Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Infect Immun ; 86(9)2018 09.
Article in English | MEDLINE | ID: mdl-29986895

ABSTRACT

Within tuberculous granulomas, a subpopulation of Mycobacterium tuberculosis resides inside foamy macrophages (FM) that contain abundant cytoplasmic lipid bodies (LB) filled with triacylglycerol (TAG). Upon fusion of LB with M. tuberculosis-containing phagosomes, TAG is hydrolyzed and reprocessed by the bacteria into their own lipids, which accumulate as intracytosolic lipid inclusions (ILI). This phenomenon is driven by many mycobacterial lipases, among which LipY participates in the hydrolysis of host and bacterial TAG. However, the functional contribution of LipY's PE domain to TAG hydrolysis remains unclear. Here, enzymatic studies were performed to compare the lipolytic activities of recombinant LipY and its truncated variant lacking the N-terminal PE domain, LipY(ΔPE). Complementarily, an FM model was used where bone marrow-derived mouse macrophages were infected with M. bovis BCG strains either overexpressing LipY or LipY(ΔPE) or carrying a lipY deletion mutation prior to being exposed to TAG-rich very-low-density lipoprotein (VLDL). Results indicate that truncation of the PE domain correlates with increased TAG hydrolase activity. Quantitative electron microscopy analyses showed that (i) in the presence of lipase inhibitors, large ILI (ILI+3) were not formed because of an absence of LB due to inhibition of VLDL-TAG hydrolysis or inhibition of LB-neutral lipid hydrolysis by mycobacterial lipases, (ii) ILI+3 profiles in the strain overexpressing LipY(ΔPE) were reduced, and (iii) the number of ILI+3 profiles in the ΔlipY mutant was reduced by 50%. Overall, these results delineate the role of LipY and its PE domain in host and mycobacterial lipid consumption and show that additional mycobacterial lipases take part in these processes.


Subject(s)
Bacterial Proteins/chemistry , Carboxylic Ester Hydrolases/chemistry , Lipid Metabolism , Macrophages/microbiology , Macrophages/physiology , Triglycerides/metabolism , Virulence Factors/chemistry , Animals , Bacterial Proteins/genetics , Carboxylic Ester Hydrolases/genetics , Catalytic Domain , Cells, Cultured , Female , Lipase/metabolism , Lipoproteins, VLDL/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron , Mycobacterium bovis , Protein Structure, Tertiary , Tuberculosis/microbiology , Virulence Factors/genetics
2.
Open Biol ; 6(11)2016 11.
Article in English | MEDLINE | ID: mdl-27906132

ABSTRACT

Mycobacterium abscessus is a pathogenic, rapidly growing mycobacterium responsible for pulmonary and cutaneous infections in immunocompetent patients and in patients with Mendelian disorders, such as cystic fibrosis (CF). Mycobacterium abscessus is known to transition from a smooth (S) morphotype with cell surface-associated glycopeptidolipids (GPL) to a rough (R) morphotype lacking GPL. Herein, we show that M. abscessus S and R variants are able to grow inside macrophages and are present in morphologically distinct phagosomes. The S forms are found mostly as single bacteria within phagosomes characterized by a tightly apposed phagosomal membrane and the presence of an electron translucent zone (ETZ) surrounding the bacilli. By contrast, infection with the R form leads to phagosomes often containing more than two bacilli, surrounded by a loose phagosomal membrane and lacking the ETZ. In contrast to the R variant, the S variant is capable of restricting intraphagosomal acidification and induces less apoptosis and autophagy. Importantly, the membrane of phagosomes enclosing the S forms showed signs of alteration, such as breaks or partial degradation. Although not frequently encountered, these events suggest that the S form is capable of provoking phagosome-cytosol communication. In conclusion, M. abscessus S exhibits traits inside macrophages that are reminiscent of slow-growing mycobacterial species.


