Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
mBio ; 15(6): e0071024, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38682908

RESUMEN

The causative agent of Legionnaires' disease, Legionella pneumophila, is an environmental bacterium, that replicates in macrophages, parasitizes amoeba, and forms biofilms. L. pneumophila employs the Legionella quorum sensing (Lqs) system and the transcription factor LvbR to control various bacterial traits, including virulence and biofilm architecture. LvbR negatively regulates the nitric oxide (NO) receptor Hnox1, linking quorum sensing to NO signaling. Here, we assessed the response of L. pneumophila to NO and investigated bacterial receptors underlying this process. Chemical NO donors, such as dipropylenetriamine (DPTA) NONOate and sodium nitroprusside (SNP), delayed and reduced the expression of the promoters for flagellin (PflaA) and the 6S small regulatory RNA (P6SRNA). Marker-less L. pneumophila mutant strains lacking individual (Hnox1, Hnox2, or NosP) or all three NO receptors (triple knockout, TKO) grew like the parental strain in media. However, in the TKO strain, the reduction of PflaA expression by DPTA NONOate was less pronounced, suggesting that the NO receptors are implicated in NO signaling. In the ΔnosP mutant, the lvbR promoter was upregulated, indicating that NosP negatively regulates LvbR. The single and triple NO receptor mutant strains were impaired for growth in phagocytes, and phenotypic heterogeneity of non-growing/growing bacteria in amoebae was regulated by the NO receptors. The single NO receptor and TKO mutant strains showed altered biofilm architecture and lack of response of biofilms to NO. In summary, we provide evidence that L. pneumophila regulates virulence, intracellular phenotypic heterogeneity, and biofilm formation through NO and three functionally non-redundant NO receptors, Hnox1, Hnox2, and NosP. IMPORTANCE: The highly reactive diatomic gas molecule nitric oxide (NO) is produced by eukaryotes and bacteria to promote short-range and transient signaling within and between neighboring cells. Despite its importance as an inter-kingdom and intra-bacterial signaling molecule, the bacterial response and the underlying components of the signaling pathways are poorly characterized. The environmental bacterium Legionella pneumophila forms biofilms and replicates in protozoan and mammalian phagocytes. L. pneumophila harbors three putative NO receptors, one of which crosstalks with the Legionella quorum sensing (Lqs)-LvbR network to regulate various bacterial traits, including virulence and biofilm architecture. In this study, we used pharmacological, genetic, and cell biological approaches to assess the response of L. pneumophila to NO and to demonstrate that the putative NO receptors are implicated in NO detection, bacterial replication in phagocytes, intracellular phenotypic heterogeneity, and biofilm formation.


Asunto(s)
Proteínas Bacterianas , Biopelículas , Regulación Bacteriana de la Expresión Génica , Legionella pneumophila , Óxido Nítrico , Transducción de Señal , Biopelículas/crecimiento & desarrollo , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Legionella pneumophila/fisiología , Legionella pneumophila/metabolismo , Óxido Nítrico/metabolismo , Virulencia , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fenotipo , Macrófagos/microbiología , Percepción de Quorum
2.
Mol Microbiol ; 120(2): 194-209, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37429596

RESUMEN

Lipid droplets (LDs) are dynamic and versatile organelles present in most eukaryotic cells. LDs consist of a hydrophobic core of neutral lipids, a phospholipid monolayer coat, and a variety of associated proteins. LDs are formed at the endoplasmic reticulum and have diverse roles in lipid storage, energy metabolism, membrane trafficking, and cellular signaling. In addition to their physiological cellular functions, LDs have been implicated in the pathogenesis of several diseases, including metabolic disorders, cancer, and infections. A number of intracellular bacterial pathogens modulate and/or interact with LDs during host cell infection. Members of the genera Mycobacterium, Legionella, Coxiella, Chlamydia, and Salmonella exploit LDs as a source of intracellular nutrients and membrane components to establish their distinct intracellular replicative niches. In this review, we focus on the biogenesis, interactions, and functions of LDs, as well as on their role in lipid metabolism of intracellular bacterial pathogens.


Asunto(s)
Dieta , Gotas Lipídicas , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos
3.
Elife ; 122023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37158597

RESUMEN

The amoeba-resistant bacterium Legionella pneumophila causes Legionnaires' disease and employs a type IV secretion system (T4SS) to replicate in the unique, ER-associated Legionella-containing vacuole (LCV). The large fusion GTPase Sey1/atlastin is implicated in ER dynamics, ER-derived lipid droplet (LD) formation, and LCV maturation. Here, we employ cryo-electron tomography, confocal microscopy, proteomics, and isotopologue profiling to analyze LCV-LD interactions in the genetically tractable amoeba Dictyostelium discoideum. Dually fluorescence-labeled D. discoideum producing LCV and LD markers revealed that Sey1 as well as the L. pneumophila T4SS and the Ran GTPase activator LegG1 promote LCV-LD interactions. In vitro reconstitution using purified LCVs and LDs from parental or Δsey1 mutant D. discoideum indicated that Sey1 and GTP promote this process. Sey1 and the L. pneumophila fatty acid transporter FadL were implicated in palmitate catabolism and palmitate-dependent intracellular growth. Taken together, our results reveal that Sey1 and LegG1 mediate LD- and FadL-dependent fatty acid metabolism of intracellular L. pneumophila.


Asunto(s)
Dictyostelium , Legionella pneumophila , Legionella , Enfermedad de los Legionarios , Humanos , Legionella pneumophila/metabolismo , GTP Fosfohidrolasas/metabolismo , Macrófagos/metabolismo , Dictyostelium/metabolismo , Gotas Lipídicas/metabolismo , Vacuolas/metabolismo , Legionella/metabolismo , Enfermedad de los Legionarios/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
4.
Sci Adv ; 8(41): eabn6845, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36223470

RESUMEN

Membrane proteins are currently investigated after detergent extraction from native cellular membranes and reconstitution into artificial liposomes or nanodiscs, thereby removing them from their physiological environment. However, to truly understand the biophysical properties of membrane proteins in a physiological environment, they must be investigated within living cells. Here, we used a spin-labeled nanobody to interrogate the conformational cycle of the ABC transporter MsbA by double electron-electron resonance. Unexpectedly, the wide inward-open conformation of MsbA, commonly considered a nonphysiological state, was found to be prominently populated in Escherichia coli cells. Molecular dynamics simulations revealed that extensive lateral portal opening is essential to provide access of its large natural substrate core lipid A to the binding cavity. Our work paves the way to investigate the conformational landscape of membrane proteins in cells.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Proteínas de Escherichia coli , Escherichia coli , Transportadoras de Casetes de Unión a ATP/química , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/metabolismo , Detergentes/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Lípido A , Liposomas/metabolismo , Proteínas de la Membrana/metabolismo , Conformación Proteica
5.
Cell Microbiol ; 23(5): e13318, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33583106

RESUMEN

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


Asunto(s)
Dictyostelium/fisiología , Retículo Endoplásmico/ultraestructura , GTP Fosfohidrolasas/metabolismo , Legionella pneumophila/fisiología , Proteínas Protozoarias/metabolismo , Vacuolas/microbiología , Dictyostelium/crecimiento & desarrollo , Dictyostelium/microbiología , Dictyostelium/ultraestructura , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Retículo Endoplásmico Rugoso/microbiología , Retículo Endoplásmico Rugoso/fisiología , GTP Fosfohidrolasas/genética , Homeostasis , Interacciones Huésped-Patógeno , Legionella pneumophila/crecimiento & desarrollo , Movimiento , Muramidasa/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Protozoarias/genética , Vacuolas/fisiología
6.
Front Immunol ; 11: 25, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32117224

RESUMEN

Legionella pneumophila is the causative agent of a severe pneumonia called Legionnaires' disease. The environmental bacterium replicates in free-living amoebae as well as in lung macrophages in a distinct compartment, the Legionella-containing vacuole (LCV). The LCV communicates with a number of cellular vesicle trafficking pathways and is formed by a plethora of secreted bacterial effector proteins, which target host cell proteins and lipids. Phosphoinositide (PI) lipids are pivotal determinants of organelle identity, membrane dynamics and vesicle trafficking. Accordingly, eukaryotic cells tightly regulate the production, turnover, interconversion, and localization of PI lipids. L. pneumophila modulates the PI pattern in infected cells for its own benefit by (i) recruiting PI-decorated vesicles, (ii) producing effectors acting as PI interactors, phosphatases, kinases or phospholipases, and (iii) subverting host PI metabolizing enzymes. The PI conversion from PtdIns(3)P to PtdIns(4)P represents a decisive step during LCV maturation. In this review, we summarize recent progress on elucidating the strategies, by which L. pneumophila subverts host PI lipids to promote LCV formation and intracellular replication.


Asunto(s)
Interacciones Huésped-Patógeno , Legionella pneumophila/enzimología , Enfermedad de los Legionarios/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Fosfatidilinositoles/metabolismo , Vacuolas/metabolismo , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/microbiología , Humanos , Enfermedad de los Legionarios/microbiología , Vesículas Secretoras/metabolismo , Vesículas Transportadoras/metabolismo
7.
mBio ; 11(2)2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32209684

RESUMEN

Legionella pneumophila governs its interactions with host cells by secreting >300 different "effector" proteins. Some of these effectors contain eukaryotic domains such as the RCC1 (regulator of chromosome condensation 1) repeats promoting the activation of the small GTPase Ran. In this report, we reveal a conserved pattern of L. pneumophila RCC1 repeat genes, which are distributed in two main clusters of strains. Accordingly, strain Philadelphia-1 contains two RCC1 genes implicated in bacterial virulence, legG1 (Legionella eukaryotic gene 1), and ppgA, while strain Paris contains only one, pieG The RCC1 repeat effectors localize to different cellular compartments and bind distinct components of the Ran GTPase cycle, including Ran modulators and the small GTPase itself, and yet they all promote the activation of Ran. The pieG gene spans the corresponding open reading frames of legG1 and a separate adjacent upstream gene, lpg1975legG1 and lpg1975 are fused upon addition of a single nucleotide to encode a protein that adopts the binding specificity of PieG. Thus, a point mutation in pieG splits the gene, altering the effector target. These results indicate that divergent evolution of RCC1 repeat effectors defines the Ran GTPase cycle targets and that modulation of different components of the cycle might fine-tune Ran activation during Legionella infection.IMPORTANCELegionella pneumophila is a ubiquitous environmental bacterium which, upon inhalation, causes a life-threatening pneumonia termed Legionnaires' disease. The opportunistic pathogen grows in amoebae and macrophages by employing a "type IV" secretion system, which secretes more than 300 different "effector" proteins into the host cell, where they subvert pivotal processes. The function of many of these effector proteins is unknown, and their evolution has not been studied. L. pneumophila RCC1 repeat effectors target the small GTPase Ran, a molecular switch implicated in different cellular processes such as nucleocytoplasmic transport and microtubule cytoskeleton dynamics. We provide evidence that one or more RCC1 repeat genes are distributed in two main clusters of L. pneumophila strains and have divergently evolved to target different components of the Ran GTPase activation cycle at different subcellular sites. Thus, L. pneumophila employs a sophisticated strategy to subvert host cell Ran GTPase during infection.


Asunto(s)
Proteínas Bacterianas/genética , Evolución Molecular , Interacciones Huésped-Patógeno , Legionella pneumophila/genética , Proteína de Unión al GTP ran/genética , Células A549 , Animales , Dictyostelium/microbiología , Células HEK293 , Humanos , Legionella pneumophila/patogenicidad , Macrófagos/microbiología , Ratones , Células RAW 264.7 , Proteína de Unión al GTP ran/metabolismo
8.
Nat Commun ; 10(1): 5216, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31740681

RESUMEN

The facultative intracellular bacterium Legionella pneumophila replicates in environmental amoebae and in lung macrophages, and causes Legionnaires' disease. Here we show that L. pneumophila reversibly forms replicating and nonreplicating subpopulations of similar size within amoebae. The nonreplicating bacteria are viable and metabolically active, display increased antibiotic tolerance and a distinct proteome, and show high virulence as well as the capacity to form a degradation-resistant compartment. Upon infection of naïve or interferon-γ-activated macrophages, the nonreplicating subpopulation comprises ca. 10% or 50%, respectively, of the total intracellular bacteria; hence, the nonreplicating subpopulation is of similar size in amoebae and activated macrophages. The numbers of nonreplicating bacteria within amoebae are reduced in the absence of the autoinducer synthase LqsA or other components of the Lqs quorum-sensing system. Our results indicate that virulent, antibiotic-tolerant subpopulations of L. pneumophila are formed during infection of evolutionarily distant phagocytes, in a process controlled by the Lqs system.


Asunto(s)
Legionella/patogenicidad , Legionelosis/microbiología , Macrófagos/microbiología , Percepción de Quorum , Amoeba/microbiología , Animales , Proteínas Bacterianas/metabolismo , Espacio Intracelular/microbiología , Legionella/crecimiento & desarrollo , Ratones , Viabilidad Microbiana , Proteoma/metabolismo , Vacuolas/microbiología , Virulencia
9.
PLoS Pathog ; 15(2): e1007551, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30730983

RESUMEN

By engulfing potentially harmful microbes, professional phagocytes are continually at risk from intracellular pathogens. To avoid becoming infected, the host must kill pathogens in the phagosome before they can escape or establish a survival niche. Here, we analyse the role of the phosphoinositide (PI) 5-kinase PIKfyve in phagosome maturation and killing, using the amoeba and model phagocyte Dictyostelium discoideum. PIKfyve plays important but poorly understood roles in vesicular trafficking by catalysing formation of the lipids phosphatidylinositol (3,5)-bisphosphate (PI(3,5)2) and phosphatidylinositol-5-phosphate (PI(5)P). Here we show that its activity is essential during early phagosome maturation in Dictyostelium. Disruption of PIKfyve inhibited delivery of both the vacuolar V-ATPase and proteases, dramatically reducing the ability of cells to acidify newly formed phagosomes and digest their contents. Consequently, PIKfyve- cells were unable to generate an effective antimicrobial environment and efficiently kill captured bacteria. Moreover, we demonstrate that cells lacking PIKfyve are more susceptible to infection by the intracellular pathogen Legionella pneumophila. We conclude that PIKfyve-catalysed phosphoinositide production plays a crucial and general role in ensuring early phagosomal maturation, protecting host cells from diverse pathogenic microbes.


Asunto(s)
Dictyostelium/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Adenosina Trifosfatasas , Animales , Línea Celular , Dictyostelium/patogenicidad , Humanos , Hidrolasas/metabolismo , Legionella pneumophila/patogenicidad , Legionelosis/metabolismo , Macrófagos , Fagocitosis , Fagosomas , Fosfatidilinositol 3-Quinasas/fisiología , Fosfatidilinositoles , Transporte de Proteínas , Infecciones por Protozoos/metabolismo
10.
Cell Microbiol ; 21(6): e13008, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30656819

RESUMEN

The causative agent of tuberculosis, Mycobacterium tuberculosis, and its close relative Mycobacterium marinum manipulate phagocytic host cells, thereby creating a replication-permissive compartment termed the Mycobacterium-containing vacuole (MCV). The phosphoinositide (PI) lipid pattern is a crucial determinant of MCV formation and is targeted by mycobacterial PI phosphatases. In this study, we establish an efficient phage transduction protocol to construct defined M. marinum deletion mutants lacking one or three phosphatases, PtpA, PtpB, and/or SapM. These strains were defective for intracellular replication in macrophages and amoebae, and the growth defect was complemented by the corresponding plasmid-borne genes. Fluorescence microscopy of M. marinum-infected Dictyostelium discoideum revealed that MCVs harbouring mycobacteria lacking PtpA, SapM, or all three phosphatases accumulate significantly more phosphatidylinositol-3-phosphate (PtdIns3P) compared with MCVs containing the parental strain. Moreover, PtpA reduced MCV acidification by blocking the recruitment of the V-ATPase, and all three phosphatases promoted bacterial escape from the pathogen vacuole to the cytoplasm. In summary, the secreted M. marinum phosphatases PtpA, PtpB, and SapM determine the MCV PI pattern, compartment acidification, and phagosomal escape.


Asunto(s)
Citosol/metabolismo , Mycobacterium marinum/crecimiento & desarrollo , Fagosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Vacuolas/metabolismo , Acanthamoeba castellanii/microbiología , Adenosina Trifosfatasas/metabolismo , Amoeba/microbiología , Animales , Proteínas Bacterianas/metabolismo , Dictyostelium/metabolismo , Dictyostelium/microbiología , Interacciones Huésped-Patógeno/genética , Macrófagos/enzimología , Macrófagos/microbiología , Ratones , Microscopía Fluorescente , Mycobacterium marinum/enzimología , Mycobacterium marinum/genética , Mycobacterium marinum/patogenicidad , Proteínas Tirosina Fosfatasas/metabolismo , Células RAW 264.7 , Vacuolas/microbiología
11.
Methods Mol Biol ; 1921: 221-238, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30694495

RESUMEN

Legionella pneumophila is a facultative intracellular bacterium, which grows in amoebae as well as in macrophages and epithelial cells. Depletion of genes of interest by RNA interference (RNAi) has proven to be a robust and economic technique to study L. pneumophila-host cell interactions. Predesigned and often validated double-stranded (ds) RNA oligonucleotides that silence specific genes are commercially available. RNAi results in a reduced level of distinct proteins, which allows studying the specific role of host cell components involved in L. pneumophila infection. Here, we describe how to assess RNAi-mediated protein depletion efficiency and cytotoxic effects in human A549 lung epithelial cells and murine RAW 264.7 macrophages. Moreover, we demonstrate how RNAi can be used to screen for novel host cell proteins involved in the formation of the Legionella-containing vacuole and intracellular replication of the pathogen.


Asunto(s)
Interacciones Huésped-Patógeno/genética , Legionella/fisiología , Legionelosis/genética , Legionelosis/microbiología , Interferencia de ARN , Animales , Línea Celular , Supervivencia Celular , Citometría de Flujo , Expresión Génica , Genes Reporteros , Humanos , Legionella pneumophila/fisiología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Sistemas de Secreción Tipo IV , Vacuolas/metabolismo , Vacuolas/microbiología
12.
Small GTPases ; 10(5): 336-342, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-28632994

RESUMEN

Rap1 belongs to the Ras family of small GTPases, which are involved in a multitude of cellular signal transduction pathways and have extensively been linked to cancer biogenesis and metastasis. The small GTPase is activated in response to various extracellular and intracellular cues. Rap1 has conserved functions in Dictyostelium discoideum amoeba and mammalian cells, which are important for cell polarity, substrate and cell-cell adhesion and other processes that involve the regulation of cytoskeletal dynamics. Moreover, our recent study has shown that Rap1 is required for the formation of the replication-permissive vacuole of an intracellular bacterial pathogen. Here we review the function and regulation of Rap1 in these distinct processes, and we discuss the underlying signal transduction pathways.


Asunto(s)
Infecciones Bacterianas/metabolismo , Citoesqueleto/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Proteínas de Unión a Telómeros/metabolismo , Animales , Infecciones Bacterianas/patología , Citoesqueleto/patología , Dictyostelium/metabolismo , Dictyostelium/microbiología , Humanos , Metástasis de la Neoplasia , Neoplasias/microbiología , Neoplasias/patología , Proteínas Protozoarias/metabolismo , Complejo Shelterina
13.
mBio ; 9(6)2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30538188

RESUMEN

Legionella pneumophila is the causative agent of a pneumonia termed Legionnaires' disease. The facultative intracellular bacterium employs the Icm/Dot type IV secretion system (T4SS) and a plethora of translocated "effector" proteins to interfere with host vesicle trafficking pathways and establish a replicative niche, the Legionella-containing vacuole (LCV). Internalization of the pathogen and the events immediately ensuing are accompanied by host cell-mediated phosphoinositide (PI) lipid changes and the Icm/Dot-controlled conversion of the LCV from a PtdIns(3)P-positive vacuole into a PtdIns(4)P-positive replication-permissive compartment, which tightly associates with the endoplasmic reticulum. The source and formation of PtdIns(4)P are ill-defined. Using dually labeled Dictyostelium discoideum amoebae and real-time high-resolution confocal laser scanning microscopy (CLSM), we show here that nascent LCVs continuously capture and accumulate PtdIns(4)P-positive vesicles from the host cell. Trafficking of these PtdIns(4)P-positive vesicles to LCVs occurs independently of the Icm/Dot system, but their sustained association requires a functional T4SS. During the infection, PtdIns(3)P-positive membranes become compacted and segregated from the LCV, and PtdIns(3)P-positive vesicles traffic to the LCV but do not fuse. Moreover, using eukaryotic and prokaryotic PtdIns(4)P probes (2×PHFAPP-green fluorescent protein [2×PHFAPP-GFP] and P4CSidC-GFP, respectively) along with Arf1-GFP, we show that PtdIns(4)P-rich membranes of the trans-Golgi network associate with the LCV. Intriguingly, the interaction dynamics of 2×PHFAPP-GFP and P4CSidC-GFP are spatially separable and reveal the specific PtdIns(4)P pool from which the LCV PI originates. These findings provide high-resolution real-time insights into how L. pneumophila exploits the cellular dynamics of membrane-bound PtdIns(4)P for LCV formation.IMPORTANCE The environmental bacterium Legionella pneumophila causes a life-threatening pneumonia termed Legionnaires' disease. The bacteria grow intracellularly in free-living amoebae as well as in respiratory tract macrophages. To this end, L. pneumophila forms a distinct membrane-bound compartment called the Legionella-containing vacuole (LCV). Phosphoinositide (PI) lipids are crucial regulators of the identity and dynamics of host cell organelles. The PI lipid PtdIns(4)P is a hallmark of the host cell secretory pathway, and decoration of LCVs with this PI is required for pathogen vacuole maturation. The source, dynamics, and mode of accumulation of PtdIns(4)P on LCVs are largely unknown. Using Dictyostelium amoebae producing different fluorescent probes as host cells, we show here that LCVs rapidly acquire PtdIns(4)P through the continuous interaction with PtdIns(4)P-positive host vesicles derived from the Golgi apparatus. Thus, the PI lipid pattern of the secretory pathway contributes to the formation of the replication-permissive pathogen compartment.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Dictyostelium/microbiología , Aparato de Golgi/metabolismo , Legionella pneumophila/crecimiento & desarrollo , Fosfatos de Fosfatidilinositol/análisis , Vacuolas/metabolismo , Vesículas Citoplasmáticas/química , Microscopía Confocal , Sistemas de Secreción Tipo IV/metabolismo , Vacuolas/microbiología
14.
Methods Mol Biol ; 1841: 59-76, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30259480

RESUMEN

The causative agent of Legionnaires' disease, Legionella pneumophila, replicates in free-living amoebae as well as in macrophages of the innate immune system within a distinct membrane-bound compartment, the "Legionella-containing-vacuole" (LCV). LCV formation is a complex process and requires the bacterial Icm/Dot type IV secretion system, which translocates approximately 300 different "effector" proteins. Intact LCVs from infected Dictyostelium discoideum amoebae or RAW 264.7 murine macrophages can be purified using a straightforward protocol. In the first step, the LCVs in cell homogenates are tagged with an antibody directed against an L. pneumophila effector protein specifically localizing to the pathogen vacuole membrane and isolated by immunomagnetic separation using a secondary antibody coupled to magnetic beads. In the second step, the LCVs are further enriched by density gradient centrifugation through a Histodenz cushion. LCVs thus purified are analyzed by mass spectrometry-based proteomics and characterized by biochemical and cell biological approaches.


Asunto(s)
Metabolismo Energético , Interacciones Huésped-Patógeno , Legionella/metabolismo , Proteómica , Proteínas Protozoarias/metabolismo , Vacuolas/metabolismo , Amoeba/metabolismo , Amoeba/microbiología , Animales , Fraccionamiento Celular/métodos , Centrifugación por Gradiente de Densidad , Cromatografía Liquida , Macrófagos/metabolismo , Macrófagos/microbiología , Espectrometría de Masas , Ratones , Fagocitos/inmunología , Fagocitos/metabolismo , Fagocitos/microbiología , Proteómica/métodos , Células RAW 264.7 , Sistemas de Secreción Tipo IV
15.
Artículo en Inglés | MEDLINE | ID: mdl-29552544

RESUMEN

Environmental bacteria of the genus Legionella naturally parasitize free-living amoebae. Upon inhalation of bacteria-laden aerosols, the opportunistic pathogens grow intracellularly in alveolar macrophages and can cause a life-threatening pneumonia termed Legionnaires' disease. Intracellular replication in amoebae and macrophages takes place in a unique membrane-bound compartment, the Legionella-containing vacuole (LCV). LCV formation requires the bacterial Icm/Dot type IV secretion system, which translocates literally hundreds of "effector" proteins into host cells, where they modulate crucial cellular processes for the pathogen's benefit. The mechanism of LCV formation appears to be evolutionarily conserved, and therefore, amoebae are not only ecologically significant niches for Legionella spp., but also useful cellular models for eukaryotic phagocytes. In particular, Acanthamoeba castellanii and Dictyostelium discoideum emerged over the last years as versatile and powerful models. Using genetic, biochemical and cell biological approaches, molecular interactions between amoebae and Legionella pneumophila have recently been investigated in detail with a focus on the role of phosphoinositide lipids, small and large GTPases, autophagy components and the retromer complex, as well as on bacterial effectors targeting these host factors.


Asunto(s)
Acanthamoeba/microbiología , Dictyostelium/microbiología , Modelos Animales de Enfermedad , Legionella/metabolismo , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/veterinaria , Acanthamoeba castellanii/microbiología , Amoeba/microbiología , Animales , Autofagia , Proteínas Bacterianas/metabolismo , Evaluación Preclínica de Medicamentos , Evolución Molecular , GTP Fosfohidrolasas , Interacciones Huésped-Patógeno/fisiología , Legionella/patogenicidad , Legionella pneumophila/metabolismo , Macrófagos/microbiología , Fosfatidilinositoles/metabolismo , Proteómica , Sistemas de Secreción Tipo IV/metabolismo , Vacuolas/metabolismo , Vacuolas/microbiología
16.
Int J Med Microbiol ; 308(1): 49-57, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28865995

RESUMEN

The environmental bacterium Legionella pneumophila replicates in free-living amoeba as well as in alveolar macrophages upon inhalation of bacteria-laden aerosols. Resistance of the opportunistic pathogen to macrophages is a prerequisite to cause a severe pneumonia called Legionnaires' disease. L. pneumophila grows intracellularly in a unique, ER-associated compartment, the Legionella-containing vacuole (LCV). The bacterial Icm/Dot type IV secretion system represents an essential virulence factor, which translocates approximately 300 "effector proteins" into protozoan or mammalian host cells. Some of these effectors contribute to the formation of the LCV by targeting conserved host factors implicated in membrane dynamics, such as phosphoinositide lipids and small GTPases. Here we review recent findings on the role of phosphoinositides, small and large GTPases as well as ER dynamics for pathogen vacuole formation and intracellular replication of L. pneumophila.


Asunto(s)
Retículo Endoplásmico/metabolismo , GTP Fosfohidrolasas/metabolismo , Legionella/fisiología , Fosfatidilinositoles/metabolismo , Vacuolas/microbiología , Amoeba/microbiología , Animales , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Legionella/metabolismo , Macrófagos/microbiología , Sistemas de Secreción Tipo IV/metabolismo , Vacuolas/metabolismo
17.
Cell Host Microbe ; 22(3): 302-316.e7, 2017 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-28867389

RESUMEN

The intracellular bacteria Legionella pneumophila encodes a type IV secretion system (T4SS) that injects effector proteins into macrophages in order to establish and replicate within the Legionella-containing vacuole (LCV). Once generated, the LCV interacts with mitochondria through unclear mechanisms. We show that Legionella uses both T4SS-independent and T4SS-dependent mechanisms to respectively interact with mitochondria and induce mitochondrial fragmentation that ultimately alters mitochondrial metabolism. The T4SS effector MitF, a Ran GTPase activator, is required for fission of the mitochondrial network. These effects of MitF occur through accumulation of mitochondrial DNM1L, a GTPase critical for fission. Furthermore mitochondrial respiration is abruptly halted in a T4SS-dependent manner, while T4SS-independent upregulation of cellular glycolysis remains elevated. Collectively, these alterations in mitochondrial dynamics promote a Warburg-like phenotype in macrophages that favors bacterial replication. Hence the rewiring of cellular bioenergetics to create a replication permissive niche in host cells is a virulence strategy of L. pneumophila.


Asunto(s)
Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/metabolismo , Macrófagos/metabolismo , Dinámicas Mitocondriales , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células Cultivadas , Dinaminas , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Legionella pneumophila/genética , Enfermedad de los Legionarios/microbiología , Enfermedad de los Legionarios/fisiopatología , Macrófagos/microbiología , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Células RAW 264.7 , Sistemas de Secreción Tipo IV/genética , Sistemas de Secreción Tipo IV/metabolismo
18.
EMBO Rep ; 18(10): 1817-1836, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28835546

RESUMEN

The pathogenic bacterium Legionella pneumophila replicates in host cells within a distinct ER-associated compartment termed the Legionella-containing vacuole (LCV). How the dynamic ER network contributes to pathogen proliferation within the nascent LCV remains elusive. A proteomic analysis of purified LCVs identified the ER tubule-resident large GTPase atlastin3 (Atl3, yeast Sey1p) and the reticulon protein Rtn4 as conserved LCV host components. Here, we report that Sey1/Atl3 and Rtn4 localize to early LCVs and are critical for pathogen vacuole formation. Sey1 overproduction promotes intracellular growth of L. pneumophila, whereas a catalytically inactive, dominant-negative GTPase mutant protein, or Atl3 depletion, restricts pathogen replication and impairs LCV maturation. Sey1 is not required for initial recruitment of ER to PtdIns(4)P-positive LCVs but for subsequent pathogen vacuole expansion. GTP (but not GDP) catalyzes the Sey1-dependent aggregation of purified, ER-positive LCVs in vitro Thus, Sey1/Atl3-dependent ER remodeling contributes to LCV maturation and intracellular replication of L. pneumophila.


Asunto(s)
Retículo Endoplásmico/fisiología , Proteínas de Unión al GTP/metabolismo , Legionella pneumophila/crecimiento & desarrollo , Proteínas de la Membrana/metabolismo , Vacuolas/metabolismo , Vacuolas/microbiología , Células A549 , Dictyostelium/microbiología , Retículo Endoplásmico/microbiología , Proteínas de Unión al GTP/genética , Humanos , Legionella pneumophila/patogenicidad , Macrófagos/microbiología , Proteínas de la Membrana/genética , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Proteómica , Sistemas de Secreción Tipo IV
19.
Mol Cell Proteomics ; 16(4): 622-641, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28183814

RESUMEN

Legionella pneumophila is an opportunistic bacterial pathogen that causes a severe lung infection termed "Legionnaires' disease." The pathogen replicates in environmental protozoa as well as in macrophages within a unique membrane-bound compartment, the Legionella-containing-vacuole (LCV). LCV formation requires the bacterial Icm/Dot type IV secretion system, which translocates ca. 300 "effector proteins" into host cells, where they target distinct host factors. The L. pneumophila "pentuple" mutant (Δpentuple) lacks 5 gene clusters (31% of the effector proteins) and replicates in macrophages but not in Dictyostelium discoideum amoeba. To elucidate the host factors defining a replication-permissive compartment, we compare here the proteomes of intact LCVs isolated from D. discoideum or macrophages infected with Δpentuple or the parental strain Lp02. This analysis revealed that the majority of host proteins are shared in D. discoideum or macrophage LCVs containing the mutant or the parental strain, respectively, whereas some proteins preferentially localize to distinct LCVs. The small GTPase Rap1 was identified on D. discoideum LCVs containing strain Lp02 but not the Δpentuple mutant and on macrophage LCVs containing either strain. The localization pattern of active Rap1 on D. discoideum or macrophage LCVs was confirmed by fluorescence microscopy and imaging flow cytometry, and the depletion of Rap1 by RNA interference significantly reduced the intracellular growth of L. pneumophila Thus, comparative proteomics identified Rap1 as a novel LCV host component implicated in intracellular replication of L. pneumophila.


Asunto(s)
Proteínas Bacterianas/genética , Dictyostelium/metabolismo , Legionella pneumophila/fisiología , Macrófagos/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteómica/métodos , Vacuolas/microbiología , Proteínas de Unión al GTP rap1/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Cromatografía Liquida , Replicación del ADN , Dictyostelium/microbiología , Eliminación de Gen , Legionella pneumophila/genética , Enfermedad de los Legionarios/microbiología , Macrófagos/microbiología , Ratones , Proteínas Protozoarias/metabolismo , Células RAW 264.7 , Espectrometría de Masas en Tándem , Vacuolas/metabolismo
20.
Mol Microbiol ; 100(2): 229-46, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26691313

RESUMEN

Amino acids represent the prime carbon and energy source for Legionella pneumophila, a facultative intracellular pathogen, which can cause a life-threatening pneumonia termed Legionnaires' disease. Genome, transcriptome and proteome studies indicate that L. pneumophila also utilizes carbon substrates other than amino acids. We show here that glycerol promotes intracellular replication of L. pneumophila in amoeba or macrophages (but not extracellular growth) dependent on glycerol-3-phosphate dehydrogenase, GlpD. An L. pneumophila mutant strain lacking glpD was outcompeted by wild-type bacteria upon co-infection of amoeba, indicating an important role of glycerol during infection. Isotopologue profiling studies using (13) C-labelled substrates were performed in a novel minimal defined medium, MDM, comprising essential amino acids, proline and phenylalanine. In MDM, L. pneumophila utilized (13) C-labelled glycerol or glucose predominantly for gluconeogenesis and the pentose phosphate pathway, while the amino acid serine was used for energy generation via the citrate cycle. Similar results were obtained for L. pneumophila growing intracellularly in amoeba fed with (13) C-labelled glycerol, glucose or serine. Collectively, these results reveal a bipartite metabolism of L. pneumophila, where glycerol and carbohydrates like glucose are mainly fed into anabolic processes, while serine serves as major energy supply.


Asunto(s)
Glicerol/metabolismo , Legionella pneumophila/metabolismo , Aminoácidos/metabolismo , Amoeba/microbiología , Animales , Proteínas Bacterianas/metabolismo , Isótopos de Carbono/metabolismo , Gluconeogénesis/fisiología , Glucosa/metabolismo , Glicerolfosfato Deshidrogenasa/metabolismo , Macrófagos/microbiología , Redes y Vías Metabólicas , Ratones , Células RAW 264.7 , Serina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA