Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Infect Immun ; 90(12): e0045322, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36350146

ABSTRACT

The genus Chlamydia consists of diverse, obligate intracellular bacteria that infect various animals, including humans. Although chlamydial species share many aspects of the typical intracellular lifestyle, such as the biphasic developmental cycle and the preference for invasion of epithelial cells, each chlamydial strain also employs sophisticated species-specific strategies that contribute to an extraordinary diversity in organ and/or tissue tropism and disease manifestation. In order to discover and understand the mechanisms underlying how these pathogens infect particular hosts and cause specific diseases, it is imperative to develop a mutagenesis approach that would be applicable to every chlamydial species. We present functional evidence that the region between Chlamydia trachomatis and Chlamydia muridarum pgp6 and pgp7, containing four 22-bp tandem repeats that are present in all chlamydial endogenous plasmids, represents the plasmid origin of replication. Furthermore, by introducing species-specific ori regions into an engineered 5.45-kb pUC19-based plasmid, we generated vectors that can be successfully transformed into and propagated under selective pressure by C. trachomatis serovars L2 and D, as well as C. muridarum. Conversely, these vectors were rapidly lost upon removal of the selective antibiotic. This conditionally replicating system was used to generate a tarP deletion mutant by fluorescence-reported allelic exchange mutagenesis in both C. trachomatis serovar D and C. muridarum. The strains were analyzed using in vitro invasion and fitness assays. The virulence of the C. muridarum strains was then assessed in a murine infection model. Our approach represents a novel and efficient strategy for targeted genetic manipulation in Chlamydia beyond C. trachomatis L2. This advance will support comparative studies of species-specific infection biology and enable studies in a well-established murine model of chlamydial pathogenesis.


Subject(s)
Chlamydia Infections , Chlamydia muridarum , Humans , Mice , Animals , Chlamydia muridarum/genetics , Gene Knockout Techniques , Gene Deletion , Chlamydia trachomatis/genetics , Replicon , Models, Animal , Chlamydia Infections/microbiology
2.
Infect Immun ; 88(5)2020 04 20.
Article in English | MEDLINE | ID: mdl-32152196

ABSTRACT

The translocated actin recruiting phosphoprotein (Tarp) is a multidomain type III secreted effector used by Chlamydia trachomatis In aggregate, existing data suggest a role of this effector in initiating new infections. As new genetic tools began to emerge to study chlamydial genes in vivo, we speculated as to what degree Tarp function contributes to Chlamydia's ability to parasitize mammalian host cells. To address this question, we generated a complete tarP deletion mutant using the fluorescence-reported allelic exchange mutagenesis (FRAEM) technique and complemented the mutant in trans with wild-type tarP or mutant tarP alleles engineered to harbor in-frame domain deletions. We provide evidence for the significant role of Tarp in C. trachomatis invasion of host cells. Complementation studies indicate that the C-terminal filamentous actin (F-actin)-binding domains are responsible for Tarp-mediated invasion efficiency. Wild-type C. trachomatis entry into HeLa cells resulted in host cell shape changes, whereas the tarP mutant did not. Finally, using a novel cis complementation approach, C. trachomatis lacking tarP demonstrated significant attenuation in a murine genital tract infection model. Together, these data provide definitive genetic evidence for the critical role of the Tarp F-actin-binding domains in host cell invasion and for the Tarp effector as a bona fide C. trachomatis virulence factor.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/pathogenicity , Mutagenesis/genetics , Actins/genetics , Alleles , Animals , Bacterial Proteins/genetics , Cell Line, Tumor , Fluorescence , Gene Deletion , HeLa Cells , Humans , Mice , Mice, Inbred C3H , Phosphoproteins/genetics , Virulence/genetics
3.
J Immunol ; 190(4): 1695-701, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23303668

ABSTRACT

Type I IFNs are induced during microbial infections and have well-characterized antiviral activities. TRAF3 is a signaling molecule crucial for type I IFN production and, therefore, represents a potential target for disarming immune responses. Chlamydia pneumoniae is a human pathogen that primarily infects respiratory epithelial cells; the onset of symptoms takes several weeks, and the course of infection is protracted. C. pneumoniae has also been associated with a variety of chronic inflammatory conditions. Thus, typical C. pneumoniae infections of humans are consistent with an impairment in inflammatory responses to the microorganism. We demonstrate that infection of epithelial cells with C. pneumoniae does not lead to IFN-ß production. Instead, infected cells are prevented from activating IFN regulatory factor 3. This effect is mediated by C. pneumoniae-dependent degradation of TRAF3, which is independent of a functional proteasome. Hence, it is likely that C. pneumoniae expresses a unique protease targeting TRAF3-dependent immune effector mechanisms.