Subject(s)
Macrophages/microbiology , Mycobacterium chelonae/growth & development , Cells, Cultured , Fluorescence Resonance Energy Transfer , Humans , Mycobacterium Infections, Nontuberculous/microbiology , Phagosomes/microbiology
3.
Article in English | MEDLINE | ID: mdl-27774438

ABSTRACT

Despite a slight decline since 2014, tuberculosis (TB) remains the major deadly infectious disease worldwide with about 1.5 million deaths each year and with about one-third of the population being latently infected with Mycobacterium tuberculosis, the etiologic agent of TB. During primo-infection, the recruitment of immune cells leads to the formation of highly organized granulomas. Among the different cells, one outstanding subpopulation is the foamy macrophage (FM), characterized by the abundance of triacylglycerol-rich lipid bodies (LB). M. tuberculosis can reside in FM, where it acquires, from host LB, the neutral lipids which are subsequently processed and stored by the bacilli in the form of intracytosolic lipid inclusions (ILI). Although host LB can be viewed as a reservoir of nutrients for the pathogen during latency, the molecular mechanisms whereby intraphagosomal mycobacteria interact with LB and assimilate the LB-derived lipids are only beginning to be understood. Past studies have emphasized that these physiological processes are critical to the M. tuberculosis infectious-life cycle, for propagation of the infection, establishment of the dormancy state and reactivation of the disease. In recent years, several animal and cellular models have been developed with the aim of dissecting these complex processes and of determining the nature and contribution of their key players. Herein, we review some of the in vitro and in vivo models which allowed to gain significant insight into lipid accumulation and consumption in M. tuberculosis, two important events that are directly linked to pathogenicity, granuloma formation/maintenance and survival of the tubercle bacillus under non-replicative conditions. We also discuss the advantages and limitations of each model, hoping that this will serve as a guide for future investigations dedicated to persistence and innovative therapeutic approaches against TB.


Subject(s)
Host-Pathogen Interactions , Lipid Metabolism , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/growth & development , Animals , Humans , Models, Theoretical
4.
Mol Microbiol ; 102(4): 611-627, 2016 11.
Article in English | MEDLINE | ID: mdl-27513974

ABSTRACT

Slow growing pathogenic mycobacteria utilize host-derived lipids and accumulate large amounts of triacylglycerol (TAG) in the form of intracytoplasmic lipid inclusions (ILI), serving as a source of carbon and energy during prolonged infection. Mycobacterium abscessus is an emerging and rapidly growing species capable to induce severe and chronic pulmonary infections. However, whether M. abscessus, like Mycobacterium tuberculosis, possesses the machinery to acquire and store host lipids, remains unaddressed. Herein, we aimed at deciphering the contribution of the seven putative M. abscessus TAG synthases (Tgs) in TAG synthesis/accumulation thanks to a combination of genetic and biochemical techniques and a well-defined foamy macrophage (FM) model along with electron microscopy. Targeted gene deletion and functional complementation studies identified the MAB_3551c product, Tgs1, as the major Tgs involved in TAG production. Tgs1 exhibits a preference for long acyl-CoA substrates and site-directed mutagenesis demonstrated that His144 and Gln145 are essential for enzymatic activity. Importantly, in the lipid-rich intracellular context of FM, M. abscessus formed large ILI in a Tgs1-dependent manner. This supports the ability of M. abscessus to assimilate host lipids and the crucial role of Tgs1 in intramycobacterial TAG production, which may represent important mechanisms for long-term storage of a rich energy supply.


Subject(s)
Fatty Acid Synthases/genetics , Nontuberculous Mycobacteria/genetics , Triglycerides/biosynthesis , Amino Acid Sequence , Fatty Acid Synthases/metabolism , Lipid Metabolism/genetics , Mutagenesis, Site-Directed , Nontuberculous Mycobacteria/enzymology , Nontuberculous Mycobacteria/metabolism , Sequence Homology, Amino Acid , Triglycerides/metabolism
5.
Mol Microbiol ; 99(5): 866-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26585558

ABSTRACT

In mycobacteria, MmpL proteins represent key components that participate in the biosynthesis of the complex cell envelope. Whole genome analysis of a spontaneous rough morphotype variant of Mycobacterium abscessus subsp. bolletii identified a conserved tyrosine that is crucial for the function of MmpL family proteins. Isogenic smooth (S) and rough (R) variants differed by a single mutation linked to a Y842H substitution in MmpL4a. This mutation caused a deficiency in glycopeptidolipid production/transport in the R variant and a gain in the capacity to produce cords in vitro. In zebrafish, increased virulence of the M. bolletii R variant over the parental S strain was found, involving massive production of serpentine cords, abscess formation and rapid larval death. Importantly, this finding allowed us to demonstrate an essential role of Tyr842 in several different MmpL proteins, including Mycobacterium tuberculosis MmpL3. Structural homology models of MmpL4a and MmpL3 identified two additional critical residues located in the transmembrane regions TM10 and TM4 that are facing each other. We propose that these central residues are part of the proton-motive force that supplies the energy for substrate transport. Hence, we provide important insights into mechanistic/structural aspects of MmpL proteins as lipid transporters and virulence determinants in mycobacteria.


Subject(s)
Membrane Transport Proteins/metabolism , Mycobacterium/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biological Transport , Cell Wall/metabolism , Conserved Sequence , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mycobacterium/genetics , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Proton-Motive Force , Virulence , Virulence Factors/metabolism , Zebrafish
6.
Virulence ; 7(1): 33-44, 2016.
Article in English | MEDLINE | ID: mdl-26606688

ABSTRACT

Brucella is a Gram-negative bacterium responsible for brucellosis, a worldwide re-emerging zoonosis. Brucella has been shown to infect and replicate within Granulocyte macrophage colony-stimulating factor (GMCSF) in vitro grown bone marrow-derived dendritic cells (BMDC). In this cell model, Brucella can efficiently control BMDC maturation. However, it has been shown that Brucella infection in vivo induces spleen dendritic cells (DC) migration and maturation. As DCs form a complex network composed by several subpopulations, differences observed may be due to different interactions between Brucella and DC subsets. Here, we compare Brucella interaction with several in vitro BMDC models. The present study shows that Brucella is capable of replicating in all the BMDC models tested with a high infection rate at early time points in GMCSF-IL15 DCs and Flt3l DCs. GMCSF-IL15 DCs and Flt3l DCs are more activated than the other studied DC models and consequently intracellular bacteria are not efficiently targeted to the ER replicative niche. Interestingly, GMCSF-DC and GMCSF-Flt3l DC response to infection is comparable. However, the key difference between these 2 models concerns IL10 secretion by GMCSF DCs observed at 48 h post-infection. IL10 secretion can explain the weak secretion of IL12p70 and TNFα in the GMCSF-DC model and the low level of maturation observed when compared to GMCSF-IL15 DCs and Flt3l DCs. These models provide good tools to understand how Brucella induce DC maturation in vivo and may lead to new therapeutic design using DCs as cellular vaccines capable of enhancing immune response against pathogens.


Subject(s)
Brucella/pathogenicity , Brucellosis/microbiology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Animals , Brucella/genetics , Brucella/growth & development , Brucella/immunology , Brucellosis/immunology , Brucellosis/pathology , Brucellosis/prevention & control , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cytokines/immunology , Dendritic Cells/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Host-Pathogen Interactions , Interleukin-10/immunology , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Transcriptome , Tumor Necrosis Factor-alpha/immunology
7.
J Control Release ; 211: 94-104, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26055640

ABSTRACT

The appearance of drug-resistant strains of Mycobacterium tuberculosis (Mtb) poses a great challenge to the development of novel treatment programmes to combat tuberculosis. Since innovative nanotechnologies might alleviate the limitations of current therapies, we have designed a new nanoformulation for use as an anti-TB drug delivery system. It consists of incorporating mycobacterial cell wall mycolic acids (MA) as targeting ligands into a drug-encapsulating Poly dl-lactic-co-glycolic acid polymer (PLGA), via a double emulsion solvent evaporation technique. Bone marrow-derived mouse macrophages, either uninfected or infected with different mycobacterial strains (Mycobacterium avium, Mycobacterium bovis BCG or Mtb), were exposed to encapsulated isoniazid-PLGA nanoparticles (NPs) using MA as a targeting ligand. The fate of the NPs was monitored by electron microscopy. Our study showed that i) the inclusion of MA in the nanoformulations resulted in their expression on the outer surface and a significant increase in phagocytic uptake of the NPs; ii) nanoparticle-containing phagosomes were rapidly processed into phagolysosomes, whether MA had been included or not; and iii) nanoparticle-containing phagolysosomes did not fuse with non-matured mycobacterium-containing phagosomes, but fusion events with mycobacterium-containing phagolysosomes were clearly observed.


Subject(s)
Antitubercular Agents/administration & dosage , Drug Delivery Systems/methods , Mycolic Acids/administration & dosage , Nanoparticles/administration & dosage , Tuberculosis , Animals , Antitubercular Agents/metabolism , Female , Humans , Ligands , Mice , Mice, Inbred C57BL , Mycobacterium bovis/drug effects , Mycobacterium bovis/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Mycolic Acids/metabolism , Nanoparticles/metabolism , Tuberculosis/drug therapy , Tuberculosis/metabolism
8.
PLoS Pathog ; 10(2): e1003928, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586151

ABSTRACT

Mycobacterium tuberculosis is an intracellular pathogen. Within macrophages, M. tuberculosis thrives in a specialized membrane-bound vacuole, the phagosome, whose pH is slightly acidic, and where access to nutrients is limited. Understanding how the bacillus extracts and incorporates nutrients from its host may help develop novel strategies to combat tuberculosis. Here we show that M. tuberculosis employs the asparagine transporter AnsP2 and the secreted asparaginase AnsA to assimilate nitrogen and resist acid stress through asparagine hydrolysis and ammonia release. While the role of AnsP2 is partially spared by yet to be identified transporter(s), that of AnsA is crucial in both phagosome acidification arrest and intracellular replication, as an M. tuberculosis mutant lacking this asparaginase is ultimately attenuated in macrophages and in mice. Our study provides yet another example of the intimate link between physiology and virulence in the tubercle bacillus, and identifies a novel pathway to be targeted for therapeutic purposes.


Subject(s)
Asparagine/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/metabolism , Nitrogen/metabolism , Phagosomes/metabolism , Stress, Physiological , Tuberculosis/metabolism , Animals , Chromatography, Liquid , Disease Models, Animal , Female , Flow Cytometry , Gene Knockout Techniques , Immunoblotting , Mass Spectrometry , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Phagosomes/microbiology
9.
Infect Immun ; 82(2): 476-90, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478064

ABSTRACT

During the dormant phase of tuberculosis, Mycobacterium tuberculosis persists in lung granulomas by residing in foamy macrophages (FM) that contain abundant lipid bodies (LB) in their cytoplasm, allowing bacilli to accumulate lipids as intracytoplasmic lipid inclusions (ILI). An experimental model of FM is presented where bone marrow-derived mouse macrophages are infected with M. avium and exposed to very-low-density lipoprotein (VLDL) as a lipid source. Quantitative analysis of detailed electron microscope observations showed the following results. (i) Macrophages became foamy, and mycobacteria formed ILI, for which host triacylglycerides, rather than cholesterol, was essential. (ii) Lipid transfer occurred via mycobacterium-induced fusion between LB and phagosomes. (iii) Mycobacteria showed a thinned cell wall and became elongated but did not divide. (iv) Upon removal of VLDL, LB and ILI declined within hours, and simultaneous resumption of mycobacterial division restored the number of mycobacteria to the same level as that found in untreated control macrophages. This showed that the presence of ILI resulted in a reversible block of division without causing a change in the mycobacterial replication rate. Fluctuation between ILI either partially or fully extending throughout the mycobacterial cytoplasm was suggestive of bacterial cell cycle events. We propose that VLDL-driven FM constitute a well-defined cellular system in which to study changed metabolic states of intracellular mycobacteria that may relate to persistence and reactivation of tuberculosis.


Subject(s)
Lipid Metabolism , Lipoproteins, VLDL/metabolism , Macrophages/microbiology , Mycobacterium avium/growth & development , Mycobacterium avium/metabolism , Animals , Cell Division , Cells, Cultured , Female , Inclusion Bodies/microbiology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mycobacterium avium/ultrastructure
10.
PLoS Negl Trop Dis ; 7(11): e2502, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24244764

ABSTRACT

BACKGROUND: Mycobacterium ulcerans, a slow-growing environmental bacterium, is the etiologic agent of Buruli ulcer, a necrotic skin disease. Skin lesions are caused by mycolactone, the main virulence factor of M. ulcerans, with dermonecrotic (destruction of the skin and soft tissues) and immunosuppressive activities. This toxin is secreted in vesicles that enhance its biological activities. Nowadays, it is well established that the main reservoir of the bacilli is localized in the aquatic environment where the bacillus may be able to colonize different niches. Here we report that plant polysaccharides stimulate M. ulcerans growth and are implicated in toxin synthesis regulation. METHODOLOGY/PRINCIPAL FINDINGS: In this study, by selecting various algal components, we have identified plant-specific carbohydrates, particularly glucose polymers, capable of stimulating M. ulcerans growth in vitro. Furthermore, we underscored for the first time culture conditions under which the polyketide toxin mycolactone, the sole virulence factor of M. ulcerans identified to date, is down-regulated. Using a quantitative proteomic approach and analyzing transcript levels by RT-qPCR, we demonstrated that its regulation is not at the transcriptional or translational levels but must involve another type of regulation. M. ulcerans produces membrane vesicles, as other mycobacterial species, in which are the mycolactone is concentrated. By transmission electron microscopy, we observed that the production of vesicles is independent from the toxin production. Concomitant with this observed decrease in mycolactone production, the production of mycobacterial siderophores known as mycobactins was enhanced. CONCLUSIONS/SIGNIFICANCE: This work is the first step in the identification of the mechanisms involved in mycolactone regulation and paves the way for the discovery of putative new drug targets in the future.


Subject(s)
Macrolides/metabolism , Mycobacterium ulcerans/metabolism , Proteomics , Reverse Transcriptase Polymerase Chain Reaction
11.
J Infect Dis ; 207(7): 1075-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303808

ABSTRACT

Brucellae replicate in a vacuole derived from the endoplasmic reticulum (ER) in epithelial cells, macrophages, and dendritic cells. In animals, trophoblasts are also key cellular targets where brucellae efficiently replicate in association with the ER. Therefore, we investigated the ability of Brucella spp. to infect human trophoblasts using both immortalized and primary trophoblasts. Brucella extensively proliferated within different subpopulations of trophoblasts, suggesting that they constitute an important niche in cases where the fetal-maternal barrier is breached. In extravillous trophoblasts (EVTs), B. abortus and B. suis replicated within single-membrane acidic lysosomal membrane-associated protein 1-positive inclusions, whereas B. melitensis replicated in the ER-derived compartment. Furthermore, B. melitensis but not B. abortus nor B. suis interfered with the invasive capacity of EVT-like cells in vitro. Because EVTs are essential for implantation during early stages of pregnancy, the nature of the replication niche may have a central role during Brucella-associated abortion in infected women.


Subject(s)
Brucella abortus/growth & development , Brucella suis/growth & development , Trophoblasts/microbiology , Autophagy , Bacterial Load , Brucella abortus/metabolism , Brucella abortus/pathogenicity , Brucella melitensis/growth & development , Brucella melitensis/metabolism , Brucella melitensis/pathogenicity , Brucella suis/metabolism , Brucella suis/pathogenicity , Brucellosis/microbiology , Brucellosis/pathology , Calnexin/metabolism , Cells, Cultured , Female , Humans , Lysosomal Membrane Proteins/metabolism , Microbial Viability , Microscopy, Fluorescence , Placenta/metabolism , Placenta/microbiology , Placenta/pathology , Pregnancy , Tetraspanin 30/metabolism , Trophoblasts/metabolism , Trophoblasts/pathology
12.
J Biol Chem ; 287(31): 26187-99, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22621931

ABSTRACT

Pathogenic mycobacteria survive within macrophages by residing in phagosomes, which they prevent from maturing and fusing with lysosomes. Although several bacterial components were seen to modulate phagosome processing, the molecular regulatory mechanisms taking part in this process remain elusive. We investigated whether the phagosome maturation block (PMB) could be modulated by signaling through Ser/Thr phosphorylation. Here, we demonstrated that mycolic acid cyclopropane synthase PcaA, but not MmaA2, was phosphorylated by mycobacterial Ser/Thr kinases at Thr-168 and Thr-183 both in vitro and in mycobacteria. Phosphorylation of PcaA was associated with a significant decrease in the methyltransferase activity, in agreement with the strategic structural localization of these two phosphoacceptors. Using a BCG ΔpcaA mutant, we showed that PcaA was required for intracellular survival and prevention of phagosome maturation in human monocyte-derived macrophages. The physiological relevance of PcaA phosphorylation was further assessed by generating PcaA phosphoablative (T168A/T183A) or phosphomimetic (T168D/T183D) mutants. In contrast to the wild-type and phosphoablative pcaA alleles, introduction of the phosphomimetic pcaA allele in the ΔpcaA mutant failed to restore the parental mycolic acid profile and cording morphotype. Importantly, the PcaA phosphomimetic strain, as the ΔpcaA mutant, exhibited reduced survival in human macrophages and was unable to prevent phagosome maturation. Our results add new insight into the importance of mycolic acid cyclopropane rings in the PMB and provide the first evidence of a Ser/Thr kinase-dependent mechanism for modulating mycolic acid composition and PMB.


Subject(s)
Bacterial Proteins/metabolism , Methyltransferases/metabolism , Mycobacterium tuberculosis/enzymology , Mycolic Acids/metabolism , Phagosomes , Amino Acid Sequence , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Conserved Sequence , Cyclopropanes/metabolism , Host-Pathogen Interactions , Humans , Macrophages/microbiology , Methyltransferases/chemistry , Methyltransferases/genetics , Microbial Viability , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mycobacterium bovis/enzymology , Mycobacterium bovis/metabolism , Mycobacterium bovis/physiology , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry
14.
Trends Microbiol ; 20(3): 106-12, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22305804

ABSTRACT

Iron, zinc and copper, among others, are transition metals with multiple biological roles that make them essential elements for life. Beyond the strict requirement of transition metals by the vertebrate immune system for its proper functioning, novel mechanisms involving direct metal intoxication of microorganisms are starting to be unveiled as important components of the immune system, in particular against Mycobacterium tuberculosis. In parallel, metal detoxification systems in bacteria have been recently characterized as crucial microbial virulence determinants. Here, we will focus on these exciting advancements implicating copper- and zinc-mediated microbial poisoning as a novel innate immune mechanism against microbial pathogens, shedding light on an emerging field in the metallobiology of host-pathogen interactions.


Subject(s)
Copper/metabolism , Host-Pathogen Interactions , Iron/metabolism , Mycobacterium tuberculosis/metabolism , Tuberculosis/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Humans , Mycobacterium tuberculosis/genetics , Tuberculosis/metabolism
15.
Cell Host Microbe ; 10(3): 248-59, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21925112

ABSTRACT

Mycobacterium tuberculosis thrives within macrophages by residing in phagosomes and preventing them from maturing and fusing with lysosomes. A parallel transcriptional survey of intracellular mycobacteria and their host macrophages revealed signatures of heavy metal poisoning. In particular, mycobacterial genes encoding heavy metal efflux P-type ATPases CtpC, CtpG, and CtpV, and host cell metallothioneins and zinc exporter ZnT1, were induced during infection. Consistent with this pattern of gene modulation, we observed a burst of free zinc inside macrophages, and intraphagosomal zinc accumulation within a few hours postinfection. Zinc exposure led to rapid CtpC induction, and ctpC deficiency caused zinc retention within the mycobacterial cytoplasm, leading to impaired intracellular growth of the bacilli. Thus, the use of P(1)-type ATPases represents a M. tuberculosis strategy to neutralize the toxic effects of zinc in macrophages. We propose that heavy metal toxicity and its counteraction might represent yet another chapter in the host-microbe arms race.


Subject(s)
Bacterial Proton-Translocating ATPases/metabolism , Macrophages/metabolism , Mycobacterium tuberculosis/enzymology , Tuberculosis/metabolism , Zinc/metabolism , Animals , Bacterial Proton-Translocating ATPases/genetics , Cells, Cultured , Female , Humans , Macrophages/drug effects , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Tuberculosis/microbiology , Zinc/toxicity
16.
PLoS Pathog ; 6(7): e1001002, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20664790

ABSTRACT

Salmonella enterica serovar Typhimurium is a Gram-negative bacterial pathogen causing gastroenteritis in humans and a systemic typhoid-like illness in mice. The capacity of Salmonella to cause diseases relies on the establishment of its intracellular replication niche, a membrane-bound compartment named the Salmonella-containing vacuole (SCV). This requires the translocation of bacterial effector proteins into the host cell by type three secretion systems. Among these effectors, SifA is required for the SCV stability, the formation of Salmonella-induced filaments (SIFs) and plays an important role in the virulence of Salmonella. Here we show that the effector SopD2 is responsible for the SCV instability that triggers the cytoplasmic release of a sifA(-) mutant. Deletion of sopD2 also rescued intra-macrophagic replication and increased virulence of sifA(-) mutants in mice. Membrane tubular structures that extend from the SCV are the hallmark of Salmonella-infected cells. Until now, these unique structures have not been observed in the absence of SifA. The deletion of sopD2 in a sifA(-) mutant strain re-established membrane trafficking from the SCV and led to the formation of new membrane tubular structures, the formation of which is dependent on other Salmonella effector(s). Taken together, our data demonstrate that SopD2 inhibits the vesicular transport and the formation of tubules that extend outward from the SCV and thereby contributes to the sifA(-) associated phenotypes. These results also highlight the antagonistic roles played by SopD2 and SifA in the membrane dynamics of the vacuole, and the complex actions of SopD2, SifA, PipB2 and other unidentified effector(s) in the biogenesis and maintenance of the Salmonella replicative niche.


Subject(s)
Bacterial Proteins/physiology , Salmonella typhimurium/physiology , Vacuoles/microbiology , Animals , Biological Transport , Glycoproteins/physiology , Host-Pathogen Interactions , Mice , Microtubules/metabolism , Salmonella Infections , Salmonella typhimurium/pathogenicity , Vacuoles/metabolism , Virulence
17.
PLoS Pathog ; 5(6): e1000487, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19557163

ABSTRACT

The intracellular pathogen Brucella abortus survives and replicates inside host cells within an endoplasmic reticulum (ER)-derived replicative organelle named the "Brucella-containing vacuole" (BCV). Here, we developed a subcellular fractionation method to isolate BCVs and characterize for the first time the protein composition of its replicative niche. After identification of BCV membrane proteins by 2 dimensional (2D) gel electrophoresis and mass spectrometry, we focused on two eukaryotic proteins: the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and the small GTPase Rab 2 recruited to the vacuolar membrane of Brucella. These proteins were previously described to localize on vesicular and tubular clusters (VTC) and to regulate the VTC membrane traffic between the endoplasmic reticulum (ER) and the Golgi. Inhibition of either GAPDH or Rab 2 expression by small interfering RNA strongly inhibited B. abortus replication. Consistent with this result, inhibition of other partners of GAPDH and Rab 2, such as COPI and PKC iota, reduced B. abortus replication. Furthermore, blockage of Rab 2 GTPase in a GDP-locked form also inhibited B. abortus replication. Bacteria did not fuse with the ER and instead remained in lysosomal-associated membrane vacuoles. These results reveal an essential role for GAPDH and the small GTPase Rab 2 in B. abortus virulence within host cells.


Subject(s)
Brucella abortus/cytology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , rab2 GTP-Binding Protein/metabolism , Animals , Brucella abortus/growth & development , Cell Line , Cell Membrane/chemistry , Cell Membrane/microbiology , Cell Survival , Endoplasmic Reticulum/microbiology , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/chemistry , Host-Pathogen Interactions/physiology , Immunohistochemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Secretory Pathway/physiology , Vacuoles/chemistry , Vacuoles/enzymology , Vacuoles/microbiology , rab2 GTP-Binding Protein/chemistry
18.
Cell Microbiol ; 11(8): 1190-207, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19388907

ABSTRACT

Pathogenic mycobacteria survive in macrophages of the host organism by residing in phagosomes which they prevent from undergoing maturation and fusion with lysosomes. Several molecular mechanisms have been associated with the phagosome maturation block. Here we show for Mycobacterium avium in mouse bone marrow-derived macrophages that the maturation block required an all-around close apposition between the mycobacterial surface and the phagosome membrane. When small (0.1 microm) latex beads were covalently attached to the mycobacterial surface to act as a spacer that interfered with a close apposition, phagosomes rapidly acquired lysosomal characteristics as indicators for maturation and fusion with lysosomes. As a result, several mycobacteria were delivered into single phagolysosomes. Detailed electron-microscope observations of phagosome morphology over a 7-day post-infection period showed a linear correlation between bead attachment and phagosome-lysosome fusion. After about 3 days post infection, conditions inside phagolysosomes caused a gradual release of beads. This allowed mycobacteria to re-establish a close apposition with the surrounding membrane and sequester themselves into individual, non-maturing phagosomes which had lost lysosomal characteristics. By rescuing themselves from phagolysosomes, mycobacteria remained fully viable and able to multiply at the normal rate. In order to unify the present observations and previously reported mechanisms for the maturation block, we discuss evidence that they may act synergistically to interfere with 'Phagosome Membrane Economics' by causing relative changes in incoming and outgoing endocytic membrane fluxes.


Subject(s)
Intracellular Membranes/metabolism , Mycobacterium avium , Phagosomes/physiology , Tuberculosis/metabolism , Animals , Cell Culture Techniques , Female , Host-Pathogen Interactions , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Microspheres , Mycobacterium avium/metabolism , Mycobacterium avium/pathogenicity , Phagocytosis , Phagosomes/ultrastructure , Tuberculosis/immunology , Tuberculosis/microbiology , Virulence
19.
Immunobiology ; 214(7): 526-42, 2009.
Article in English | MEDLINE | ID: mdl-19261352

ABSTRACT

A major virulence factor of pathogenic mycobacteria is their ability to parasitize the host's scavenger cells and more particularly macrophages. The present overview discusses the known cellular and molecular mechanisms of intracellular survival of Mtb and other pathogenic mycobacteria within different intracellular niches, i.e. the macrophage in which they replicate and the granuloma in which they persist in a non-replicating state. After phagocytic uptake by macrophages, mycobacteria reside in phagosomes which they prevent from maturing and, as a result, from fusing with acidic and hydrolase-rich lysosomes. Two major points are highlighted: (i) the requirement for a close apposition between the phagosome membrane and the mycobacterial surface all around, and (ii) the ability for mycobacteria targeted to phagolysosomes to avoid degradation and to be rescued from this cytolytic environment to again reside in non-maturing phagosomes with a closely apposed membrane in which they can replicate. Concerning Mtb in granulomatous lesions, this review discusses the occurence of mycobacteria in lipid-rich foamy macrophages in which they persist in a non-replicating state. This overview highlights the major contribution of host cholesterol and/or fatty acids (triacylglycerol) in both prevention of phagosome maturation and persistence in granulomatous lesions.


Subject(s)
Macrophages/immunology , Mycobacterium Infections/immunology , Mycobacterium Infections/microbiology , Mycobacterium/immunology , Animals , Cholesterol/metabolism , Granuloma , Host-Pathogen Interactions , Humans , Mycobacterium/growth & development , Mycobacterium/pathogenicity , Mycobacterium Infections/pathology , Mycobacterium Infections/physiopathology , Phagosomes/immunology , Virulence
20.
PLoS Pathog ; 4(11): e1000204, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19002241

ABSTRACT

Tuberculosis (TB) is characterized by a tight interplay between Mycobacterium tuberculosis and host cells within granulomas. These cellular aggregates restrict bacterial spreading, but do not kill all the bacilli, which can persist for years. In-depth investigation of M. tuberculosis interactions with granuloma-specific cell populations are needed to gain insight into mycobacterial persistence, and to better understand the physiopathology of the disease. We have analyzed the formation of foamy macrophages (FMs), a granuloma-specific cell population characterized by its high lipid content, and studied their interaction with the tubercle bacillus. Within our in vitro human granuloma model, M. tuberculosis long chain fatty acids, namely oxygenated mycolic acids (MA), triggered the differentiation of human monocyte-derived macrophages into FMs. In these cells, mycobacteria no longer replicated and switched to a dormant non-replicative state. Electron microscopy observation of M. tuberculosis-infected FMs showed that the mycobacteria-containing phagosomes migrate towards host cell lipid bodies (LB), a process which culminates with the engulfment of the bacillus into the lipid droplets and with the accumulation of lipids within the microbe. Altogether, our results suggest that oxygenated mycolic acids from M. tuberculosis play a crucial role in the differentiation of macrophages into FMs. These cells might constitute a reservoir used by the tubercle bacillus for long-term persistence within its human host, and could provide a relevant model for the screening of new antimicrobials against non-replicating persistent mycobacteria.


Subject(s)
Cell Differentiation , Foam Cells/microbiology , Granuloma/microbiology , Macrophages/microbiology , Mycolic Acids , Tuberculosis/microbiology , Humans , Lipids , Macrophages/pathology , Macrophages/ultrastructure , Mycobacterium Infections/immunology , Mycobacterium tuberculosis/physiology , Phagocytosis , Tuberculosis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...