Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Infect Immun ; 91(4): e0040522, 2023 04 18.
Article in English | MEDLINE | ID: mdl-36877064

ABSTRACT

Chlamydia trachomatis is an obligate intracellular bacterial pathogen that causes ocular and urogenital infections in humans. The ability of C. trachomatis to grow intracellularly in a pathogen-containing vacuole (known as an inclusion) depends on chlamydial effector proteins transported into the host cell by a type III secretion system. Among these effectors, several inclusion membrane proteins (Incs) insert in the vacuolar membrane. Here, we show that human cell lines infected by a C. trachomatis strain deficient for Inc CT288/CTL0540 (renamed IncM) displayed less multinucleation than when infected by IncM-producing strains (wild type or complemented). This indicated that IncM is involved in the ability of Chlamydia to inhibit host cell cytokinesis. The capacity of IncM to induce multinucleation in infected cells was shown to be conserved among its chlamydial homologues and appeared to require its two larger regions predicted to be exposed to the host cell cytosol. C. trachomatis-infected cells also displayed IncM-dependent defects in centrosome positioning, Golgi distribution around the inclusion, and morphology and stability of the inclusion. The altered morphology of inclusions containing IncM-deficient C. trachomatis was further affected by depolymerization of host cell microtubules. This was not observed after depolymerization of microfilaments, and inclusions containing wild-type C. trachomatis did not alter their morphology upon depolymerization of microtubules. Overall, these findings suggest that IncM may exert its effector function by acting directly or indirectly on host cell microtubules.


Subject(s)
Chlamydia Infections , Cytokinesis , Humans , Cytokinesis/physiology , Chlamydia trachomatis/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , HeLa Cells , Vacuoles/metabolism , Centrosome/metabolism , Chlamydia Infections/microbiology , Host-Pathogen Interactions
3.
Front Cell Infect Microbiol ; 12: 902210, 2022.
Article in English | MEDLINE | ID: mdl-35903198

ABSTRACT

Chlamydia trachomatis is an obligate intracellular bacterium causing ocular and urogenital infections in humans that are a significant burden worldwide. The completion of its characteristic infectious cycle relies on the manipulation of several host cell processes by numerous chlamydial type III secretion effector proteins. We previously identified the C. trachomatis CteG effector and showed it localizes at the host cell plasma membrane at late stages of infection. Here, we showed that, from 48 h post-infection, mammalian cells infected by wild-type C. trachomatis contained more infectious chlamydiae in the culture supernatant than cells infected by a CteG-deficient strain. This phenotype was CteG-dependent as it could be complemented in cells infected by the CteG-deficient strain carrying a plasmid encoding CteG. Furthermore, we detected a CteG-dependent defect on host cell cytotoxicity, indicating that CteG mediates chlamydial lytic exit. Previous studies showed that Pgp4, a global regulator of transcription encoded in the C. trachomatis virulence plasmid, also mediates chlamydial lytic exit. However, by using C. trachomatis strains encoding or lacking Pgp4, we showed that production and localization of CteG are not regulated by Pgp4. A C. trachomatis strain lacking both CteG and Pgp4 was as defective in promoting host cell cytotoxicity as mutant strains lacking only CteG or Pgp4. Furthermore, CteG overproduction in a plasmid suppressed the host cell cytotoxic defect of CteG- and Pgp4-deficient chlamydiae. Overall, we revealed the first chlamydial type III secretion effector involved in host cell lytic exit. Our data indicates that CteG and Pgp4 participate in a single cascade of events, but involving multiple layers of regulation, leading to lysis of host cells and release of the infectious chlamydiae.


Subject(s)
Chlamydia Infections , Chlamydia trachomatis , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chlamydia Infections/microbiology , HeLa Cells , Humans , Mammals/genetics , Plasmids/genetics
4.
Front Cell Infect Microbiol ; 12: 864626, 2022.
Article in English | MEDLINE | ID: mdl-35711665

ABSTRACT

Legionella pneumophila is an accidental human pathogen that causes the potentially fatal Legionnaires' disease, a severe type of pneumonia. The main virulence mechanism of L. pneumophila is a Type 4B Secretion System (T4SS) named Icm/Dot that transports effector proteins into the host cell cytosol. The concerted action of effectors on several host cell processes leads to the formation of an intracellular Legionella-containing vacuole that is replication competent and avoids phagolysosomal degradation. To date over 300 Icm/Dot substrates have been identified. In this study, we searched the genome of a L. pneumophila strain (Pt/VFX2014) responsible for the second largest L. pneumophila outbreak worldwide (in Vila Franca de Xira, Portugal, in 2014) for genes encoding potential novel Icm/Dot substrates. This strain Pt/VFX2014 belongs to serogroup 1 but phylogenetically segregates from all other serogroup 1 strains previously sequenced, displaying a unique mosaic genetic backbone. The ability of the selected putative effectors to be delivered into host cells by the T4SS was confirmed using the TEM-1 ß-lactamase reporter assay. Two previously unknown Icm/Dot effectors were identified, VFX05045 and VFX10045, whose homologs Lpp1450 and Lpp3070 in clinical strain L. pneumophila Paris were also confirmed as T4SS substrates. After delivery into the host cell cytosol, homologs VFX05045/Lpp1450 remained diffused in the cell, similarly to Lpp3070. In contrast, VFX10045 localized to the host cell nucleus. To understand how VFX10045 and Lpp3070 (94% of identity at amino acid level) are directed to distinct sites, we carried out a comprehensive site-directed mutagenesis followed by analyses of the subcellular localization of the mutant proteins. This led to the delineation of region in the C-terminal part (residues 380 to 534) of the 583 amino acid-long VFX10045 as necessary and sufficient for nuclear targeting and highlighted the fundamental function of the VFX10045-specific R440 and I441 residues in this process. These studies revealed a strain-specific nucleotropism for new effector VFX10045/Lpp3070, which anticipates distinct functions between these homologs.


Subject(s)
Legionella pneumophila , Legionella , Legionnaires' Disease , Amino Acids/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Legionella/metabolism , Legionella pneumophila/metabolism
5.
PLoS One ; 17(2): e0264292, 2022.
Article in English | MEDLINE | ID: mdl-35192658

ABSTRACT

Chlamydia trachomatis causes genital and ocular infections in humans. This bacterial pathogen multiplies exclusively within host cells in a characteristic vacuole (inclusion) and delivers proteins such as inclusion membrane proteins (Incs) into the host cell. Here, we identified CT006 as a novel C. trachomatis protein that when expressed ectopically eukaryotic cells can associate with lipid droplets (LDs). A screen using Saccharomyces cerevisiae identified two Incs causing vacuolar protein sorting defects and seven Incs showing tropism for eukaryotic organelles. Ectopic expression in yeast and mammalian cells of genes encoding different fragments of CT006 revealed tropism for the endoplasmic reticulum and LDs. We identified a LD-targeting region within the first 88 amino acid residues of CT006, and positively charged residues important for this targeting. Comparing with the parental wild-type strain, cells infected by a newly generated C. trachomatis strain overproducing CT006 with a double hemagglutinin tag showed a slight increase in the area occupied by LDs within the inclusion region. However, we could not correlate this effect with the LD-targeting regions within CT006. We further showed that both the amino and carboxy-terminal regions of CT006, flanking the Inc-characteristic bilobed hydrophobic domain, are exposed to the host cell cytosol during C. trachomatis infection, supporting their availability to interact with host cell targets. Altogether, our data suggest that CT006 might participate in the interaction of LDs with C. trachomatis inclusions.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/pathogenicity , Lipid Droplets/metabolism , Membrane Proteins/metabolism , Animals , Chlorocebus aethiops , HeLa Cells , Humans , Lipid Droplets/microbiology , Vero Cells
6.
Microb Cell ; 6(9): 414-449, 2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31528632

ABSTRACT

Chlamydia trachomatis serovars are obligate intracellular bacterial pathogens mainly causing ocular and urogenital infections that affect millions of people worldwide and which can lead to blindness or sterility. They reside and multiply intracellularly within a membrane-bound vacuolar compartment, known as inclusion, and are characterized by a developmental cycle involving two morphologically and physiologically distinct chlamydial forms. Completion of the developmental cycle involves the secretion of > 70 C. trachomatis proteins that function in the host cell cytoplasm and nucleus, in the inclusion membrane and lumen, and in the extracellular milieu. These proteins can, for example, interfere with the host cell cytoskeleton, vesicular and non-vesicular transport, metabolism, and immune signalling. Generally, this promotes C. trachomatis invasion into, and escape from, host cells, the acquisition of nutrients by the chlamydiae, and evasion of cell-autonomous, humoral and cellular innate immunity. Here, we present an in-depth review on the current knowledge and outstanding questions about these C. trachomatis secreted proteins.

7.
Sci Rep ; 9(1): 6133, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992493

ABSTRACT

Chlamydia trachomatis is a bacterial pathogen causing ocular and genital infections in humans. C. trachomatis multiplies exclusively inside host cells within a characteristic vacuole, from where it manipulates host cells by injecting them with type III secretion effector proteins. Here, we identified CteG as the first C. trachomatis effector associated with the Golgi. For this, C. trachomatis strains expressing candidate effectors fused to a double hemagglutinin (2HA) tag were constructed. Then, among these strains, immunofluorescence microscopy revealed that CteG-2HA was delivered into the cytoplasm of infected cells. Between 16-20 h post-infection, CteG-2HA mostly associated with the Golgi; however, CteG-2HA also appeared at the host cell plasma membrane, and at 30 or 40 h post-infection this was its predominant localization. This change in the main localization of CteG-2HA was independent of intact microfilaments or microtubules. Ectopic expression of different regions of CteG (656 amino acid residues) in uninfected cells revealed that its first 100 residues contain a Golgi targeting region. Although a C. trachomatis cteG mutant did not display a defect in intracellular multiplication, CteG induced a vacuolar protein sorting defect when expressed in Saccharomyces cerevisiae. This suggested that CteG might function by subverting host cell vesicular transport.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia Infections/pathology , Chlamydia trachomatis/pathogenicity , Golgi Apparatus/metabolism , Animals , Bacterial Proteins/genetics , Cell Membrane/metabolism , Chlamydia Infections/microbiology , Chlamydia trachomatis/metabolism , Chlorocebus aethiops , HeLa Cells , Humans , Mutation , Transport Vesicles/metabolism , Vacuoles/metabolism , Vero Cells
8.
mBio ; 9(5)2018 10 02.
Article in English | MEDLINE | ID: mdl-30279280

ABSTRACT

Nonflagellar type III secretion systems (nf T3SSs) form a cell surface needle-like structure and an associated translocon that deliver bacterial effector proteins into eukaryotic host cells. This involves a tightly regulated hierarchy of protein secretion. A switch involving SctP and SctU stops secretion of the needle protein. The gatekeeper protein SctW is required for secretion of translocon proteins and controls a second switch to start effector secretion. Salmonella enterica serovar Typhimurium encodes two T3SSs in Salmonella pathogenicity island 1 (SPI-1) and SPI-2. The acidic vacuole containing intracellular bacteria stimulates assembly of the SPI-2 T3SS and its translocon. Sensing the nearly neutral host cytosolic pH is required for effector translocation. Here, we investigated the involvement of SPI-2-encoded proteins SsaP (SctP), SsaU (SctU), SsaV (SctV), and SsaL (SctW) in regulation of secretion. We found that SsaP and SsaU are involved in the first but not the second secretion switch. A random-mutagenesis screen identified amino acids of SsaV that regulate translocon and effector secretion. Single substitutions in subdomain 4 of SsaV or InvA (SPI-1-encoded SctV) phenocopied mutations of their corresponding gatekeepers with respect to translocon and effector protein secretion and host cell interactions. SsaL interacted with SsaV in bacteria exposed to low ambient pH but not after the pH was raised to 7.2. We propose that SsaP and SsaU enable the apparatus to become competent for a secretion switch and facilitate the SsaL-SsaV interaction. This mediates secretion of translocon proteins until neutral pH is sensed, which causes their dissociation, resulting in arrest of translocon secretion and derepression of effector translocation.IMPORTANCESalmonella Typhimurium is an intracellular pathogen that uses the SPI-2 type III secretion system to deliver virulence proteins across the vacuole membrane surrounding intracellular bacteria. This involves a tightly regulated hierarchy of protein secretion controlled by two molecular switches. We found that SPI-2-encoded proteins SsaP and SsaU are involved in the first but not the second secretion switch. We identify key amino acids of the inner membrane protein SsaV that are required to interact with the so-called gatekeeper protein SsaL and show that the dissociation of SsaV-SsaL causes the second switch, leading to delivery of effector proteins. Our results provide insights into the molecular events controlling virulence-associated type III secretion and suggest a broader model describing how the process is regulated.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Genomic Islands , Protein Interaction Mapping , Type III Secretion Systems/genetics , Amino Acid Substitution , Bacterial Proteins/genetics , DNA Mutational Analysis , Hydrogen-Ion Concentration , Protein Binding , Protein Multimerization
9.
Article in English | MEDLINE | ID: mdl-30094225

ABSTRACT

Chlamydia trachomatis is an obligate intracellular human pathogen causing mainly ocular and genital infections of significant clinical and public health impact. C. trachomatis multiplies intracellularly in a membrane bound vacuole, known as inclusion. Both extracellularly and from within the inclusion, C. trachomatis uses a type III secretion system to deliver several effector proteins into the cytoplasm of host cells. A large proportion of these effectors, the inclusion membrane (Inc) proteins, are exposed to the host cell cytosol but possess a characteristic hydrophobic domain mediating their insertion in the inclusion membrane. By yeast two-hybrid, we found that C. trachomatis Inc CT288 interacts with the human centrosomal protein CCDC146 (coiled-coil domain-containing protein 146). The interaction was also detected by co-immunoprecipitation in mammalian cells either ectopically expressing CCDC146 and CT288 or ectopically expressing CCDC146 and infected by a C. trachomatis strain expressing epitope-tagged and inclusion membrane-localized CT288. In uninfected mammalian cells, ectopically expressed full-length CCDC146 (955 amino acid residues) localized at the centrosome; but in cells infected by wild-type C. trachomatis, its centrosomal localization was less evident and CCDC146 accumulated around the inclusion. Recruitment of CCDC146 to the inclusion periphery did not require intact host Golgi, microtubules or microfilaments, but was dependent on chlamydial protein synthesis. Full-length CCDC146 also accumulated at the periphery of the inclusion in cells infected by a C. trachomatis ct288 mutant; however, a C-terminal fragment of CCDC146 (residues 692-955), which interacts with CT288, showed differences in localization at the periphery of the inclusion in cells infected by wild-type or ct288 mutant C. trachomatis. This suggests a model in which chlamydial proteins other than CT288 recruit CCDC146 to the periphery of the inclusion, where the CT288-CCDC146 interaction might contribute to modulate the function of this host protein.


Subject(s)
Bacterial Proteins/metabolism , Chlamydia trachomatis/physiology , Host-Pathogen Interactions , Inclusion Bodies/microbiology , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Vacuoles/microbiology , Animals , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , Inclusion Bodies/chemistry , Protein Binding , Two-Hybrid System Techniques , Vacuoles/chemistry , Vero Cells
10.
Methods Mol Biol ; 1615: 501-515, 2017.
Article in English | MEDLINE | ID: mdl-28667633

ABSTRACT

The identification of effector proteins delivered into mammalian host cells by bacterial pathogens possessing syringelike nanomachines is an important step toward understanding the mechanisms underlying the virulence of these pathogens. In this chapter, we describe a method based on mammalian tissue culture infection models where incubation with a nonionic detergent (Triton X-100) enables solubilization of host cell membranes but not of bacterial membranes. This allows the isolation of a Triton-soluble fraction lacking bacteria but enriched in proteins present in the host cell cytoplasm and plasma membrane. Using appropriate controls, this fraction can be probed by immunoblotting for the presence of bacterial effector proteins delivered into host cells.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Cell Line , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Immunoblotting , Mice , Protein Transport , RAW 264.7 Cells , Solubility , Type III Secretion Systems
11.
PLoS One ; 12(6): e0178856, 2017.
Article in English | MEDLINE | ID: mdl-28622339

ABSTRACT

Chlamydia trachomatis is a human bacterial pathogen causing ocular and genital infections. It multiplies exclusively within an intracellular membrane-bound vacuole, the inclusion, and uses a type III secretion system to manipulate host cells by injecting them with bacterially-encoded effector proteins. In this work, we characterized the expression and subcellular localization in infected host cells of the C. trachomatis CT142, CT143, and CT144 proteins, which we previously showed to be type III secretion substrates. Transcriptional analyses in C. trachomatis confirmed the prediction that ct142, ct143 and ct144 are organized in an operon and revealed that their expression is likely driven by the main σ factor, σ66. In host cells infected by C. trachomatis, production of CT142 and CT143 could be detected by immunoblotting from 20-26 h post-infection. Immunofluorescence microscopy of infected cells revealed that from 20 h post-infection CT143 appeared mostly as globular structures outside of the bacterial cells but within the lumen of the inclusion. Furthermore, immunofluorescence microscopy of cells infected by C. trachomatis strains carrying plasmids producing CT142, CT143, or CT144 under the control of the ct142 promoter and with a C-terminal double hemagglutinin (2HA) epitope tag revealed that CT142-2HA, CT143-2HA or CT144-2HA showed an identical localization to chromosomally-encoded CT143. Moreover, CT142-2HA or CT144-2HA and CT143 produced by the same bacteria co-localized in the lumen of the inclusion. Overall, these data suggest that the CT142, CT143, and CT144 type III secretion substrates are secreted into the lumen of the inclusion where they might form a protein complex.


Subject(s)
Bacterial Proteins , Bacterial Secretion Systems , Chlamydia trachomatis , Plasmids , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/metabolism , Chlamydia trachomatis/genetics , Chlamydia trachomatis/metabolism , Chlorocebus aethiops , HeLa Cells , Humans , Plasmids/genetics , Plasmids/metabolism , Vero Cells
12.
Antimicrob Agents Chemother ; 60(6): 3323-32, 2016 06.
Article in English | MEDLINE | ID: mdl-26976875

ABSTRACT

Quaternary ammonium compounds (QAC) are widely used, cheap, and chemically stable disinfectants and topical antiseptics with wide-spectrum antimicrobial activities. Within this group of compounds, we recently showed that there are significant differences between the pharmacodynamics of n-alkyl quaternary ammonium surfactants (QAS) with a short (C12) alkyl chain when in vitro toxicities toward bacterial and mammalian epithelial cells are compared. These differences result in an attractive therapeutic window that justifies studying short-chain QAS as prophylactics for sexually transmitted infections (STI) and perinatal vertically transmitted urogenital infections (UGI). We have evaluated the antimicrobial activities of short-chain (C12) n-alkyl QAS against several STI and UGI pathogens as well as against commensal Lactobacillus species. Inhibition of infection of HeLa cells by Neisseria gonorrhoeae and Chlamydia trachomatis was studied at concentrations that were not toxic to the HeLa cells. We show that the pathogenic bacteria are much more susceptible to QAS toxic effects than the commensal vaginal flora and that QAS significantly attenuate the infectivity of N. gonorrhoeae and C. trachomatis without affecting the viability of epithelial cells of the vaginal mucosa. N-Dodecylpyridinium bromide (C12PB) was found to be the most effective QAS. Our results strongly suggest that short-chain (C12) n-alkyl pyridinium bromides and structurally similar compounds are promising microbicide candidates for topical application in the prophylaxis of STI and perinatal vertical transmission of UGI.


Subject(s)
Anti-Infective Agents/pharmacology , Chlamydia trachomatis/drug effects , Gonorrhea/drug therapy , Quaternary Ammonium Compounds/pharmacology , Streptococcus/drug effects , Surface-Active Agents/pharmacology , HeLa Cells , Humans , Infectious Disease Transmission, Vertical/prevention & control , Neisseria gonorrhoeae/drug effects , Sexually Transmitted Diseases/microbiology
13.
Cell Microbiol ; 18(7): 949-69, 2016 07.
Article in English | MEDLINE | ID: mdl-26676327

ABSTRACT

Many bacterial pathogens use specialized secretion systems to deliver virulence effector proteins into eukaryotic host cells. The function of these effectors depends on their localization within infected cells, but the mechanisms determining subcellular targeting of each effector are mostly elusive. Here, we show that the Salmonella type III secretion effector SteA binds specifically to phosphatidylinositol 4-phosphate [PI(4)P]. Ectopically expressed SteA localized at the plasma membrane (PM) of eukaryotic cells. However, SteA was displaced from the PM of Saccharomyces cerevisiae in mutants unable to synthesize the local pool of PI(4)P and from the PM of HeLa cells after localized depletion of PI(4)P. Moreover, in infected cells, bacterially translocated or ectopically expressed SteA localized at the membrane of the Salmonella-containing vacuole (SCV) and to Salmonella-induced tubules; using the PI(4)P-binding domain of the Legionella type IV secretion effector SidC as probe, we found PI(4)P at the SCV membrane and associated tubules throughout Salmonella infection of HeLa cells. Both binding of SteA to PI(4)P and the subcellular localization of ectopically expressed or bacterially translocated SteA were dependent on a lysine residue near the N-terminus of the protein. Overall, this indicates that binding of SteA to PI(4)P is necessary for its localization within host cells.


Subject(s)
Bacterial Proteins/metabolism , Host-Pathogen Interactions/physiology , Phosphatidylinositol Phosphates/metabolism , Salmonella typhimurium/metabolism , Virulence Factors/metabolism , Bacterial Proteins/genetics , Cell Membrane/metabolism , Cell Membrane/microbiology , HeLa Cells/microbiology , Humans , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Vacuoles/metabolism , Virulence Factors/genetics
14.
Microbiologyopen ; 5(1): 118-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26626407

ABSTRACT

The Legionella pneumophila effector protein VipA is an actin nucleator that co-localizes with actin filaments and early endosomes in infected macrophages and which interferes with organelle trafficking when expressed in yeast. To identify the regions of VipA involved in its subcellular localization and functions, we ectopically expressed specific VipA mutant proteins in eukaryotic cells. This indicated that the characteristic punctate distribution of VipA depends on its NH2 -terminal (amino acid residues 1-133) and central coiled-coil (amino acid residues 133-206) regions, and suggested a role for the COOH-terminal (amino acid residues 206-339) region in association with actin filaments and for the NH2 -terminal in co-localization with early endosomes. Co-immunoprecipitation and in vitro assays showed that the COOH-terminal region of VipA is necessary and sufficient to mediate actin binding, and is essential but insufficient to induce microfilament formation. Assays in yeast revealed that the NH2 and the COOH-terminal regions, and possibly an NPY motif within the NH2 region of VipA, are necessary for interference with organelle trafficking. Overall, this suggests that subversion of eukaryotic vesicular trafficking by VipA involves both its ability to associate with early endosomes via its NH2 -terminal region and its capacity to bind and polymerize actin through its COOH-terminal region.


Subject(s)
Actins/metabolism , Bacterial Proteins/metabolism , Legionella pneumophila/metabolism , Organelles/metabolism , Protein Transport/physiology , Animals , CHO Cells , Cell Line , Cricetulus , Endosomes/metabolism , Escherichia coli/metabolism , Green Fluorescent Proteins , Legionella pneumophila/genetics , Macrophages/microbiology , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/metabolism
15.
Pathog Dis ; 73(9): ftv078, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416078

ABSTRACT

The actin cytoskeleton is a key target of numerous microbial pathogens, including protozoa, fungi, bacteria and viruses. In particular, bacterial pathogens produce and deliver virulence effector proteins that hijack actin dynamics to enable bacterial invasion of host cells, allow movement within the host cytosol, facilitate intercellular spread or block phagocytosis. Many of these effector proteins directly or indirectly target the major eukaryotic actin nucleator, the Arp2/3 complex, by either mimicking nucleation promoting factors or activating upstream small GTPases. In contrast, this review is focused on a recently identified class of effector proteins from Gram-negative bacteria that function as direct actin nucleators. These effector proteins mimic functional activities of formins, WH2-nucleators and Ena/VASP assembly promoting factors demonstrating that bacteria have coopted the complete set of eukaryotic actin assembly pathways. Structural and functional analyses of these nucleators have revealed several motifs and/or mechanistic activities that are shared with eukaryotic actin nucleators. However, functional effects of these proteins during infection extend beyond plain actin polymerization leading to interference with other host cell functions such as vesicle trafficking, cell cycle progression and cell death. Therefore, their use as model systems could not only help in the understanding of the mechanistic details of actin polymerization but also provide novel insights into the connection between actin dynamics and other cellular pathways.


Subject(s)
Actins/metabolism , Eukaryotic Cells/microbiology , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/pathogenicity , Host-Pathogen Interactions , Protein Multimerization , Virulence Factors/metabolism
16.
Expert Opin Drug Discov ; 10(4): 373-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25727140

ABSTRACT

INTRODUCTION: The type III secretion system (T3SS) injectisome is an essential virulence mechanism used by many bacterial pathogens to inject host cells with effector proteins. Bacteria harboring T3SSs can cause significant disease in humans. As bacterial antibiotic resistance is a major concern, alternative prophylaxis and therapeutics are needed and T3SSs are a target for anti-virulence drugs. AREAS COVERED: In this article, the authors review whole-cell-based high-throughput screens (HTSs), which have been the main approach used to identify small molecules inhibiting T3SSs. The authors review this in the context of particular characteristics of T3SSs. Furthermore, they also describe the follow-up approaches used to study the inhibitors found. The authors also highlight target-based approaches to find inhibitors of specific T3SS components. Finally, the authors briefly review strategies used to find inhibitors of effectors or of effector-activated host cell pathways, and approaches based on T3SSs for active or passive immunization and rational vaccine design. EXPERT OPINION: Future efforts targeting T3SS to prevent or treat bacterial infections should focus on deciphering the mode of action of inhibitors and on target-based approaches. The aim should not only be to find anti-T3SS drugs but also to develop novel or improved vaccines. Continuous efforts to understand many remaining fundamental questions about the structure and function of T3SSs will also be needed.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Type III Secretion Systems/antagonists & inhibitors , Animals , Bacteria/drug effects , Bacteria/pathogenicity , Bacterial Infections/microbiology , Bacterial Infections/prevention & control , Bacterial Vaccines/administration & dosage , Drug Design , Drug Resistance, Bacterial , Humans
17.
Infect Immun ; 82(7): 2923-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24778114

ABSTRACT

Salmonella enterica serovar Typhimurium is a bacterial pathogen causing gastroenteritis in humans and a typhoid-like systemic disease in mice. S. Typhimurium virulence is related to its capacity to multiply intracellularly within a membrane-bound compartment, the Salmonella-containing vacuole (SCV), and depends on type III secretion systems that deliver bacterial effector proteins into host cells. Here, we analyzed the cellular function of the Salmonella effector SteA. We show that, compared to cells infected by wild-type S. Typhimurium, cells infected by ΔsteA mutant bacteria displayed fewer Salmonella-induced filaments (SIFs), an increased clustering of SCVs, and morphologically abnormal vacuoles containing more than one bacterium. The increased clustering of SCVs and the appearance of vacuoles containing more than one bacterium were suppressed by inhibition of the activity of the microtubule motor dynein or kinesin-1. Clustering and positioning of SCVs are controlled by the effectors SseF and SseG, possibly by helping to maintain a balanced activity of microtubule motors on the bacterial vacuoles. Deletion of steA in S. Typhimurium ΔsseF or ΔsseG mutants revealed that SteA contributes to the characteristic scattered distribution of ΔsseF or ΔsseG mutant SCVs in infected cells. Overall, this shows that SteA participates in the control of SCV membrane dynamics. Moreover, it indicates that SteA is functionally linked to SseF and SseG and suggests that it might contribute directly or indirectly to the regulation of microtubule motors on the bacterial vacuoles.


Subject(s)
Bacterial Proteins/metabolism , Salmonella typhimurium/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Cell Line , Dyneins , Gene Expression Regulation/immunology , Humans , Kinesins/genetics , Kinesins/metabolism , Mice , Mutation , Virulence Factors/genetics
18.
BMC Microbiol ; 14: 40, 2014 Feb 17.
Article in English | MEDLINE | ID: mdl-24533538

ABSTRACT

BACKGROUND: Chlamydia trachomatis is an obligate intracellular human pathogen causing ocular and urogenital infections that are a significant clinical and public health concern. This bacterium uses a type III secretion (T3S) system to manipulate host cells, through the delivery of effector proteins into their cytosol, membranes, and nucleus. In this work, we aimed to find previously unidentified C. trachomatis T3S substrates. RESULTS: We first analyzed the genome of C. trachomatis L2/434 strain for genes encoding mostly uncharacterized proteins that did not appear to possess a signal of the general secretory pathway and which had not been previously experimentally shown to be T3S substrates. We selected several genes with these characteristics and analyzed T3S of the encoding proteins using Yersinia enterocolitica as a heterologous system. We identified 23 C. trachomatis proteins whose first 20 amino acids were sufficient to drive T3S of the mature form of ß-lactamase TEM-1 by Y. enterocolitica. We found that 10 of these 23 proteins were also type III secreted in their full-length versions by Y. enterocolitica, providing additional support that they are T3S substrates. Seven of these 10 likely T3S substrates of C. trachomatis were delivered by Y. enterocolitica into host cells, further suggesting that they could be effectors. Finally, real-time quantitative PCR analysis of expression of genes encoding the 10 likely T3S substrates of C. trachomatis showed that 9 of them were clearly expressed during infection of host cells. CONCLUSIONS: Using Y. enterocolitica as a heterologous system, we identified 10 likely T3S substrates of C. trachomatis (CT053, CT105, CT142, CT143, CT144, CT161, CT338, CT429, CT656, and CT849) and could detect translocation into host cells of CT053, CT105, CT142, CT143, CT161, CT338, and CT429. Therefore, we revealed several C. trachomatis proteins that could be effectors subverting host cell processes.


Subject(s)
Bacterial Secretion Systems , Chlamydia trachomatis/genetics , Chlamydia trachomatis/metabolism , Virulence Factors/metabolism , Cloning, Molecular , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression , HeLa Cells , Humans , Protein Transport , Yersinia enterocolitica/genetics , Yersinia enterocolitica/metabolism
19.
PLoS One ; 8(2): e56292, 2013.
Article in English | MEDLINE | ID: mdl-23431368

ABSTRACT

Chlamydia trachomatis is an obligate intracellular bacterial pathogen of humans that uses a type III secretion (T3S) system to manipulate host cells through the delivery of effector proteins into their cytosol and membranes. The function of T3S systems depends on small bacterial cytosolic chaperone-like proteins, which bind T3S substrates and ensure their appropriate secretion. To find novel T3S chaperone-substrate complexes of C. trachomatis we first searched its genome for genes encoding proteins with features of T3S chaperones. We then systematically tested for interactions between candidate chaperones and chlamydial T3S substrates by bacterial two-hybrid. This revealed interactions between Slc1 (a known T3S chaperone) or CT584 and several T3S substrates. Co-immunoprecipitation after protein expression in Yersinia enterocolitica and protein overlay binding assays indicated that Slc1 interacted with the N-terminal region of the known T3S substrates Tarp (a previously described substrate of Slc1), CT694, and CT695, and that CT584 interacted with a central region of CT082, which we identified as a C. trachomatis T3S substrate using Y. enterocolitica as a heterologous system. Further T3S assays in Yersinia indicated that Slc1 or CT584 increased the amount of secreted Tarp, CT694, and CT695, or CT082, respectively. Expression of CT584 increased the intra-bacterial stability of CT082, while Slc1 did not affect the stability of its substrates. Overall, this indicated that in C. trachomatis Slc1 is a chaperone of multiple T3S substrates and that CT584 is a chaperone of the newly identified T3S substrate CT082.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Chlamydia trachomatis/metabolism , Molecular Chaperones/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , HeLa Cells , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Two-Hybrid System Techniques , Yersinia enterocolitica
20.
Science ; 338(6109): 963-7, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23162002

ABSTRACT

Salmonella enterica is an intracellular bacterial pathogen that replicates within membrane-bound vacuoles through the action of effector proteins translocated into host cells. Salmonella vacuoles have characteristics of lysosomes but are reduced in hydrolytic enzymes transported by mannose-6-phosphate receptors (MPRs). We found that the effector SifA subverted Rab9-dependent retrograde trafficking of MPRs, thereby attenuating lysosome function. This required binding of SifA to its host cell target SKIP/PLEKHM2. Furthermore, SKIP regulated retrograde trafficking of MPRs in noninfected cells. Translocated SifA formed a stable complex with SKIP and Rab9 in infected cells. Sequestration of Rab9 by SifA-SKIP accounted for the effect of SifA on MPR transport and lysosome function. Growth of Salmonella increased in cells with reduced lysosomal activity and decreased in cells with higher lysosomal activity. These results suggest that Salmonella vacuoles undergo fusion with lysosomes whose potency has been reduced by SifA.


Subject(s)
Bacterial Proteins/metabolism , Glycoproteins/metabolism , Lysosomes/metabolism , Receptor, IGF Type 2/metabolism , Salmonella enterica/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , HeLa Cells , Humans , Membrane Fusion , Protein Transport , RNA, Small Interfering/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...