitted to humans via inhalation of contaminated aerosols. Iron is known as a key requirement for the growth of L. pneumophila in the environment and within its hosts. Many studies were performed to understand iron utilization by L. pneumophila but no global approaches were conducted. In this study, transcriptomic analyses were performed, comparing gene expression in L. pneumophila in standard versus iron restricted conditions. Among the regulated genes, a newly described one, lpp_2867, was highly induced in iron-restricted conditions. Mutants lacking this gene in L. pneumophila were not affected in siderophore synthesis or utilization. On the contrary, they were defective for growth on iron-depleted solid media and for ferrous iron uptake. A sequence analysis predicts that Lpp_2867 is a membrane protein, suggesting that it is involved in ferrous iron transport. We thus named it IroT, for iron transporter. Infection assays showed that the mutants are highly impaired in intracellular growth within their environmental host Acanthamoeba castellanii and human macrophages. Taken together, our results show that IroT is involved, directly or indirectly, in ferrous iron transport and is a key virulence factor for L. pneumophila.


Assuntos
Amoeba/microbiologia , Ferro/metabolismo , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidade , Doença dos Legionários/microbiologia , Macrófagos/microbiologia , Proteínas de Membrana/metabolismo , Fatores de Virulência/metabolismo , Proteínas de Bactérias/metabolismo , Sequência de Bases , Transporte Biológico , Humanos , Legionella pneumophila/genética , Proteínas de Membrana/genética , Dados de Sequência Molecular , Virulência , Fatores de Virulência/genética
14.
Environ Microbiol ; 16(2): 359-81, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23957615

RESUMO

Legionella pneumophila uses aquatic protozoa as replication niche and protection from harsh environments. Although L. pneumophila is not known to have a circadian clock, it encodes homologues of the KaiBC proteins of Cyanobacteria that regulate circadian gene expression. We show that L. pneumophila kaiB, kaiC and the downstream gene lpp1114, are transcribed as a unit under the control of the stress sigma factor RpoS. KaiC and KaiB of L. pneumophila do not interact as evidenced by yeast and bacterial two-hybrid analyses. Fusion of the C-terminal residues of cyanobacterial KaiB to Legionella KaiB restores their interaction. In contrast, KaiC of L. pneumophila conserved autophosphorylation activity, but KaiB does not trigger the dephosphorylation of KaiC like in Cyanobacteria. The crystal structure of L. pneumophila KaiB suggests that it is an oxidoreductase-like protein with a typical thioredoxin fold. Indeed, mutant analyses revealed that the kai operon-encoded proteins increase fitness of L. pneumophila in competitive environments, and confer higher resistance to oxidative and sodium stress. The phylogenetic analysis indicates that L. pneumophila KaiBC resemble Synechosystis KaiC2B2 and not circadian KaiB1C1. Thus, the L. pneumophila Kai proteins do not encode a circadian clock, but enhance stress resistance and adaption to changes in the environments.


Assuntos
Proteínas de Bactérias/metabolismo , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/metabolismo , Legionella pneumophila/genética , Óperon , Estresse Fisiológico , Acanthamoeba castellanii/microbiologia , Acanthamoeba castellanii/fisiologia , Adaptação Fisiológica , Proteínas de Bactérias/genética , Relógios Circadianos , Peptídeos e Proteínas de Sinalização do Ritmo Circadiano/genética , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Aptidão Genética , Legionella pneumophila/fisiologia , Fosforilação , Filogenia , Estrutura Terciária de Proteína , RNA Bacteriano/genética
15.
Cell Host Microbe ; 13(4): 395-405, 2013 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-23601102

RESUMO

Histone posttranslational modifications control eukaryotic gene expression and regulate many biological processes including immunity. Pathogens alter host epigenetic control to aid pathogenesis. We find that the intracellular bacterial pathogen Legionella pneumophila uses a Dot/Icm type IV secreted effector, RomA, to uniquely modify the host chromatin landscape. RomA, a SET domain-containing methyltransferase, trimethylates K14 of histone H3, a histone mark not previously described in mammals. RomA localizes to the infected cell nucleus where it promotes a burst of H3K14 methylation and consequently decreases H3K14 acetylation, an activating histone mark, to repress host gene expression. ChIP-seq analysis identified 4,870 H3K14 methylated promoter regions, including innate immune genes. Significantly reduced replication of a RomA-deleted strain in host cells was trans-complemented by wild-type, but not by catalytically inactive, RomA. Thus, a secreted L. pneumophila effector targets the host cell nucleus and modifies histones to repress gene expression and promote efficient intracellular replication.


Assuntos
Proteínas de Bactérias/genética , Cromatina/genética , Replicação do DNA/genética , Expressão Gênica/genética , Legionella pneumophila/genética , Sequência de Aminoácidos , Proteínas de Bactérias/imunologia , Linhagem Celular , Cromatina/imunologia , Replicação do DNA/imunologia , Expressão Gênica/imunologia , Histonas/genética , Histonas/imunologia , Humanos , Imunidade Inata/genética , Imunidade Inata/imunologia , Legionella pneumophila/imunologia , Metilação , Dados de Sequência Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Alinhamento de Sequência , Transcrição Gênica/genética , Transcrição Gênica/imunologia
16.
Environ Microbiol ; 15(2): 646-62, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23033905

RESUMO

Legionella pneumophila is an amoeba-resistant opportunistic pathogen that performs cell-cell communication through the signalling molecule 3-hydroxypentadecane-4-one (LAI-1, Legionella autoinducer-1). The lqs (Legionella quorum sensing) gene cluster encodes the LAI-1 autoinducer synthase LqsA, the cognate sensor kinase LqsS and the response regulator LqsR. Here we show that the Lqs system includes an 'orphan' homologue of LqsS termed LqsT. Compared with wild-type L. pneumophila, strains lacking lqsT or both lqsS and lqsT show increased salt resistance, greatly enhanced natural competence for DNA acquisition and impaired uptake by phagocytes. Sensitive novel single round growth assays and competition experiments using Acanthamoeba castellanii revealed that ΔlqsT and ΔlqsS-ΔlqsT, as well as ΔlqsA and other lqs mutant strains are impaired for intracellular growth and cannot compete against wild-type bacteria upon co-infection. In contrast to the ΔlqsS strain, ΔlqsT does not produce extracellular filaments. The phenotypes of the ΔlqsS-ΔlqsT strain are partially complemented by either lqsT or lqsS, but are not reversed by overexpression of lqsA, suggesting that LqsT and LqsS are the sole LAI-1-responsive sensor kinases in L. pneumophila. In agreement with the different phenotypes of the ΔlqsT and ΔlqsS strains, lqsT and lqsS are differentially expressed in the post-exponential growth phase, and transcriptome studies indicated that 90% of the genes, which are downregulated in absence of lqsT, are upregulated in absence of lqsS. Reciprocally regulated genes encode components of a 133 kb genomic 'fitness island' or translocated effector proteins implicated in virulence. Together, these results reveal a unique organization of the L. pneumophila Lqs system comprising two partially antagonistic LAI-1-responsive sensor kinases, LqsT and LqsS, which regulate distinct pools of genes implicated in pathogen-host cell interactions, competence, expression of a genomic island or production of extracellular filaments.


Assuntos
Proteínas de Bactérias/metabolismo , Competência de Transformação por DNA/genética , Interações Hospedeiro-Patógeno/fisiologia , Legionella pneumophila/fisiologia , Fosfotransferases/metabolismo , Fatores de Transcrição/metabolismo , Acanthamoeba castellanii/microbiologia , Proteínas de Bactérias/genética , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/genética , Legionella pneumophila/crescimento & desenvolvimento , Fosfotransferases/genética , Percepção de Quorum/genética , Sais/farmacologia
17.
Methods Mol Biol ; 954: 555-66, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150420

RESUMO

The adaptation of Legionella pneumophila to the different conditions it encounters in the environment and in the host is governed by a complex regulatory system. Current knowledge of these regulatory networks and the transcriptome responses of L. pneumophila is mainly based on microarray analysis and limited to transcriptional products of annotated protein-coding genes. The application of the Next-Generation Sequencing (NGS) technology allows now genome-wide strand-specific sequencing and accurate determination of all expressed regions of the genome to reveal the complete transcriptional network and the dynamic interplay of specific regulators on a genome-wide level. NGS-based techniques promote deeper understanding of the global transcriptional organization of L. pneumophila by identifying transcription start sites (TSS), alternative TSS and operon organization, noncoding RNAs, antisense RNAs, and 5'-/3'-untranslated regions. In this chapter we describe the construction of cDNA libraries for (1) RNA deep sequencing (RNA-seq) and (2) TSS mapping using the Illumina technology.


Assuntos
Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Legionella pneumophila/genética , Transcriptoma , Perfilação da Expressão Gênica , RNA Bacteriano/isolamento & purificação , Sítio de Iniciação de Transcrição
18.
Methods Mol Biol ; 954: 583-93, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23150422

RESUMO

Transcriptional and posttranscriptional regulators play a critical role in allowing a bacterium to adapt to the diverse environments and conditions it encounters. In order to characterize the role of these regulators the identification of their specific interaction partners is of utmost importance. Co-immunoprecipitation (IP) is based on antigen/antibody complex formation to purify a protein of interest from the rest of the samples together with its interaction partner. This method allows us to study direct interaction of a regulator with its specific binding partners like protein-RNA, protein-DNA, or protein-protein interactions. IP typically requires careful optimization and troubleshooting depending on the varying physicochemical characteristics of the protein of interest. In this chapter we present a starting point and the basic guidelines to obtain the best possible results from an IP experiment with subsequent use of new-generation sequencing techniques to detect mRNA or ncRNA targets (RIPseq) and protein-DNA interactions (ChIPseq).


Assuntos
Proteínas de Bactérias/metabolismo , DNA Bacteriano/metabolismo , Imunoprecipitação/métodos , Legionella pneumophila/genética , Legionella pneumophila/metabolismo , RNA Bacteriano/metabolismo , Imunoprecipitação da Cromatina/métodos , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Ligação Proteica
19.
RNA Biol ; 9(4): 503-19, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22546937

RESUMO

The bacterium Legionella pneumophila is found ubiquitously in aquatic environments and can cause a severe pneumonia in humans called Legionnaires' disease. How this bacterium switches from intracellular to extracellular life and adapts to different hosts and environmental conditions is only partly understood. Here we used RNA deep sequencing from exponentially (replicative) and post exponentially (virulent) grown L. pneumophila to analyze the transcriptional landscape of its entire genome. We established the complete operon map and defined 2561 primary transcriptional start sites (TSS). Interestingly, 187 of the 1805 TSS of protein-coding genes contained tandem promoters of which 93 show alternative usage dependent on the growth phase. Similarly, over 60% of 713 here identified ncRNAs are phase dependently regulated. Analysis of their conservation among the seven L. pneumophila genomes sequenced revealed many strain specific differences suggesting that L. pneumophila contains a highly dynamic pool of ncRNAs. Analysis of six ncRNAs exhibiting the same expression pattern as virulence genes showed that two, Lppnc0584 and Lppnc0405 are indeed involved in intracellular growth of L. pneumophila in A. castellanii. Furthermore, L. pneumophila encodes a small RNA named RsmX that functions together with RsmY and RsmZ in the LetA-CsrA regulatory pathway, crucial for the switch to the virulent phenotype. Together our data provide new insight into the transcriptional organization of the L. pneumophila genome, identified many new ncRNAs and will provide a framework for the understanding of virulence and adaptation properties of L. pneumophila.


Assuntos
Regulação Bacteriana da Expressão Gênica , Legionella pneumophila/genética , RNA Bacteriano/genética , Pequeno RNA não Traduzido/genética , Regiões 5' não Traduzidas , Acanthamoeba castellanii/microbiologia , Proteínas de Bactérias/genética , Sequência de Bases , Mapeamento Cromossômico , Sequência Consenso , Sequenciamento de Nucleotídeos em Larga Escala , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/patogenicidade , Anotação de Sequência Molecular , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Óperon , Regiões Promotoras Genéticas , RNA Bacteriano/metabolismo , Pequeno RNA não Traduzido/metabolismo , Análise de Sequência de RNA , Sítio de Iniciação de Transcrição , Transcriptoma , Virulência/genética
20.
Water Res ; 46(3): 808-16, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22192759

RESUMO

Legionella pneumophila is a ubiquitous environmental microorganism found in freshwater that can cause an acute form of pneumonia known as Legionnaires' disease. Despite widespread use of chlorine to ensure drinking water quality and awareness that L. pneumophila may escape these treatments, little is known about its effects on L. pneumophila. The aim of this study was to investigate the L. pneumophila transcriptional response induced by chlorine treatment. Transcriptome analysis, using DNA arrays, showed that a sublethal dose of chlorine induces a differential expression of 391 genes involved in stress response, virulence, general metabolism, information pathways and transport. Many of the stress response genes were significantly upregulated, whereas a significant number of virulence genes were repressed. In particular, exposure of L. pneumophila to chlorine induced the expression of cellular antioxidant proteins, stress proteins and transcriptional regulators. In addition, glutathione S-transferase specific activity was enhanced following chlorine treatment. Our results clearly indicate that chlorine induces expression of proteins involved in cellular defence mechanisms against oxidative stress that might be involved in adaptation or resistance to chlorine treatment.


Assuntos
Cloro/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/genética , Transcrição Gênica/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Perfilação da Expressão Gênica , Genes Bacterianos/genética , Legionella pneumophila/patogenicidade , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Virulência/efeitos dos fármacos , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...