Subject(s)
Chlamydophila pneumoniae/immunology , Down-Regulation/immunology , Immunity, Innate , Respiratory Mucosa/immunology , TNF Receptor-Associated Factor 3/metabolism , Active Transport, Cell Nucleus/immunology , Cell Line, Tumor , Chlamydophila Infections/immunology , Chlamydophila Infections/microbiology , Chlamydophila Infections/pathology , Chlamydophila pneumoniae/pathogenicity , Down-Regulation/genetics , Gene Targeting , HeLa Cells , Humans , Immunity, Innate/genetics , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Type I/antagonists & inhibitors , Interferon Type I/biosynthesis , Interferon Type I/genetics , Phosphorylation/immunology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , TNF Receptor-Associated Factor 3/antagonists & inhibitors , TNF Receptor-Associated Factor 3/genetics
4.
J Bacteriol ; 195(18): 4221-30, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23852872

ABSTRACT

Salicylidene acylhydrazides (SAHs) inhibit the type III secretion system (T3S) of Yersinia and other Gram-negative bacteria. In addition, SAHs restrict the growth and development of Chlamydia species. However, since the inhibition of Chlamydia growth by SAH is suppressed by the addition of excess iron and since SAHs have an iron-chelating capacity, their role as specific T3S inhibitors is unclear. We investigated here whether SAHs exhibit a function on C. trachomatis that goes beyond iron chelation. We found that the iron-saturated SAH INP0341 (IS-INP0341) specifically affects C. trachomatis infectivity with reduced generation of infectious elementary body (EB) progeny. Selection and isolation of spontaneous SAH-resistant mutant strains revealed that mutations in hemG suppressed the reduced infectivity caused by IS-INP0341 treatment. Structural modeling of C. trachomatis HemG predicts that the acquired mutations are located in the active site of the enzyme, suggesting that IS-INP0341 inhibits this domain of HemG and that protoporphyrinogen oxidase (HemG) and heme metabolism are important for C. trachomatis infectivity.


Subject(s)
Bacterial Proteins/genetics , Chlamydia trachomatis/drug effects , Chlamydia trachomatis/genetics , Hydrazines/pharmacology , Mutation , Protoporphyrinogen Oxidase/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Chlamydia trachomatis/enzymology , Chlamydia trachomatis/pathogenicity , Drug Resistance, Bacterial , HeLa Cells , Heme/metabolism , Humans , Iron/metabolism , Iron/pharmacology , Models, Molecular , Molecular Sequence Data , Protoporphyrinogen Oxidase/chemistry , Protoporphyrinogen Oxidase/metabolism
5.
J Biol Chem ; 287(33): 28078-86, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22711538

ABSTRACT

The Chlamydia trachomatis type three-secreted effector protein CT694 is expressed during late-cycle development yet is secreted by infectious particles during the invasion process. We have previously described the presence of at least two functional domains within CT694. CT694 was found to interact with the human protein Ahnak through a C-terminal domain and affect formation of host-cell actin stress fibers. Immunolocalization analyses of ectopically expressed pEGFP-CT694 also revealed plasma membrane localization for CT694 that was independent of Ahnak binding. Here we provide evidence that CT694 contains multiple functional domains. Plasma membrane localization and CT694-induced alterations in host cell morphology are dependent on an N-terminal domain. We demonstrate that membrane association of CT694 is dependent on a domain resembling a membrane localization domain (MLD) found in anti-host proteins from Yersinia, Pseudomonas, and Salmonella spp. This domain is necessary and sufficient for localization and morphology changes but is not required for Ahnak binding. Further, the CT694 MLD is able to complement ExoS ΔMLD when ectopically expressed. Taken together, our data indicate that CT694 is a multidomain protein with the potential to modulate multiple host cell processes.


Subject(s)
Bacterial Proteins/metabolism , Cell Membrane/metabolism , Chlamydia Infections/metabolism , Chlamydia trachomatis/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Stress Fibers/metabolism , Bacterial Proteins/genetics , Cell Membrane/genetics , Chlamydia Infections/genetics , Chlamydia trachomatis/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Protein Structure, Tertiary , Pseudomonas/genetics , Pseudomonas/metabolism , Salmonella/genetics , Salmonella/metabolism , Stress Fibers/genetics , Yersinia/genetics , Yersinia/metabolism
6.
J Biol Chem ; 287(34): 28738-44, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22761422

ABSTRACT

In eukaryotic cells, there are two well characterized pathways that regulate translation initiation in response to stress, and each have been shown to be targeted by various viruses. We recently showed in a yeast-based model that the bacterial virulence factor YopJ disrupts one of these pathways, which is centered on the α-subunit of the translation factor eIF2. Here, we show in mammalian cells that induction of the eIF2 signaling pathway occurs following infection with bacterial pathogens and that, consistent with our yeast-based findings, YopJ reduces eIF2 signaling in response to endoplasmic reticulum stress, heavy metal toxicity, dsRNA, and bacterial infection. We demonstrate that the well documented activities of YopJ, inhibition of NF-κB activation and proinflammatory cytokine expression, are both dependent on an intact eIF2 signaling pathway. Unexpectedly, we found that cells with defective eIF2 signaling were more susceptible to bacterial invasion. This was true for pathogenic Yersinia, a facultative intracellular pathogen, as well as for the intracellular pathogens Listeria monocytogenes and Chlamydia trachomatis. Collectively, our data indicate that the highly conserved eIF2 signaling pathway, which is vitally important for antiviral responses, plays a variety of heretofore unrecognized roles in antibacterial responses.


Subject(s)
Chlamydia Infections/metabolism , Chlamydia trachomatis/metabolism , Cytokines/biosynthesis , Eukaryotic Initiation Factor-2/metabolism , Inflammation Mediators/metabolism , Listeria monocytogenes/metabolism , Listeriosis/metabolism , Signal Transduction , Yersinia Infections/metabolism , Yersinia/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Cell Line , Chlamydia Infections/genetics , Chlamydia Infections/immunology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , Cytokines/genetics , Cytokines/immunology , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/immunology , Inflammation Mediators/immunology , Listeria monocytogenes/genetics , Listeria monocytogenes/immunology , Listeriosis/genetics , Listeriosis/immunology , Mice , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Yersinia/genetics , Yersinia/immunology , Yersinia Infections/genetics , Yersinia Infections/immunology
7.
Front Cell Infect Microbiol ; 13: 1232391, 2023.
Article in English | MEDLINE | ID: mdl-37483386

ABSTRACT

Chlamydia trachomatis is an obligate intracellular pathogen that actively promotes invasion of epithelial cells. A virulence-associated type III secretion system contributes to chlamydial entry and at least four effectors have been described that are deployed during this time. Two of these invasion-related effectors, the translocated membrane-associated effectors A and B (TmeA and TmeB), are encoded in a bi-cistronic operon. TmeA directly activates host N-WASP to stimulate Arp2/3-dependent actin polymerization. According to current working models, TmeA-mediated N-WASP activation contributes to invasion. TmeB has not been functionally characterized. Unlike a tmeA null strain, loss of tmeB does not impact invasion efficiency of C. trachomatis. Using strains deficient for multiple genes, we provide evidence that TmeA is dispensable for invasion in the absence of TmeB. Our data indicate that overabundance of TmeB interferes with invasion and that this activity requires active Arp2/3 complex. We further show that TmeB is capable of interfering with Arp2/3-mediated actin polymerization. In aggregate, these data point to opposing functions for TmeA and TmeB that manifest during the invasion process. These studies raise intriguing questions regarding the dynamic interplay between TmeA, TmeB, and branched actin polymerization during chlamydial entry.


Subject(s)
Actins , Chlamydia trachomatis , Humans , HeLa Cells , Chlamydia trachomatis/genetics , Bacterial Proteins/genetics , Polymerization
8.
J Bacteriol ; 193(14): 3490-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21571996

ABSTRACT

The Chlamydia pneumoniae CopN protein is a member of the YopN/TyeA/InvE/MxiC family of secreted proteins that function to regulate the secretion of type III secretion system (T3SS) translocator and effector proteins. In this study, the Scc1 (CP0432) and Scc4 (CP0033) proteins of C. pneumoniae AR-39 were demonstrated to function together as a type III secretion chaperone that binds to an N-terminal region of CopN. The Scc1/Scc4 chaperone promoted the efficient secretion of CopN via a heterologous T3SS, whereas, the Scc3 chaperone, which binds to a C-terminal region of CopN, reduced CopN secretion.


Subject(s)
Bacterial Proteins/metabolism , Chlamydophila pneumoniae/metabolism , Molecular Chaperones/metabolism , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Chlamydophila pneumoniae/chemistry , Chlamydophila pneumoniae/genetics , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Protein Binding , Protein Transport
9.
mBio ; 12(1)2021 01 19.
Article in English | MEDLINE | ID: mdl-33468693

ABSTRACT

Chlamydia trachomatis is a medically significant human pathogen and is an epithelial-tropic obligate intracellular parasite. Invasion of nonprofessional phagocytes represents a crucial step in the infection process and has likely promoted the evolution of a redundant mechanism and routes of entry. Like many other viral and invasive bacterial pathogens, manipulation of the host cell cytoskeleton represents a focal point in Chlamydia entry. The advent of genetic techniques in C. trachomatis, such as creation of complete gene deletions via fluorescence-reported allelic exchange mutagenesis (FRAEM), is providing important tools to unravel the contributions of bacterial factors in these complex pathways. The type III secretion chaperone Slc1 directs delivery of at least four effectors during the invasion process. Two of these, TarP and TmeA, have been associated with manipulation of actin networks and are essential for normal levels of invasion. The functions of TarP are well established, whereas TmeA is less well characterized. We leverage chlamydial genetics and proximity labeling here to provide evidence that TmeA directly targets host N-WASP to promote Arp2/3-dependent actin polymerization. Our work also shows that TmeA and TarP influence separate, yet synergistic pathways to accomplish chlamydial entry. These data further support an appreciation that a pathogen, confined by a reductionist genome, retains the ability to commit considerable resources to accomplish bottle-neck steps during the infection process.IMPORTANCE The increasing genetic tractability of Chlamydia trachomatis is accelerating the ability to characterize the unique infection biology of this obligate intracellular parasite. These efforts are leading to a greater understanding of the molecular events associated with key virulence requirements. Manipulation of the host actin cytoskeleton plays a pivotal role throughout Chlamydia infection, yet a thorough understanding of the molecular mechanisms initiating and orchestrating actin rearrangements has lagged. Our work highlights the application of genetic manipulation to address open questions regarding chlamydial invasion, a process essential to survival. We provide definitive insight regarding the role of the type III secreted effector TmeA and how that activity relates to another prominent effector, TarP. In addition, our data implicate at least one source that contributes to the functional divergence of entry mechanisms among chlamydial species.


Subject(s)
Actins/genetics , Bacterial Proteins/genetics , Chlamydia trachomatis/genetics , Cytoskeleton/metabolism , Molecular Chaperones/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Actin-Related Protein 2/genetics , Actin-Related Protein 2/metabolism , Actin-Related Protein 3/genetics , Actin-Related Protein 3/metabolism , Actins/metabolism , Bacterial Proteins/metabolism , Cell Line , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/metabolism , Cytoskeleton/microbiology , Cytoskeleton/ultrastructure , Epithelial Cells/microbiology , Gene Expression Regulation , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Molecular Chaperones/metabolism , Polymerization , Signal Transduction , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
10.
Cell Microbiol ; 11(5): 769-79, 2009 May.
Article in English | MEDLINE | ID: mdl-19159390

ABSTRACT

Chlamydia pneumoniae is a common respiratory pathogen that has been associated with a variety of chronic diseases including asthma and atherosclerosis. Chlamydiae are obligate intracellular parasites that primarily infect epithelial cells where they develop within a membrane-bound vacuole, termed an inclusion. Interactions between the microorganism and eukaryotic cell can be mediated by chlamydial proteins inserted into the inclusion membrane. We describe here a novel C. pneumoniae-specific inclusion membrane protein (Inc) CP0236, which contains domains exposed to the host cytoplasm. We demonstrate that, in a yeast two-hybrid screen, CP0236 interacts with the NFκB activator 1 (Act1) and this interaction was confirmed in HeLa 229 cells where ectopically expressed CP0236 was co-immunoprecipitated with endogenous Act1. Furthermore, we demonstrate that Act1 displays an altered distribution in the cytoplasm of HeLa cells infected with C. pneumoniae where it associates with the chlamydial inclusion membrane. This sequestration of Act1 by chlamydiae inhibited recruitment of the protein to the interleukin-17 (IL-17) receptor upon stimulation of C. pneumoniae-infected cells with IL-17A. Such inhibition of the IL-17 signalling pathway led to protection of Chlamydia-infected cells from NFκB activation in IL-17-stimulated cells. We describe here a unique strategy employed by C. pneumoniae to achieve inhibition of NFκB activation via interaction of CP0236 with mammalian Act1.


Subject(s)
Bacterial Proteins/metabolism , Chlamydophila pneumoniae/pathogenicity , Interleukin-17/metabolism , Membrane Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Adaptor Proteins, Signal Transducing , Bacterial Proteins/chemistry , Bacterial Proteins/physiology , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/physiology , Models, Biological , NF-kappa B/metabolism
11.
Front Immunol ; 11: 1490, 2020.
Article in English | MEDLINE | ID: mdl-32760406

ABSTRACT

Dynamic interactions that govern the balance between host and pathogen determine the outcome of infection and are shaped by evolutionary pressures. Eukaryotic hosts have evolved elaborate and formidable defense mechanisms that provide the basis for innate and adaptive immunity. Proteins containing a membrane attack complex/Perforin (MACPF) domain represent an important class of immune effectors. These pore-forming proteins induce cell killing by targeting microbial or host membranes. Intracellular bacteria can be shielded from MACPF-mediated killing, and Chlamydia spp. represent a successful paradigm of obligate intracellular parasitism. Ancestors of present-day Chlamydia likely originated at evolutionary times that correlated with or preceded many host defense pathways. We discuss the current knowledge regarding how chlamydiae interact with the MACPF proteins Complement C9, Perforin-1, and Perforin-2. Current evidence indicates a degree of resistance by Chlamydia to MACPF effector mechanisms. In fact, chlamydiae have acquired and adapted their own MACPF-domain protein to facilitate infection.


Subject(s)
Chlamydia Infections/immunology , Chlamydia/physiology , Complement C9/metabolism , Complement Membrane Attack Complex/metabolism , Perforin/metabolism , Animals , Biological Evolution , Complement C9/genetics , Complement Membrane Attack Complex/genetics , Host-Pathogen Interactions , Humans , Immunity, Innate , Perforin/genetics
12.
J Vis Exp ; (155)2020 01 30.
Article in English | MEDLINE | ID: mdl-32065159

ABSTRACT

Chlamydia trachomatis is an obligate intracellular pathogen that has been historically difficult to genetically manipulate. Definitive progress in elucidating the mechanisms that C. trachomatis use to create and maintain a privileged intracellular niche has been limited due to a lack of genetic tools. Fortunately, there have recently been several new advances in genetic manipulation techniques. Among these is the development of fluorescence-reported allelic exchange mutagenesis (FRAEM). This method allows targeted gene deletion coupled with insertion of a selection cassette encoding antibiotic resistance and green fluorescent protein (GFP). Reliance on this strategy can be complicated when targeting genes within polycistronic operons due to the potential of polar effects on downstream genes. Floxed cassette allelic exchange mutagenesis (FLAEM), the protocol for which is described here, was developed to alleviate cassette-induced polar effects. FLAEM utilizes Cre-loxP genome editing to remove the selection cassette after targeted deletion by allelic exchange. The resulting strains contain markerless gene deletions of one or more coding sequences. This technique facilitates direct assessment of gene function and expands the repertoire of tools for genetic manipulation in C. trachomatis.


Subject(s)
Alleles , Chlamydia trachomatis/genetics , Gene Deletion , Mutagenesis, Insertional/genetics , Mutagenesis/genetics , Base Sequence , DNA, Bacterial/genetics , Genome, Bacterial , Green Fluorescent Proteins/genetics , Integrases/metabolism , Transformation, Genetic
13.
Methods Mol Biol ; 2042: 151-164, 2019.
Article in English | MEDLINE | ID: mdl-31385275

ABSTRACT

Progress in understanding molecular mechanisms contributing to chlamydial pathogenesis has been greatly facilitated by recent advances in genetic manipulation of C. trachomatis. Valuable approaches such as random, chemically induced mutagenesis or targeted, insertion-based gene disruption have led to significant discoveries. We describe herein a technique for generating definitive null strains via complete deletion of chromosomal genes in C. trachomatis. Fluorescence-reported allelic exchange mutagenesis (FRAEM), using the suicide vector pSUmC, enables targeted deletion of desired chromosomal DNA. The protocol provided here describes steps required to produce transformation competent chlamydiae, generate a specific allelic exchange plasmid construct, carry out mutagenesis, and isolate clonal populations of resulting mutant strains.


Subject(s)
Chlamydia trachomatis/genetics , Gene Deletion , Gene Targeting/methods , Mutagenesis , Alleles , Chlamydia Infections/microbiology , Chromosomes, Bacterial , Humans , Transformation, Genetic
14.
Microbes Infect ; 20(7-8): 445-450, 2018.
Article in English | MEDLINE | ID: mdl-29409975

ABSTRACT

The intonation "The king is dead, long live the king" aptly describes the state of Chlamydia research. Genetic-based approaches are rapidly replacing correlative strategies to provide new insights. We describe how current transformation technologies are enhancing progress in understanding Chlamydia infection biology and present key opportunities for further development.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia/physiology , Animals , Bacterial Proteins/genetics , Chlamydia/genetics , Chlamydia/pathogenicity , Chlamydia Infections/physiopathology , Cloning, Molecular , DNA Transformation Competence , Gene Transfer Techniques , Humans , Mutagenesis , Virulence Factors/genetics
15.
Curr Protoc Microbiol ; 45: 11A.3.1-11A.3.15, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28510361

ABSTRACT

Gene inactivation is essential for forward and reverse genetic approaches to establish protein function. Techniques such as insertion or chemical mutagenesis have been developed to mutagenize chlamydiae via targeted or random mutagenesis, respectively. Both of these approaches require transformation of chlamydiae to either introduce insertion elements or complement mutants. We have recently developed a targeted mutagenesis strategy, fluorescence-reported allelic exchange mutagenesis (FRAEM), to delete Chlamydia trachomatis L2 genes. This approach overcomes several barriers for genetically manipulating intracellular bacteria. Perhaps most significantly, FRAEM employs fluorescence reporting to indicate successful transformation and subsequent recombination events. Three protocols are provided that detail methods to construct gene-specific suicide vectors, transform C. trachomatis L2 to select for recombinants, and isolate clonal populations via limiting dilution. In aggregate, these protocols will allow investigators to engineer C. trachomatis L2 strains carrying complete deletions of desired gene(s). © 2017 by John Wiley & Sons, Inc.


Subject(s)
Chlamydia trachomatis/genetics , Gene Knockout Techniques/methods , Transformation, Bacterial
16.
Microbes Infect ; 18(2): 84-92, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26515030

ABSTRACT

It has been appreciated for almost 20 years that members of the Chlamydiales possess a virulence-associated type III secretion mechanism. Given the obligate intracellular nature of these bacteria, defining exactly how type III secretion functions to promote pathogenesis has been challenging. We present a working model herein that is based on current evidence.


Subject(s)
Chlamydia/pathogenicity , Type III Secretion Systems/metabolism , Virulence Factors/metabolism , Chlamydia/chemistry , Chlamydia/metabolism , Models, Biological , Models, Molecular , Type III Secretion Systems/chemistry
17.
mBio ; 7(1): e01817-15, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26787828

ABSTRACT

UNLABELLED: Although progress in Chlamydia genetics has been rapid, genomic modification has previously been limited to point mutations and group II intron insertions which truncate protein products. The bacterium has thus far been intractable to gene deletion or more-complex genomic integrations such as allelic exchange. Herein, we present a novel suicide vector dependent on inducible expression of a chlamydial gene that renders Chlamydia trachomatis fully genetically tractable and permits rapid reverse genetics by fluorescence-reported allelic exchange mutagenesis (FRAEM). We describe the first available system of targeting chlamydial genes for deletion or allelic exchange as well as curing plasmids from C. trachomatis serovar L2. Furthermore, this approach permits the monitoring of mutagenesis by fluorescence microscopy without disturbing bacterial growth, a significant asset when manipulating obligate intracellular organisms. As proof of principle, trpA was successfully deleted and replaced with a sequence encoding both green fluorescent protein (GFP) and ß-lactamase. The trpA-deficient strain was unable to grow in indole-containing medium, and this phenotype was reversed by complementation with trpA expressed in trans. To assess reproducibility at alternate sites, FRAEM was repeated for genes encoding type III secretion effectors CTL0063, CTL0064, and CTL0065. In all four cases, stable mutants were recovered one passage after the observation of transformants, and allelic exchange was limited to the specific target gene, as confirmed by whole-genome sequencing. Deleted sequences were not detected by quantitative real-time PCR (qPCR) from isogenic mutant populations. We demonstrate that utilization of the chlamydial suicide vector with FRAEM renders C. trachomatis highly amenable to versatile and efficient genetic manipulation. IMPORTANCE: The obligate intracellular nature of a variety of infectious bacteria presents a significant obstacle to the development of molecular genetic tools for dissecting pathogenicity. Although progress in chlamydial genetics has been rapid, genomic modification has previously been limited to point mutations and group II intron insertions which truncate protein products. The bacterium has thus far been intractable to gene deletion or more-complex genomic integrations such as allelic exchange. Here, we present a novel suicide vector dependent on inducible expression of a chlamydial gene that renders Chlamydia trachomatis fully genetically tractable and permits rapid reverse genetics by fluorescence-reported allelic exchange mutagenesis (FRAEM). We describe the first available system of targeting chlamydial genes for deletion or allelic exchange as well as curing plasmids from C. trachomatis L2. Furthermore, this approach permits monitoring of mutagenesis by fluorescence microscopy without disturbing bacterial growth, a significant asset when manipulating obligate intracellular organisms.


Subject(s)
Chlamydia trachomatis/genetics , Fluorescence , Gene Deletion , Gene Knockout Techniques/methods , Genes, Reporter , Genetics, Microbial/methods , Chromosomes, Bacterial , Microscopy, Fluorescence , Plasmids
18.
PLoS One ; 10(8): e0135295, 2015.
Article in English | MEDLINE | ID: mdl-26258949

ABSTRACT

Chlamydia spp. utilize multiple secretion systems, including the type III secretion system (T3SS), to deploy host-interactive effector proteins into infected host cells. Elucidation of secreted proteins has traditionally required ectopic expression in a surrogate T3SS followed by immunolocalization of endogenous candidate effectors to confirm secretion by chlamydiae. The ability to transform Chlamydia and achieve stable expression of recombinant gene products has enabled a more direct assessment of secretion. We adapted TEM-1 ß-lactamase as a reporter system for assessment of chlamydial protein secretion. We provide evidence that this system facilitates visualization of secretion in the context of infection. Specifically, our findings provide definitive evidence that C. trachomatis CT695 is secreted during infection. Follow-up indirect immunofluorescence studies confirmed CT695 secretion and indicate that this effector can be secreted at multiple points during the chlamydial developmental cycle. Our results indicate that the BlaM-fusion reporter assay will allow efficacious identification of novel secreted proteins. Moreover, this approach can easily be adapted to enable more sophisticated studies of the secretion process in Chlamydia.


Subject(s)
Bacterial Proteins/genetics , Chlamydia trachomatis/genetics , Gene Expression Regulation, Bacterial , Type III Secretion Systems/genetics , beta-Lactamases/genetics , Bacterial Proteins/metabolism , Base Sequence , Chlamydia trachomatis/metabolism , Chlamydia trachomatis/pathogenicity , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Sequence Data , Neisseria meningitidis/chemistry , Neisseria meningitidis/genetics , Plasmids/chemistry , Plasmids/metabolism , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transformation, Bacterial , Type III Secretion Systems/metabolism , beta-Lactamases/metabolism , Red Fluorescent Protein
19.
PLoS One ; 8(5): e63732, 2013.
Article in English | MEDLINE | ID: mdl-23700432

ABSTRACT

Pannexin 1 (Panx1) is a plasma membrane channel glycoprotein that plays a role in innate immune response through association with the inflammasome complex. Probenecid, a classic pharmacological agent for gout, has also been used historically in combination therapy with antibiotics to prevent cellular drug efflux and has been reported to inhibit Panx1. As the inflammasome has been implicated in the progression of Chlamydia infections, and with chlamydial infections at record levels in the US, we therefore investigated whether probenecid would have a direct effect on Chlamydia trachomatis development through inhibition of Panx1. We found chlamydial development to be inhibited in a dose-dependent, yet reversible manner in the presence of probenecid. Drug treatment induced an aberrant chlamydial morphology consistent with persistent bodies. Although Panx1 was shown to localize to the chlamydial inclusion, no difference was seen in chlamydial development during infection of cells derived from wild-type and Panx1 knockout mice. Therefore, probenecid may inhibit C. trachomatis growth by an as yet unresolved mechanism.


Subject(s)
Chlamydia Infections/metabolism , Chlamydia trachomatis/physiology , Connexins/physiology , Nerve Tissue Proteins/physiology , Animals , Anti-Bacterial Agents/pharmacology , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/drug effects , Chlamydia trachomatis/immunology , Connexins/antagonists & inhibitors , HeLa Cells , Host-Pathogen Interactions , Humans , Immunity, Innate , Mice , Mice, Knockout , Microbial Sensitivity Tests , Nerve Tissue Proteins/antagonists & inhibitors , Probenecid/pharmacology , Protein Transport
20.
PLoS One ; 8(7): e68754, 2013.
Article in English | MEDLINE | ID: mdl-23874749

ABSTRACT

Here we show that cells lacking the heme-regulated inhibitor (HRI) are highly resistant to infection by bacterial pathogens. By examining the infection process in wild-type and HRI null cells, we found that HRI is required for pathogens to execute their virulence-associated cellular activities. Specifically, unlike wild-type cells, HRI null cells infected with the gram-negative bacterial pathogen Yersinia are essentially impervious to the cytoskeleton-damaging effects of the Yop virulence factors. This effect is due to reduced functioning of the Yersinia type 3 secretion (T3S) system which injects virulence factors directly into the host cell cytosol. Reduced T3S activity is also observed in HRI null cells infected with the bacterial pathogen Chlamydia which results in a dramatic reduction in its intracellular proliferation. We go on to show that a HRI-mediated process plays a central role in the cellular infection cycle of the Gram-positive pathogen Listeria. For this pathogen, HRI is required for the post-invasion trafficking of the bacterium to the infected host cytosol. Thus by depriving Listeria of its intracellular niche, there is a highly reduced proliferation of Listeria in HRI null cells. We provide evidence that these infection-associated functions of HRI (an eIF2α kinase) are independent of its activity as a regulator of protein synthesis. This is the first report of a host factor whose absence interferes with the function of T3S secretion and cytosolic access by pathogens and makes HRI an excellent target for inhibitors due to its broad virulence-associated activities.


Subject(s)
Bacteria/pathogenicity , Host-Pathogen Interactions , Virulence Factors/physiology , eIF-2 Kinase/physiology , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Cells, Cultured , Chlamydia trachomatis/pathogenicity , Disease Resistance/genetics , Female , Gene Knockdown Techniques , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Listeria monocytogenes/pathogenicity , Male , Mice , Yersinia pseudotuberculosis/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL