Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
mBio ; 9(1)2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29437918

RESUMO

Coinciding with major changes to its municipal water system, Flint, MI, endured Legionnaires' disease outbreaks in 2014 and 2015. By sampling premise plumbing in Flint in the fall of 2016, we found that 12% of homes harbored legionellae, a frequency similar to that in residences in neighboring areas. To evaluate the genetic diversity of Legionella pneumophila in Southeast Michigan, we determined the sequence type (ST) and serogroup (SG) of the 18 residential isolates from Flint and Detroit, MI, and the 33 clinical isolates submitted by hospitals in three area counties in 2013 to 2016. Common to one environmental and four clinical samples were strains of L. pneumophila SG1 and ST1, the most prevalent ST worldwide. Among the Flint premise plumbing isolates, 14 of 16 strains were of ST367 and ST461, two closely related SG6 strain types isolated previously from patients and corresponding environmental samples. Each of the representative SG1 clinical strains and SG6 environmental isolates from Southeast Michigan infected and survived within macrophage cultures at least as well as a virulent laboratory strain, as judged by microscopy and by enumerating CFU. Likewise, 72 h after infection, the yield of viable-cell counts increased >100-fold for each of the representative SG1 clinical isolates, Flint premise plumbing SG6 ST367 and -461 isolates, and two Detroit residential isolates. We verified by immunostaining that SG1-specific antibody does not cross-react with the SG6 L. pneumophila environmental strains. Because the widely used urinary antigen diagnostic test does not readily detect non-SG1 L. pneumophila, Legionnaires' disease caused by SG6 L. pneumophila is likely underreported worldwide.IMPORTANCEL. pneumophila is the leading cause of disease outbreaks associated with drinking water in the United States. Compared to what is known of the established risks of colonization within hospitals and hotels, relatively little is known about residential exposure to L. pneumophila One year after two outbreaks of Legionnaires' disease in Genesee County, MI, that coincided with damage to the Flint municipal water system, our multidisciplinary team launched an environmental surveillance and laboratory research campaign aimed at informing risk management strategies to provide safe public water supplies. The most prevalent L. pneumophila strains isolated from residential plumbing were closely related strains of SG6. In laboratory tests of virulence, the SG6 environmental isolates resembled SG1 clinical strains, yet they are not readily detected by the common diagnostic urinary antigen test, which is specific for SG1. Therefore, our study complements the existing epidemiological literature indicating that Legionnaires' disease due to non-SG1 strains is underreported around the globe.


Assuntos
Água Potável/microbiologia , Variação Genética , Legionella pneumophila/classificação , Legionella pneumophila/isolamento & purificação , Legionelose/microbiologia , Engenharia Sanitária , Sorogrupo , Humanos , Legionella pneumophila/genética , Macrófagos/microbiologia , Michigan , Viabilidade Microbiana , Tipagem de Sequências Multilocus , Prevalência
2.
Mol Biol Cell ; 29(5): 657-668, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29282279

RESUMO

As professional phagocytes, macrophages are susceptible to endolysosomal membrane damage inflicted by the pathogens and noxious particles they ingest. Whether macrophages have mechanisms for limiting such damage is not well understood. Previously, we reported a phenomenon, termed "inducible renitence," in which lipopolysaccharide (LPS) activation of macrophages protected their endolysosomes against damage initiated by the phagocytosis of silica beads. To gain mechanistic insight into the process, we analyzed the kinetics of renitence and morphological features of LPS-activated versus resting macrophages following silica bead-mediated injury. We discovered novel vacuolar structures that form in LPS-activated but not resting macrophages following silica bead phagocytosis. Because of their correlation with renitence and damage-resistant nature, we termed these structures "renitence vacuoles" (RVs). RVs formed coincident with silica bead uptake in a process associated with membrane ruffling and macropinocytosis. However, unlike normal macropinosomes (MPs), which shrink within 20 min of formation, RVs persisted around bead-containing phagosomes. RVs fused with lysosomes, whereas associated phagosomes typically did not. These findings are consistent with a model in which RVs, as persistent MPs, prevent fusion between damaged phagosomes and intact lysosomes and thereby preserve endolysosomal integrity.


Assuntos
Endossomos/metabolismo , Membranas Intracelulares/efeitos dos fármacos , Macrófagos/citologia , Fagossomos/fisiologia , Vacúolos/fisiologia , Animais , Lipopolissacarídeos/farmacologia , Lisossomos/fisiologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Pinocitose
4.
J Bacteriol ; 198(3): 553-64, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26598366

RESUMO

UNLABELLED: Bacterial evolution is accelerated by mobile genetic elements. To spread horizontally and to benefit the recipient bacteria, genes encoded on these elements must be properly regulated. Among the legionellae are multiple integrative conjugative elements (ICEs) that each encode a paralog of the broadly conserved regulator csrA. Using bioinformatic analyses, we deduced that specific csrA paralogs are coinherited with particular lineages of the type IV secretion system that mediates horizontal spread of its ICE, suggesting a conserved regulatory interaction. As a first step to investigate the contribution of csrA regulators to this class of mobile genetic elements, we analyzed here the activity of the csrA paralog encoded on Legionella pneumophila ICE-ßox. Deletion of this gene, which we name csrT, had no observed effect under laboratory conditions. However, ectopic expression of csrT abrogated the protection to hydrogen peroxide and macrophage degradation that ICE-ßox confers to L. pneumophila. When ectopically expressed, csrT also repressed L. pneumophila flagellin production and motility, a function similar to the core genome's canonical csrA. Moreover, csrT restored the repression of motility to csrA mutants of Bacillus subtilis, a finding consistent with the predicted function of CsrT as an mRNA binding protein. Since all known ICEs of legionellae encode coinherited csrA-type IV secretion system pairs, we postulate that CsrA superfamily proteins regulate ICE activity to increase their horizontal spread, thereby expanding L. pneumophila versatility. IMPORTANCE: ICEs are mobile DNA elements whose type IV secretion machineries mediate spread among bacterial populations. All surveyed ICEs within the Legionella genus also carry paralogs of the essential life cycle regulator csrA. It is striking that the csrA loci could be classified into distinct families based on either their sequence or the subtype of the adjacent type IV secretion system locus. To investigate whether ICE-encoded csrA paralogs are bona fide regulators, we analyzed ICE-ßox as a model system. When expressed ectopically, its csrA paralog inhibited multiple ICE-ßox phenotypes, as well as the motility of not only Legionella but also Bacillus subtilis. Accordingly, we predict that CsrA regulators equip legionellae ICEs to promote their spread via dedicated type IV secretion systems.


Assuntos
Proteínas de Bactérias/metabolismo , Conjugação Genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Legionella pneumophila/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Genes Reguladores , Legionella pneumophila/genética , Lisossomos , Macrófagos , Camundongos , Filogenia , Ligação Proteica , Transporte Proteico , RNA Bacteriano/genética , RNA Bacteriano/metabolismo
5.
mBio ; 5(3): e01091-14, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24781744

RESUMO

ABSTRACT Integrative conjugative elements (ICEs) are mobile blocks of DNA that can contribute to bacterial evolution by self-directed transmission of advantageous traits. Here, we analyze the activity of a putative 65-kb ICE harbored by Legionella pneumophila using molecular genetics, conjugation assays, a phenotype microarray screen, and macrophage infections. The element transferred to a naive L. pneumophila strain, integrated site-specifically, and conferred increased resistance to oxacillin, penicillin, hydrogen peroxide, and bleach. Furthermore, the element increased survival of L. pneumophila within restrictive mouse macrophages. In particular, this ICE protects L. pneumophila from phagocyte oxidase activity, since mutation of the macrophage NADPH oxidase eliminated the fitness difference between strains that carried and those that lacked the mobile element. Renamed ICE-ßox (for ß-lactam antibiotics and oxidative stress), this transposable element is predicted to contribute to the emergence of L. pneumophila strains that are more fit in natural and engineered water systems and in macrophages. IMPORTANCE Bacteria evolve rapidly by acquiring new traits via horizontal gene transfer. Integrative conjugative elements (ICEs) are mobile blocks of DNA that encode the machinery necessary to spread among bacterial populations. ICEs transfer antibiotic resistance and other bacterial survival factors as cargo genes carried within the element. Here, we show that Legionella pneumophila, the causative agent of Legionnaires' disease, carries ICE-ßox, which enhances the resistance of this opportunistic pathogen to bleach and ß-lactam antibiotics. Moreover, L. pneumophila strains encoding ICE-ßox are more resistant to macrophages that carry phagocyte oxidase. Accordingly, ICE-ßox is predicted to increase the fitness of L. pneumophila in natural and engineered waters and in humans. To our knowledge, this is the first description of an ICE that confers oxidative stress resistance to a nosocomial pathogen.


Assuntos
Elementos de DNA Transponíveis , Legionella pneumophila/fisiologia , Macrófagos/microbiologia , Estresse Oxidativo/genética , Animais , Feminino , Ordem dos Genes , Genes Bacterianos , Aptidão Genética , Loci Gênicos , Macrófagos/metabolismo , Camundongos , Mutagênese Insercional , NADPH Oxidases/metabolismo , Oxidantes/farmacologia , Resistência beta-Lactâmica/genética
6.
Infect Immun ; 82(2): 720-30, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478086

RESUMO

The phagosomal transporter (Pht) family of the major facilitator superfamily (MFS) is encoded by phylogenetically related intracellular gammaproteobacteria, including the opportunistic pathogen Legionella pneumophila. The location of the pht genes between the putative thymidine kinase (tdk) and phosphopentomutase (deoB) genes suggested that the phtC and phtD loci contribute to thymidine salvage in L. pneumophila. Indeed, a phtC(+) allele in trans restored pyrimidine uptake to an Escherichia coli mutant that lacked all known nucleoside transporters, whereas a phtD(+) allele did not. The results of phenotypic analyses of L. pneumophila strains lacking phtC or phtD strongly indicate that L. pneumophila requires PhtC and PhtD function under conditions where sustained dTMP synthesis is compromised. First, in broth cultures that mimicked thymidine limitation or starvation, L. pneumophila exhibited a marked requirement for PhtC function. Conversely, mutation of phtD conferred a survival advantage. Second, in medium that lacked thymidine, multicopy phtC(+) or phtD(+) alleles enhanced the survival of L. pneumophila thymidylate synthase (thyA)-deficient strains, which cannot synthesize dTMP endogenously. Third, under conditions in which transport of the pyrimidine nucleoside analog 5-fluorodeoxyuridine (FUdR) would inhibit growth, PhtC and PhtD conferred a growth advantage to L. pneumophila thyA(+) strains. Finally, when cultured in macrophages, L. pneumophila required the phtC-phtD locus to replicate. Accordingly, we propose that PhtC and PhtD contribute to protect L. pneumophila from dTMP starvation during its intracellular life cycle.


Assuntos
Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana Transportadoras/metabolismo , Timidina/metabolismo , Animais , Células Cultivadas , Meios de Cultura/química , Feminino , Deleção de Genes , Legionella pneumophila/genética , Proteínas de Membrana Transportadoras/genética , Camundongos , Viabilidade Microbiana
7.
mBio ; 4(1): e00620-12, 2013 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-23404401

RESUMO

UNLABELLED: When microbes contaminate the macrophage cytoplasm, leukocytes undergo a proinflammatory death that is initiated by nucleotide-binding-domain-, leucine-rich-repeat-containing proteins (NLR proteins) that bind and activate caspase-1. We report that these inflammasome components also regulate autophagy, a vesicular pathway to eliminate cytosolic debris. In response to infection with flagellate Legionella pneumophila, C57BL/6J mouse macrophages equipped with caspase-1 and the NLR proteins NAIP5 and NLRC4 stimulated autophagosome turnover. A second trigger of inflammasome assembly, K(+) efflux, also rapidly activated autophagy in macrophages that produced caspase-1. Autophagy protects infected macrophages from pyroptosis, since caspase-1-dependent cell death occurred more frequently when autophagy was dampened pharmacologically by either 3-methyladenine or an inhibitor of the Atg4 protease. Accordingly, in addition to coordinating pyroptosis, both (pro-) caspase-1 protein and NLR components of inflammasomes equip macrophages to recruit autophagy, a disposal pathway that raises the threshold of contaminants necessary to trigger proinflammatory leukocyte death. IMPORTANCE: An exciting development in the innate-immunity field is the recognition that macrophages enlist autophagy to protect their cytoplasm from infection. Nutrient deprivation has long been known to induce autophagy; how infection triggers this disposal pathway is an active area of research. Autophagy is encountered by many of the intracellular pathogens that are known to trigger pyroptosis, an inflammatory cell death initiated when nucleotide-binding-domain-, leucine-rich-repeat-containing proteins (NLR proteins) activate caspase-1 within inflammasome complexes. Therefore, we tested the hypothesis that NLR proteins and caspase-1 also coordinate autophagy as a barrier to cytosolic infection. By exploiting classical bacterial and mouse genetics and kinetic assays of autophagy, we demonstrate for the first time that, when confronted with cytosolic contamination, primary mouse macrophages rely not only on the NLR proteins NAIP5 and NLRC4 but also on (pro-)caspase-1 protein to mount a rapid autophagic response that wards off proinflammatory cell death.


Assuntos
Autofagia , Inflamassomos/metabolismo , Legionella pneumophila/imunologia , Macrófagos/imunologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Caspase 1/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Inibidora de Apoptose Neuronal/metabolismo
8.
Infect Immun ; 81(3): 945-55, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319553

RESUMO

In response to environmental fluctuations or stresses, bacteria can activate transcriptional and phenotypic programs to coordinate an adaptive response. The intracellular pathogen Legionella pneumophila converts from a noninfectious replicative form to an infectious transmissive form when the bacterium encounters alterations in either amino acid concentrations or fatty acid biosynthesis. Here, we report that L. pneumophila differentiation is also triggered by nicotinic acid, a precursor of the central metabolite NAD(+). In particular, when replicative L. pneumophila are treated with 5 mM nicotinic acid, the bacteria induce numerous transmissive-phase phenotypes, including motility, cytotoxicity toward macrophages, sodium sensitivity, and lysosome avoidance. Transcriptional profile analysis determined that nicotinic acid induces the expression of a panel of genes characteristic of transmissive-phase L. pneumophila. Moreover, an additional 213 genes specific to nicotinic acid treatment were altered. Although nearly 25% of these genes lack an assigned function, the gene most highly induced by nicotinic acid treatment encodes a putative major facilitator superfamily transporter, Lpg0273. Indeed, lpg0273 protects L. pneumophila from toxic concentrations of nicotinic acid as judged by analyzing the growth of the corresponding mutant. The broad utility of the nicotinic acid pathway to couple central metabolism and cell fate is underscored by this small metabolite's modulation of gene expression by diverse microbes, including Candida glabrata, Bordetella pertussis, Escherichia coli, and L. pneumophila.


Assuntos
Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Legionella pneumophila/efeitos dos fármacos , Legionella pneumophila/metabolismo , Niacina/farmacologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Proliferação de Células , Feminino , Legionella pneumophila/patogenicidade , Lisossomos , Macrófagos , Camundongos , Modelos Moleculares , Conformação Proteica , Fatores de Tempo , Transcriptoma , Virulência
9.
PLoS Pathog ; 6(3): e1000822, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20333253

RESUMO

Legionella pneumophila is an intracellular pathogen responsible for Legionnaires' disease. This bacterium uses the Dot/Icm type IV secretion system to inject a large number of bacterial proteins into host cells to facilitate the biogenesis of a phagosome permissive for its intracellular growth. Like many highly adapted intravacuolar pathogens, L. pneumophila is able to maintain a neutral pH in the lumen of its phagosome, particularly in the early phase of infection. However, in all cases, the molecular mechanisms underlying this observation remain unknown. In this report, we describe the identification and characterization of a Legionella protein termed SidK that specifically targets host v-ATPase, the multi-subunit machinery primarily responsible for organelle acidification in eukaryotic cells. Our results indicate that after being injected into infected cells by the Dot/Icm secretion system, SidK interacts with VatA, a key component of the proton pump. Such binding leads to the inhibition of ATP hydrolysis and proton translocation. When delivered into macrophages, SidK inhibits vacuole acidification and impairs the ability of the cells to digest non-pathogenic E. coli. We also show that a domain located in the N-terminal portion of SidK is responsible for its interactions with VatA. Furthermore, expression of sidK is highly induced when bacteria begin to enter new growth cycle, correlating well with the potential temporal requirement of its activity during infection. Our results indicate that direct targeting of v-ATPase by secreted proteins constitutes a virulence strategy for L. pneumophila, a vacuolar pathogen of macrophages and amoebae.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Legionella pneumophila/enzimologia , Doença dos Legionários/microbiologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Vacúolos/enzimologia , Ácidos/metabolismo , Animais , Células Cultivadas , Meios de Cultura , Feminino , Concentração de Íons de Hidrogênio , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Lisossomos/enzimologia , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos A , Fagossomos/enzimologia , Prótons , Saccharomyces cerevisiae/crescimento & desenvolvimento , Especificidade por Substrato , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores , Virulência
10.
Infect Immun ; 78(6): 2571-83, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20351136

RESUMO

When confronted with metabolic stress, replicative Legionella pneumophila bacteria convert to resilient, infectious cells equipped for transmission. Differentiation is promoted by the LetA/LetS two-component system, which belongs to a family of signal-transducing proteins that employ a four-step phosphorelay to regulate gene expression. Histidine 307 of LetS was essential to switch on the transmission profile, but a threonine substitution at position 311 (T311M) suggested a rheostat-like function. The letS(T311M) bacteria resembled the wild type (WT) for some traits and letS null mutants for others, whereas they displayed intermediate levels of infectivity, cytotoxicity, and lysosome evasion. Although only 30 to 50% of letS(T311M) mutants became motile, flow cytometry determined that every cell eventually activated the flagellin promoter to WT levels, but expression was delayed. Likewise, letS(T311M) mutants exhibited delayed induction of RsmY and RsmZ, regulatory RNAs that relieve CsrA repression of transmission traits. Transcriptional profile analysis revealed that letS(T311M) mutants expressed the flagellar regulon and multiple other transmissive-phase loci at a higher cell density than the WT. Accordingly, we postulate that the letS(T311M) mutant may relay phosphate less efficiently than the WT LetS sensor protein, leading to sluggish gene expression and a variety of phenotypic profiles. Thus, as first described for BvgA/BvgS, rather than acting as on/off switches, this family of two-component systems exhibit rheostat activity that likely confers versatility as microbes adapt to fluctuating environments.


Assuntos
Proteínas de Bactérias/fisiologia , Regulação Bacteriana da Expressão Gênica , Legionella pneumophila/fisiologia , Transdução de Sinais , Substituição de Aminoácidos/genética , Animais , Proteínas de Bactérias/genética , Deleção de Genes , Macrófagos/microbiologia , Camundongos , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Fosfatos/metabolismo
11.
Mol Microbiol ; 76(1): 200-19, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20199605

RESUMO

To transit between hosts, intracellular Legionella pneumophila transform into a motile, infectious, transmissive state. Here we exploit the pathogen's life cycle to examine how guanosine tetraphosphate (ppGpp) and DksA cooperate to govern bacterial differentiation. Transcriptional profiling revealed that during transmission alarmone accumulation increases the mRNA for flagellar and Type IV-secretion components, secreted host effectors and regulators, and decreases transcripts for translation, membrane modification and ATP synthesis machinery. DksA is critical for differentiation, since mutants are defective for stationary phase survival, flagellar gene activation, lysosome avoidance and macrophage cytotoxicity. The roles of ppGpp and DksA depend on the context. For macrophage transmission, ppGpp is essential, whereas DksA is dispensable, indicating that ppGpp can act autonomously. In broth, DksA promotes differentiation when ppGpp levels increase, or during fatty acid stress, as judged by flaA expression and evasion of degradation by macrophages. For flagella morphogenesis, DksA is required for basal fliA (sigma(28)) promoter activity. When alarmone levels increase, DksA cooperates with ppGpp to generate a pulse of Class II rod RNA or to amplify the Class III sigma factor and Class IV flagellin RNAs. Thus, DksA responds to the level of ppGpp and other stress signals to co-ordinate L. pneumophila differentiation.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica , Guanosina Tetrafosfato/metabolismo , Legionella pneumophila/fisiologia , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Adesão Celular , Sobrevivência Celular , Contagem de Colônia Microbiana , Flagelos/fisiologia , Deleção de Genes , Perfilação da Expressão Gênica , Legionella pneumophila/crescimento & desenvolvimento , Legionella pneumophila/metabolismo , Legionella pneumophila/patogenicidade , Macrófagos/microbiologia , Dados de Sequência Molecular , Fatores de Virulência/genética
12.
Infect Immun ; 78(1): 423-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19841075

RESUMO

Legionella pneumophila, a motile opportunistic pathogen of humans, is restricted from replicating in the lungs of C57BL/6 mice. Resistance of mouse macrophages to L. pneumophila depends on recognition of cytosolic flagellin. Once detected by the NOD-like receptors Naip5 and Ipaf (Nlrc4), flagellin triggers pyroptosis, a proinflammatory cell death. In contrast, motile strains of L. parisiensis and L. tucsonensis replicate profusely within C57BL/6 macrophages, similar to flagellin-deficient L. pneumophila. To gain insight into how motile species escape innate defense mechanisms of mice, we compared their impacts on macrophages. L. parisiensis and L. tucsonensis do not induce proinflammatory cell death, as measured by lactate dehydrogenase (LDH) release and interleukin-1beta (IL-1beta) secretion. However, flagellin isolated from L. parisiensis and L. tucsonensis triggers cell death and IL-1beta secretion when transfected into the cytosol of macrophages. Neither strain displays three characteristics of the canonical L. pneumophila Dot/Icm type IV secretion system: sodium sensitivity, LAMP-1 evasion, and pore formation. Therefore, we postulate that when L. parisiensis and L. tucsonensis invade a mouse macrophage, flagellin is confined to the phagosome, protecting the bacteria from recognition by the cytosolic surveillance system and allowing Legionella to replicate. Despite their superior capacity to multiply in mouse macrophages, L. parisiensis and L. tucsonensis have been associated with only two cases of disease, both in renal transplant patients. These results point to the complexity of disease, a product of the pathogenic potential of the microbe, as defined in the laboratory, and the capacity of the host to mount a measured defense.


Assuntos
Legionella/fisiologia , Macrófagos/microbiologia , Animais , Morte Celular , Células Cultivadas , Flagelina/genética , Flagelina/metabolismo , Variação Genética , Camundongos , Camundongos Endogâmicos C57BL
13.
Methods Enzymol ; 452: 383-402, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19200894

RESUMO

Macrophages enlist autophagy to combat infection by a variety of bacteria, viruses, and parasites. In response to this selective pressure, some pathogenic microbes have acquired strategies to evade or tolerate autophagy. Accordingly, infected cells may accumulate numerous autophagic vacuoles/autophagosomes when microbial products either stimulate their formation or inhibit their maturation. To distinguish between the two mechanisms, we describe methods to assess the impact of infection on the kinetics and amplitude of autophagosome formation and maturation within mouse macrophages by microscopy or Western analysis using antibodies specific for endogenous or recombinant LC3 protein.


Assuntos
Autofagia/fisiologia , Macrófagos/metabolismo , Fagossomos/metabolismo , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência
14.
Mol Microbiol ; 71(3): 640-58, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19040633

RESUMO

During its life cycle, Legionella pneumophila alternates between a replicative and a transmissive state. To determine their contributions to L. pneumophila differentiation, the two ppGpp synthetases, RelA and SpoT, were disrupted. Synthesis of ppGpp was required for transmission, as relA spoT mutants were killed during entry to and exit from macrophages. RelA, which senses amino acid starvation induced by serine hydroxamate, is dispensable in macrophages, as relA mutants spread efficiently. SpoT monitors fatty acid biosynthesis (FAB), since following cerulenin treatment, wild-type and relA strains expressed the flaA transmissive gene, but relA spoT mutants did not. As in Escherichia coli, the SpoT response to FAB perturbation likely required an interaction with acyl-carrier protein (ACP), as judged by the failure of the spoT-A413E allele to rescue transmissive trait expression of relA spoT bacteria. Furthermore, SpoT was essential for transmission between macrophages, since secondary infections by relA spoT mutants were restored by induction of spoT, but not relA. To resume replication, ppGpp must be degraded, as mutants lacking spoT hydrolase activity failed to convert from the transmissive to the replicative phase in either bacteriological medium or macrophages. Thus, L. pneumophila requires SpoT to monitor FAB and to alternate between replication and transmission in macrophages.


Assuntos
Proteínas de Bactérias/metabolismo , Legionella pneumophila/genética , Ligases/metabolismo , Macrófagos/microbiologia , Pirofosfatases/metabolismo , Animais , Proteínas de Bactérias/genética , Células Cultivadas , Feminino , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Legionella pneumophila/enzimologia , Legionella pneumophila/patogenicidade , Ligases/genética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos A , Mutagênese Insercional , Pirofosfatases/genética
15.
Autophagy ; 2(3): 179-82, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16874080

RESUMO

Autophagy is a conserved membrane traffic pathway that equips eukaryotic cells to capture cytoplasmic components within a double-membrane vacuole, or autophagosome, for delivery to lysosomes. Although best known as a mechanism to survive starvation, autophagy is now recognized to combat infection by a variety of microbes.(1-3) Not surprisingly, to establish a replication niche in host cells, some intracellular pathogens have acquired mechanisms either to evade or subvert the autophagic pathway. Because they are amenable to genetic manipulation, these microbes can be exploited as experimental tools to investigate the contribution of autophagy to immunity. Here we discuss the mouse macrophage response to L. pneumophila, the facultative intracellular bacterium responsible for an acute form of pneumonia, Legionnaire's disease.


Assuntos
Autofagia/fisiologia , Modelos Animais de Doenças , Legionella pneumophila/patogenicidade , Doença dos Legionários/microbiologia , Animais , Apoptose/fisiologia , Inflamação/microbiologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Vacúolos/microbiologia
16.
Infect Immun ; 74(6): 3285-95, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16714556

RESUMO

When cultured in broth to the transmissive phase, Legionella pneumophila infects macrophages by inhibiting phagosome maturation, whereas replicative-phase cells are transported to the lysosomes. Here we report that the ability of L. pneumophila to inhibit phagosome-lysosome fusion correlated with developmentally regulated modifications of the pathogen's surface, as judged by its lipopolysaccharide profile and by its binding to a sialic acid-specific lectin and to the hydrocarbon hexadecane. Likewise, the composition of membrane vesicles shed by L. pneumophila was developmentally regulated, based on binding to the lectin and to the lipopolysaccharide-specific monoclonal antibody 3/1. Membrane vesicles were sufficient to inhibit phagosome-lysosome fusion by a mechanism independent of type IV secretion, since only approximately 25% of beads suspended with or coated by vesicles from transmissive phase wild type or dotA secretion mutants colocalized with lysosomal probes, whereas approximately 75% of beads were lysosomal when untreated or presented with vesicles from the L. pneumophila letA regulatory mutant or E. coli. As observed previously for L. pneumophila infection of mouse macrophages, vesicles inhibited phagosome-lysosome fusion only temporarily; by 10 h after treatment with vesicles, macrophages delivered approximately 72% of ingested beads to lysosomes. Accordingly, in the context of the epidemiology of the pneumonia Legionnaires' disease and virulence mechanisms of Leishmania and Mycobacteria, we discuss a model here in which L. pneumophila developmentally regulates its surface composition and releases vesicles into phagosomes that inhibit their fusion with lysosomes.


Assuntos
Legionella pneumophila/patogenicidade , Lisossomos/fisiologia , Fusão de Membrana , Fagossomos/fisiologia , Animais , Membrana Celular/química , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Lipopolissacarídeos/análise , Lipopolissacarídeos/metabolismo , Macrófagos/microbiologia , Camundongos
17.
J Exp Med ; 203(4): 1093-104, 2006 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-16606669

RESUMO

To restrict infection by Legionella pneumophila, mouse macrophages require Naip5, a member of the nucleotide-binding oligomerization domain leucine-rich repeat family of pattern recognition receptors, which detect cytoplasmic microbial products. We report that mouse macrophages restricted L. pneumophila replication and initiated a proinflammatory program of cell death when flagellin contaminated their cytosol. Nuclear condensation, membrane permeability, and interleukin-1beta secretion were triggered by type IV secretion-competent bacteria that encode flagellin. The macrophage response to L. pneumophila was independent of Toll-like receptor signaling but correlated with Naip5 function and required caspase 1 activity. The L. pneumophila type IV secretion system provided only pore-forming activity because listeriolysin O of Listeria monocytogenes could substitute for its contribution. Flagellin monomers appeared to trigger the macrophage response from perforated phagosomes: once heated to disassemble filaments, flagellin triggered cell death but native flagellar preparations did not. Flagellin made L. pneumophila vulnerable to innate immune mechanisms because Naip5+ macrophages restricted the growth of virulent microbes, but flagellin mutants replicated freely. Likewise, after intratracheal inoculation of Naip5+ mice, the yield of L. pneumophila in the lungs declined, whereas the burden of flagellin mutants increased. Accordingly, macrophages respond to cytosolic flagellin by a mechanism that requires Naip5 and caspase 1 to restrict bacterial replication and release proinflammatory cytokines that control L. pneumophila infection.


Assuntos
Citosol/imunologia , Flagelina/imunologia , Legionella pneumophila/imunologia , Macrófagos/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose/fisiologia , Células Cultivadas , Feminino , Imunidade Inata , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , Proteína Inibidora de Apoptose Neuronal/fisiologia , Transdução de Sinais/fisiologia , Receptores Toll-Like/fisiologia
18.
Infect Immun ; 73(9): 5720-34, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16113289

RESUMO

Legionella pneumophila is a motile intracellular pathogen of macrophages and amoebae. When nutrients become scarce, the bacterium induces expression of transmission traits, some of which are dependent on the flagellar sigma factor FliA (sigma(28)). To test how particular components of the L. pneumophila flagellar regulon contribute to virulence, we compared a fliA mutant with strains whose flagellar construction is disrupted at various stages. We find that L. pneumophila requires FliA to avoid lysosomal degradation in murine bone marrow-derived macrophages (BMM), to regulate production of a melanin-like pigment, and to regulate binding to the dye crystal violet, whereas motility, flagellar secretion, and external flagella or flagellin are dispensable for these activities. Thus, in addition to flagellar genes, the FliA sigma factor regulates an effector(s) or regulator(s) that contributes to other transmissive traits, notably inhibition of phagosome maturation. Whether or not the microbes produced flagellin, all nonmotile L. pneumophila mutants bound BMM less efficiently than the wild type, resulting in poor infectivity and a loss of contact-dependent death of BMM. Therefore, bacterial motility increases contact with host cells during infection, but flagellin is not an adhesin. When BMM contact by each nonmotile strain was promoted by centrifugation, all the mutants bound BMM similarly, but only those microbes that synthesized flagellin induced BMM death. Thus, the flagellar regulon equips the aquatic pathogen L. pneumophila to coordinate motility with multiple traits vital to virulence.


Assuntos
Flagelos/fisiologia , Legionella pneumophila/enzimologia , Legionella pneumophila/imunologia , Doença dos Legionários , Macrófagos/microbiologia , Muramidase/fisiologia , Regulon/fisiologia , Animais , Proteínas de Bactérias/fisiologia , Morte Celular , Células Cultivadas , Feminino , Flagelina/metabolismo , Temperatura Alta , Legionella pneumophila/genética , Legionella pneumophila/patogenicidade , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos A , Fator sigma/fisiologia , Sódio/farmacologia , Virulência
19.
Infect Immun ; 73(8): 4494-504, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16040960

RESUMO

Legionella pneumophila and Coxiella burnetii are phylogenetically related intracellular bacteria that cause aerosol-transmitted lung infections. In host cells both pathogens proliferate in vacuoles whose biogenesis displays some common features. To test the functional similarity of their respective intracellular niches, African green monkey kidney epithelial (Vero) cells, A/J mouse bone marrow-derived macrophages, human macrophages, and human dendritic cells (DC) containing mature C. burnetii replication vacuoles were superinfected with L. pneumophila, and then the acidity, lysosome-associated membrane protein (LAMP) content, and cohabitation of mature replication vacuoles was assessed. In all cell types, wild-type L. pneumophila occupied distinct vacuoles in close association with acidic, LAMP-positive C. burnetii replication vacuoles. In murine macrophages, but not primate macrophages, DC, or epithelial cells, L. pneumophila replication vacuoles were acidic and LAMP positive. Unlike wild-type L. pneumophila, type IV secretion-deficient dotA mutants trafficked to lysosome-like C. burnetii vacuoles in Vero cells where they survived but failed to replicate. In primate macrophages, DC, or epithelial cells, growth of L. pneumophila was as robust in superinfected cell cultures as in those singly infected. Thus, despite their noted similarities, L. pneumophila and C. burnetii are exquisitely adapted for replication in unique replication vacuoles, and factors that maintain the C. burnetii replication vacuole do not alter biogenesis of an adjacent L. pneumophila replication vacuole. Moreover, L. pneumophila can replicate efficiently in either lysosomal vacuoles of A/J mouse cells or in nonlysosomal vacuoles of primate cells.


Assuntos
Coxiella burnetii/metabolismo , Legionella pneumophila/metabolismo , Doença dos Legionários/microbiologia , Vacúolos/microbiologia , Animais , Antígenos de Bactérias , Proliferação de Células , Chlorocebus aethiops , Coxiella burnetii/patogenicidade , Células Dendríticas/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Legionella pneumophila/patogenicidade , Macrófagos/microbiologia , Camundongos , Febre Q/microbiologia , Células Vero , Virulência
20.
Proc Natl Acad Sci U S A ; 102(28): 9924-9, 2005 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-15998735

RESUMO

Differentiation in response to environmental cues is integral to the success of many intracellular pathogens. By characterizing a Legionella pneumophila mutant defective for differentiation in broth and replication in macrophages, we identified a subfamily of major facilitator superfamily transporters, here named Pht (phagosomal transporter), that also is conserved in two other vacuolar pathogens, Coxiella burnetii and Francisella tularensis. Biolog phenotype microarray analysis indicated that PhtA transports threonine, an essential amino acid. Either excess threonine or threonine peptides bypass phtA function. In minimal medium, phtA mutants do not replicate; in rich broth, the bacteria prematurely differentiate to the transmissive phase, as judged by the kinetics of flaA-gfp expression, heat resistance, and sodium sensitivity. PhtA is dispensable for transmissive L. pneumophila to establish and persist within a replication vacuole but is essential for their differentiation to the replicative phase, based on phenotypic and RT-PCR analysis. Accordingly, we propose that the Pht transporter family equips transmissive L. pneumophila, C. burnetii, and F. tularensis to assess their phagosomal nutrient supply before committing to reenter the cell cycle.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Legionella pneumophila/metabolismo , Legionella pneumophila/fisiologia , Macrófagos/metabolismo , Fagossomos/metabolismo , Fenótipo , Coxiella burnetii/metabolismo , Primers do DNA , Francisella tularensis/metabolismo , Teste de Complementação Genética , Análise em Microsséries , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Treonina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA