Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 224
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(33): e2405209121, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39106308

RÉSUMÉ

The obligatory intracellular bacterium Anaplasma phagocytophilum causes human granulocytic anaplasmosis, an emerging zoonosis. Anaplasma has limited biosynthetic and metabolic capacities, yet it effectively replicates inside of inclusions/vacuoles of eukaryotic host cells. Here, we describe a unique Type IV secretion system (T4SS) effector, ER-Golgi exit site protein of Anaplasma (EgeA). In cells infected by Anaplasma, secreted native EgeA, EgeA-GFP, and the C-terminal half of EgeA (EgeA-C)-GFP localized to Anaplasma-containing inclusions. In uninfected cells, EgeA-C-GFP localized to cis-Golgi, whereas the N-terminal half of EgeA-GFP localized to the ER. Pull-down assays identified EgeA-GFP binding to a transmembrane protein in the ER, Transport and Golgi organization protein 1 (TANGO1). By yeast two-hybrid analysis, EgeA-C directly bound Sec1 family domain-containing protein 1 (SCFD1), a host protein of the cis-Golgi network that binds TANGO1 at ER-Golgi exit sites (ERES). Both TANGO1 and SCFD1 localized to the Anaplasma inclusion surface. Furthermore, knockdown of Anaplasma EgeA or either host TANGO1 or SCFD1 significantly reduced Anaplasma infection. TANGO1 and SCFD1 prevent ER congestion and stress by facilitating transport of bulky or unfolded proteins at ERES. A bulky cargo collagen and the ER-resident chaperon BiP were transported into Anaplasma inclusions, and several ER stress marker genes were not up-regulated in Anaplasma-infected cells. Furthermore, EgeA transfection reduced collagen overexpression-induced BiP upregulation. These results suggest that by binding to the two ERES proteins, EgeA redirects the cargo-adapted ERES to pathogen-occupied inclusions and reduces ERES congestion, which facilitates Anaplasma nutrient acquisition and reduces ER stress for Anaplasma survival and proliferation.


Sujet(s)
Anaplasma phagocytophilum , Protéines bactériennes , Réticulum endoplasmique , Appareil de Golgi , Anaplasma phagocytophilum/métabolisme , Anaplasma phagocytophilum/pathogénicité , Réticulum endoplasmique/métabolisme , Humains , Appareil de Golgi/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Corps d'inclusion/métabolisme , Corps d'inclusion/microbiologie , Animaux , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/génétique , Interactions hôte-pathogène
2.
J Infect Dis ; 230(1): 188-197, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39052722

RÉSUMÉ

The subtilisin-like protease-1 (SspA-1) plays an important role in the pathogenesis of a highly virulent strain of Streptococcus suis 2. However, the mechanism of SspA-1-triggered excessive inflammatory response is still unknown. In this study, we demonstrated that activation of type I IFN signaling is required for SspA-1-induced excessive proinflammatory cytokine production. Further experiments showed that the TLR2 endosomal pathway mediates SspA-1-induced type I IFN signaling and the inflammatory response. Finally, we mapped the major signaling components of the related pathway and found that the TIR adaptor proteins Mal, TRAM, and MyD88 and the downstream activation of IRF1 and IRF7 were involved in this pathway. These results explain the molecular mechanism by which SspA-1 triggers an excessive inflammatory response and reveal a novel effect of type I IFN in S. suis 2 infection, possibly providing further insights into the pathogenesis of this highly virulent S. suis 2 strain.


Sujet(s)
Cytokines , Endosomes , Interféron de type I , Transduction du signal , Streptococcus suis , Récepteur de type Toll-2 , Streptococcus suis/immunologie , Streptococcus suis/pathogénicité , Streptococcus suis/métabolisme , Interféron de type I/métabolisme , Récepteur de type Toll-2/métabolisme , Cytokines/métabolisme , Animaux , Endosomes/métabolisme , Souris , Infections à streptocoques/immunologie , Infections à streptocoques/microbiologie , Infections à streptocoques/métabolisme , Protéines bactériennes/métabolisme , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/génétique , Humains , Facteur de différenciation myéloïde-88/métabolisme , Facteur de différenciation myéloïde-88/génétique , Souris de lignée C57BL
3.
ISME J ; 18(1)2024 Jan 08.
Article de Anglais | MEDLINE | ID: mdl-38959853

RÉSUMÉ

Effector proteins secreted by bacteria that infect mammalian and plant cells often subdue eukaryotic host cell defenses by simultaneously affecting multiple targets. However, instances when a bacterial effector injected in the competing bacteria sabotage more than a single target have not been reported. Here, we demonstrate that the effector protein, LtaE, translocated by the type IV secretion system from the soil bacterium Lysobacter enzymogenes into the competing bacterium, Pseudomonas protegens, affects several targets, thus disabling the antibacterial defenses of the competitor. One LtaE target is the transcription factor, LuxR1, that regulates biosynthesis of the antimicrobial compound, orfamide A. Another target is the sigma factor, PvdS, required for biosynthesis of another antimicrobial compound, pyoverdine. Deletion of the genes involved in orfamide A and pyoverdine biosynthesis disabled the antibacterial activity of P. protegens, whereas expression of LtaE in P. protegens resulted in the near-complete loss of the antibacterial activity against L. enzymogenes. Mechanistically, LtaE inhibits the assembly of the RNA polymerase complexes with each of these proteins. The ability of LtaE to bind to LuxR1 and PvdS homologs from several Pseudomonas species suggests that it can sabotage defenses of various competitors present in the soil or on plant matter. Our study thus reveals that the multi-target effectors have evolved to subdue cell defenses not only in eukaryotic hosts but also in bacterial competitors.


Sujet(s)
Protéines bactériennes , Lysobacter , Pseudomonas , Systèmes de sécrétion de type IV , Pseudomonas/génétique , Pseudomonas/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Lysobacter/génétique , Lysobacter/métabolisme , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Régulation de l'expression des gènes bactériens , Oligopeptides/métabolisme , Oligopeptides/génétique , Transactivateurs/génétique , Transactivateurs/métabolisme , Protéines de répression/génétique , Protéines de répression/métabolisme , Facteur sigma/génétique , Facteur sigma/métabolisme
4.
Curr Opin Microbiol ; 80: 102495, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38908045

RÉSUMÉ

Legionella species are Gram-negative intracellular bacteria that evolved in soil and freshwater environments, where they infect and replicate within various unicellular protozoa. The primary virulence factor of Legionella is the expression of a type IV secretion system (T4SS), which contributes to the translocation of effector proteins that subvert biological processes of the host cells. Because of its evolution in unicellular organisms, T4SS effector proteins are not adapted to subvert specific mammalian signaling pathways and immunity. Consequently, Legionella pneumophila has emerged as an interesting infection model for investigating immune responses against pathogenic bacteria in multicellular organisms. This review highlights recent advances in our understanding of mammalian innate immunity derived from studies involving L. pneumophila. This includes recent insights into inflammasome-mediated mechanisms restricting bacterial replication in macrophages, mechanisms inducing cell death in response to infection, induction of effector-triggered immunity, activation of specific pulmonary cell types in mammalian lungs, and the protective role of recruiting monocyte-derived cells to infected lungs.


Sujet(s)
Immunité innée , Legionella pneumophila , Maladie des légionnaires , Legionella pneumophila/immunologie , Legionella pneumophila/pathogénicité , Humains , Animaux , Maladie des légionnaires/immunologie , Maladie des légionnaires/microbiologie , Phagocytes/immunologie , Phagocytes/microbiologie , Systèmes de sécrétion de type IV/immunologie , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Inflammasomes/immunologie , Inflammasomes/métabolisme , Monocytes/immunologie , Monocytes/microbiologie , Facteurs de virulence/immunologie , Facteurs de virulence/métabolisme , Macrophages/immunologie , Macrophages/microbiologie , Interactions hôte-pathogène/immunologie
5.
mBio ; 15(8): e0048824, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-38940556

RÉSUMÉ

Conjugative type 4 secretion systems (T4SSs) are the main driver for the spread of antibiotic resistance genes and virulence factors in bacteria. To deliver the DNA substrate to recipient cells, it must cross the cell envelopes of both donor and recipient bacteria. In the T4SS from the enterococcal conjugative plasmid pCF10, PrgK is known to be the active cell wall degrading enzyme. It has three predicted extracellular hydrolase domains: metallo-peptidase (LytM), soluble lytic transglycosylase (SLT), and cysteine, histidine-dependent amidohydrolases/peptidases (CHAP). Here, we report the structure of the LytM domain and show that its active site is degenerate and lacks the active site metal. Furthermore, we show that only the predicted SLT domain is functional in vitro and that it unexpectedly has a muramidase instead of a lytic transglycosylase activity. While we did not observe any peptidoglycan hydrolytic activity for the LytM or CHAP domain, we found that these domains downregulated the SLT muramidase activity. The CHAP domain was also found to be involved in PrgK dimer formation. Furthermore, we show that PrgK interacts with PrgL, which likely targets PrgK to the rest of the T4SS. The presented data provides important information for understanding the function of Gram-positive T4SSs.IMPORTANCEAntibiotic resistance is a large threat to human health and is getting more prevalent. One of the major contributors to the spread of antibiotic resistance among different bacteria is type 4 secretion systems (T4SS). However, mainly T4SSs from Gram-negative bacteria have been studied in detail. T4SSs from Gram-positive bacteria, which stand for more than half of all hospital-acquired infections, are much less understood. The significance of our research is in identifying the function and regulation of a cell wall hydrolase, a key component of the pCF10 T4SS from Enterococcus faecalis. This system is one of the best-studied Gram-positive T4SSs, and this added knowledge aids in our understanding of horizontal gene transfer in E. faecalis as well as other medically relevant Gram-positive bacteria.


Sujet(s)
Paroi cellulaire , Enterococcus faecalis , Lysozyme , Systèmes de sécrétion de type IV , Paroi cellulaire/métabolisme , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/génétique , Enterococcus faecalis/génétique , Enterococcus faecalis/enzymologie , Enterococcus faecalis/métabolisme , Lysozyme/métabolisme , Lysozyme/génétique , Plasmides/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Conjugaison génétique , Domaine catalytique
6.
mBio ; 15(7): e0072624, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38847540

RÉSUMÉ

The modulation of actin polymerization is a common theme among microbial pathogens. Even though microorganisms show a wide repertoire of strategies to subvert the activity of actin, most of them converge in the ones that activate nucleating factors, such as the Arp2/3 complex. Brucella spp. are intracellular pathogens capable of establishing chronic infections in their hosts. The ability to subvert the host cell response is dependent on the capacity of the bacterium to attach, invade, avoid degradation in the phagocytic compartment, replicate in an endoplasmic reticulum-derived compartment and egress. Even though a significant number of mechanisms deployed by Brucella in these different phases have been identified and characterized, none of them have been described to target actin as a cellular component. In this manuscript, we describe the identification of a novel virulence factor (NpeA) that promotes niche formation. NpeA harbors a short linear motif (SLiM) present within an amphipathic alpha helix that has been described to bind the GTPase-binding domain (GBD) of N-WASP and stabilizes the autoinhibited state. Our results show that NpeA is secreted in a Type IV secretion system-dependent manner and that deletion of the gene diminishes the intracellular replication capacity of the bacterium. In vitro and ex vivo experiments demonstrate that NpeA binds N-WASP and that the short linear motif is required for the biological activity of the protein.IMPORTANCEThe modulation of actin-binding effectors that regulate the activity of this fundamental cellular protein is a common theme among bacterial pathogens. The neural Wiskott-Aldrich syndrome protein (N-WASP) is a protein that several pathogens target to hijack actin dynamics. The highly adapted intracellular bacterium Brucella has evolved a wide repertoire of virulence factors that modulate many activities of the host cell to establish successful intracellular replication niches, but, to date, no effector proteins have been implicated in the modulation of actin dynamics. We present here the identification of a virulence factor that harbors a short linear motif (SLiM) present within an amphipathic alpha helix that has been described to bind the GTPase-binding domain (GBD) of N-WASP stabilizing its autoinhibited state. We demonstrate that this protein is a Type IV secretion effector that targets N-WASP-promoting intracellular survival and niche formation.


Sujet(s)
Protéines bactériennes , Facteurs de virulence , Protéine neuronale du syndrome de Wiskott-Aldrich , Facteurs de virulence/métabolisme , Facteurs de virulence/génétique , Protéine neuronale du syndrome de Wiskott-Aldrich/métabolisme , Protéine neuronale du syndrome de Wiskott-Aldrich/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Humains , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/génétique , Animaux , Souris , Liaison aux protéines , Brucella/métabolisme , Brucella/génétique , Brucella/pathogénicité , Motifs d'acides aminés , Actines/métabolisme , Brucellose/microbiologie , Macrophages/microbiologie , Interactions hôte-pathogène
7.
mSphere ; 9(7): e0035424, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38940509

RÉSUMÉ

Bacterial conjugation systems pose a major threat to human health through their widespread dissemination of mobile genetic elements (MGEs) carrying cargoes of antibiotic resistance genes. Using the Cre Recombinase Assay for Translocation (CRAfT), we recently reported that the IncFV pED208 conjugation system also translocates at least 16 plasmid-encoded proteins to recipient bacteria. Here, we deployed a high-throughput CRAfT screen to identify the repertoire of chromosomally encoded protein substrates of the pED208 system. We identified 32 substrates encoded by the Escherichia coli W3110 genome with functions associated with (i) DNA/nucleotide metabolism, (ii) stress tolerance/physiology, (iii) transcriptional regulation, or (iv) toxin inhibition. The respective gene deletions did not impact pED208 transfer proficiencies, nor did Group 1 (DNA/nucleotide metabolism) mutations detectably alter the SOS response elicited in new transconjugants upon acquisition of pED208. However, MC4100(pED208) donor cells intrinsically exhibit significantly higher SOS activation than plasmid-free MC4100 cells, and this plasmid carriage-induced stress response is further elevated in donor cells deleted of several Group 1 genes. Among 10 characterized substrates, we gained evidence of C-terminal or internal translocation signals that could function independently or synergistically for optimal protein transfer. Remarkably, nearly all tested proteins were also translocated through the IncN pKM101 and IncP RP4 conjugation systems. This repertoire of E. coli protein substrates, here termed the F plasmid "conjutome," is thus characterized by functions of potential benefit to new transconjugants, diverse TSs, and the capacity for promiscuous transfer through heterologous conjugation systems. IMPORTANCE: Conjugation systems comprise a major subfamily of the type IV secretion systems (T4SSs) and are the progenitors of a second large T4SS subfamily dedicated to translocation of protein effectors. This study examined the capacity of conjugation machines to function as protein translocators. Using a high-throughput reporter screen, we determined that 32 chromosomally encoded proteins are delivered through an F plasmid conjugation system. The translocated proteins potentially enhance the establishment of the co-transferred F plasmid or mitigate mating-induced stresses. Translocation signals located C-terminally or internally conferred substrate recognition by the F system and, remarkably, many substrates also were translocated through heterologous conjugation systems. Our findings highlight the plasticity of conjugation systems in their capacities to co-translocate DNA and many protein substrates.


Sujet(s)
Conjugaison génétique , Protéines Escherichia coli , Escherichia coli , Systèmes de sécrétion de type IV , Escherichia coli/génétique , Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , Protéines Escherichia coli/métabolisme , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Transport des protéines , Facteur F/génétique , Facteur F/métabolisme
8.
mBio ; 15(7): e0119824, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38832773

RÉSUMÉ

Stenotrophomonas maltophilia expresses a type IV protein secretion system (T4SS) that promotes contact-dependent killing of other bacteria and does so partly by secreting the effector TfcB. Here, we report the structure of TfcB, comprising an N-terminal domain similar to the catalytic domain of glycosyl hydrolase (GH-19) chitinases and a C-terminal domain for recognition and translocation by the T4SS. Utilizing a two-hybrid assay to measure effector interactions with the T4SS coupling protein VirD4, we documented the existence of five more T4SS substrates. One of these was protein 20845, an annotated nuclease. A S. maltophilia mutant lacking the gene for 20845 was impaired for killing Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. Moreover, the cloned 20845 gene conferred robust toxicity, with the recombinant E. coli being rescued when 20845 was co-expressed with its cognate immunity protein. The 20845 effector was an 899 amino-acid protein, comprised of a GHH-nuclease domain in its N-terminus, a large central region of indeterminant function, and a C-terminus for secretion. Engineered variants of the 20845 gene that had mutations in the predicted catalytic site did not impede E. coli, indicating that the antibacterial effect of 20845 involves its nuclease activity. Using flow cytometry with DNA staining, we determined that 20845, but not its mutant variants, confers a loss in DNA content of target bacteria. Database searches revealed that uncharacterized homologs of 20845 occur within a range of bacteria. These data indicate that the S. maltophilia T4SS promotes interbacterial competition through the action of multiple toxic effectors, including a potent, novel DNase.IMPORTANCEStenotrophomonas maltophilia is a multi-drug-resistant, Gram-negative bacterium that is an emerging pathogen of humans. Patients with cystic fibrosis are particularly susceptible to S. maltophilia infection. In hospital water systems and various types of infections, S. maltophilia co-exists with other bacteria, including other pathogens such as Pseudomonas aeruginosa. We previously demonstrated that S. maltophilia has a functional VirB/D4 type VI protein secretion system (T4SS) that promotes contact-dependent killing of other bacteria. Since most work on antibacterial systems involves the type VI secretion system, this observation remains noteworthy. Moreover, S. maltophilia currently stands alone as a model for a human pathogen expressing an antibacterial T4SS. Using biochemical, genetic, and cell biological approaches, we now report both the discovery of a novel antibacterial nuclease (TfdA) and the first structural determination of a bactericidal T4SS effector (TfcB).


Sujet(s)
Protéines bactériennes , Stenotrophomonas maltophilia , Systèmes de sécrétion de type IV , Stenotrophomonas maltophilia/génétique , Stenotrophomonas maltophilia/enzymologie , Stenotrophomonas maltophilia/métabolisme , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/composition chimique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Pseudomonas aeruginosa/génétique , Pseudomonas aeruginosa/enzymologie , Pseudomonas aeruginosa/métabolisme , Escherichia coli/génétique , Escherichia coli/métabolisme , Conformation des protéines
9.
Nat Commun ; 15(1): 5498, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38944647

RÉSUMÉ

IncX3 plasmids carrying the New Delhi metallo-ß-lactamase-encoding gene, blaNDM-5, are rapidly spreading globally in both humans and animals. Given that carbapenems are listed on the WHO AWaRe watch group and are prohibited for use in animals, the drivers for the successful dissemination of Carbapenem-Resistant Enterobacterales (CRE) carrying blaNDM-5-IncX3 plasmids still remain unknown. We observe that E. coli carrying blaNDM-5-IncX3 can persist in chicken intestines either under the administration of amoxicillin, one of the largest veterinary ß-lactams used in livestock, or without any antibiotic pressure. We therefore characterise the blaNDM-5-IncX3 plasmid and identify a transcription regulator, VirBR, that binds to the promoter of the regulator gene actX enhancing the transcription of Type IV secretion systems (T4SS); thereby, promoting conjugation of IncX3 plasmids, increasing pili adhesion capacity and enhancing the colonisation of blaNDM-5-IncX3 transconjugants in animal digestive tracts. Our mechanistic and in-vivo studies identify VirBR as a major factor in the successful spread of blaNDM-5-IncX3 across one-health AMR sectors. Furthermore, VirBR enhances the plasmid conjugation and T4SS expression by the presence of copper and zinc ions, thereby having profound ramifications on the use of universal animal feeds.


Sujet(s)
Antibactériens , Poulets , Conjugaison génétique , Escherichia coli , Plasmides , bêta-Lactamases , Animaux , Plasmides/génétique , bêta-Lactamases/génétique , bêta-Lactamases/métabolisme , Poulets/microbiologie , Humains , Escherichia coli/génétique , Escherichia coli/effets des médicaments et des substances chimiques , Antibactériens/pharmacologie , Protéines Escherichia coli/génétique , Protéines Escherichia coli/métabolisme , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Amoxicilline/pharmacologie , Régions promotrices (génétique)/génétique , Infections à Escherichia coli/médecine vétérinaire , Infections à Escherichia coli/microbiologie , Infections à Escherichia coli/transmission , Régulation de l'expression des gènes bactériens/effets des médicaments et des substances chimiques , Intestins/microbiologie
10.
EMBO J ; 43(15): 3287-3306, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38886579

RÉSUMÉ

Conjugative type IV secretion systems (T4SS) mediate bacterial conjugation, a process that enables the unidirectional exchange of genetic materials between a donor and a recipient bacterial cell. Bacterial conjugation is the primary means by which antibiotic resistance genes spread among bacterial populations (Barlow 2009; Virolle et al, 2020). Conjugative T4SSs form pili: long extracellular filaments that connect with recipient cells. Previously, we solved the cryo-electron microscopy (cryo-EM) structure of a conjugative T4SS. In this article, based on additional data, we present a more complete T4SS cryo-EM structure than that published earlier. Novel structural features include details of the mismatch symmetry within the OMCC, the presence of a fourth VirB8 subunit in the asymmetric unit of both the arches and the inner membrane complex (IMC), and a hydrophobic VirB5 tip in the distal end of the stalk. Additionally, we provide previously undescribed structural insights into the protein VirB10 and identify a novel regulation mechanism of T4SS-mediated pilus biogenesis by this protein, that we believe is a key checkpoint for this process.


Sujet(s)
Cryomicroscopie électronique , Fimbriae bactériens , Systèmes de sécrétion de type IV , Fimbriae bactériens/métabolisme , Fimbriae bactériens/ultrastructure , Fimbriae bactériens/génétique , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/composition chimique , Modèles moléculaires , Conjugaison génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/composition chimique , Escherichia coli/métabolisme , Escherichia coli/génétique , Conformation des protéines
11.
Proc Natl Acad Sci U S A ; 121(20): e2310348121, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38709922

RÉSUMÉ

The evolutionary conserved YopJ family comprises numerous type-III-secretion system (T3SS) effectors of diverse mammalian and plant pathogens that acetylate host proteins to dampen immune responses. Acetylation is mediated by a central acetyltransferase domain that is flanked by conserved regulatory sequences, while a nonconserved N-terminal extension encodes the T3SS-specific translocation signal. Bartonella spp. are facultative-intracellular pathogens causing intraerythrocytic bacteremia in their mammalian reservoirs and diverse disease manifestations in incidentally infected humans. Bartonellae do not encode a T3SS, but most species possess a type-IV-secretion system (T4SS) to translocate Bartonella effector proteins (Beps) into host cells. Here we report that the YopJ homologs present in Bartonellae species represent genuine T4SS effectors. Like YopJ family T3SS effectors of mammalian pathogens, the "Bartonella YopJ-like effector A" (ByeA) of Bartonella taylorii also targets MAP kinase signaling to dampen proinflammatory responses, however, translocation depends on a functional T4SS. A split NanoLuc luciferase-based translocation assay identified sequences required for T4SS-dependent translocation in conserved regulatory regions at the C-terminus and proximal to the N-terminus of ByeA. The T3SS effectors YopP from Yersinia enterocolitica and AvrA from Salmonella Typhimurium were also translocated via the Bartonella T4SS, while ByeA was not translocated via the Yersinia T3SS. Our data suggest that YopJ family T3SS effectors may have evolved from an ancestral T4SS effector, such as ByeA of Bartonella. In this evolutionary scenario, the signal for T4SS-dependent translocation encoded by N- and C-terminal sequences remained functional in the derived T3SS effectors due to the essential role these sequences coincidentally play in regulating acetyltransferase activity.


Sujet(s)
Protéines bactériennes , Bartonella , Systèmes de sécrétion de type IV , Bartonella/métabolisme , Bartonella/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Humains , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/génétique , Transport des protéines , Animaux
12.
Life Sci Alliance ; 7(8)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38811160

RÉSUMÉ

A major pathway for horizontal gene transfer is the transmission of DNA from donor to recipient cells via plasmid-encoded type IV secretion systems (T4SSs). Many conjugative plasmids encode for a single-stranded DNA-binding protein (SSB) together with their T4SS. Some of these SSBs have been suggested to aid in establishing the plasmid in the recipient cell, but for many, their function remains unclear. Here, we characterize PrgE, a proposed SSB from the Enterococcus faecalis plasmid pCF10. We show that PrgE is not essential for conjugation. Structurally, it has the characteristic OB-fold of SSBs, but it has very unusual DNA-binding properties. Our DNA-bound structure shows that PrgE binds ssDNA like beads on a string supported by its N-terminal tail. In vitro studies highlight the plasticity of PrgE oligomerization and confirm the importance of the N-terminus. Unlike other SSBs, PrgE binds both double- and single-stranded DNA equally well. This shows that PrgE has a quaternary assembly and DNA-binding properties that are very different from the prototypical bacterial SSB, but also different from eukaryotic SSBs.


Sujet(s)
Protéines bactériennes , ADN simple brin , Protéines de liaison à l'ADN , Enterococcus faecalis , Plasmides , Plasmides/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/composition chimique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Enterococcus faecalis/génétique , Enterococcus faecalis/métabolisme , ADN simple brin/métabolisme , ADN simple brin/génétique , Liaison aux protéines , Conjugaison génétique/génétique , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Modèles moléculaires , Transfert horizontal de gène , ADN bactérien/génétique , ADN bactérien/métabolisme
13.
mBio ; 15(5): e0075923, 2024 May 08.
Article de Anglais | MEDLINE | ID: mdl-38564675

RÉSUMÉ

Recent metagenome-assembled genome (MAG) analyses have profoundly impacted Rickettsiology systematics. The discovery of basal lineages (novel families Mitibacteraceae and Athabascaceae) with predicted extracellular lifestyles exposed an evolutionary timepoint for the transition to host dependency, which seemingly occurred independent of mitochondrial evolution. Notably, these basal rickettsiae carry the Rickettsiales vir homolog (rvh) type IV secretion system and purportedly use rvh to kill congener microbes rather than parasitize host cells as described for later-evolving rickettsial pathogens. MAG analysis also substantially increased diversity for the genus Rickettsia and delineated a sister lineage (the novel genus Tisiphia) that stands to inform on the emergence of human pathogens from protist and invertebrate endosymbionts. Herein, we probed Rickettsiales MAG and genomic diversity for the distribution of Rickettsia rvh effectors to ascertain their origins. A sparse distribution of most Rickettsia rvh effectors outside of Rickettsiaceae lineages illuminates unique rvh evolution from basal extracellular species and other rickettsial families. Remarkably, nearly every effector was found in multiple divergent forms with variable architectures, indicating profound roles for gene duplication and recombination in shaping effector repertoires in Rickettsia pathogens. Lateral gene transfer plays a prominent role in shaping the rvh effector landscape, as evinced by the discovery of many effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchange between Rickettsia and Legionella species. Our study exemplifies how MAGs can yield insight into pathogen effector origins, particularly how effector architectures might become tailored to the discrete host cell functions of different eukaryotic hosts.IMPORTANCEWhile rickettsioses are deadly vector-borne human diseases, factors distinguishing Rickettsia pathogens from the innumerable bevy of environmental rickettsial endosymbionts remain lacking. Recent metagenome-assembled genome (MAG) studies revealed evolutionary timepoints for rickettsial transitions to host dependency. The rvh type IV secretion system was likely repurposed from congener killing in basal extracellular species to parasitizing host cells in later-evolving pathogens. Our analysis of MAG diversity for over two dozen rvh effectors unearthed their presence in some non-pathogens. However, most effectors were found in multiple divergent forms with variable architectures, indicating gene duplication and recombination-fashioned effector repertoires of Rickettsia pathogens. Lateral gene transfer substantially shaped pathogen effector arsenals, evinced by the discovery of effectors on plasmids and conjugative transposons, as well as pervasive effector gene exchanges between Rickettsia and Legionella species. Our study exemplifies how MAGs yield insight into pathogen effector origins and evolutionary processes tailoring effectors to eukaryotic host cell biology.


Sujet(s)
Génome bactérien , Métagénome , Phylogenèse , Rickettsia , Rickettsia/génétique , Rickettsia/classification , Évolution moléculaire , Rickettsiales/génétique , Rickettsiales/classification , Variation génétique , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Transfert horizontal de gène , Humains , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme
14.
Commun Biol ; 7(1): 499, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38664513

RÉSUMÉ

Bacterial cooperation and antagonism mediated by secretion systems are among the ways in which bacteria interact with one another. Here we report the discovery of an antagonistic property of a type IV secretion system (T4SS) sourced from a conjugative plasmid, RP4, using engineering approaches. We scrutinized the genetic determinants and suggested that this antagonistic activity is independent of molecular cargos, while we also elucidated the resistance genes. We further showed that a range of Gram-negative bacteria and a mixed bacterial population can be eliminated by this T4SS-dependent antagonism. Finally, we showed that such an antagonistic property is not limited to T4SS sourced from RP4, rather it can also be observed in a T4SS originated from another conjugative plasmid, namely R388. Our results are the first demonstration of conjugative T4SS-dependent antagonism between Gram-negative bacteria on the genetic level and provide the foundation for future mechanistic studies.


Sujet(s)
Conjugaison génétique , Plasmides , Systèmes de sécrétion de type IV , Plasmides/génétique , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Bactéries à Gram négatif/génétique , Bactéries à Gram négatif/métabolisme , Escherichia coli/génétique , Escherichia coli/métabolisme
15.
PLoS Genet ; 20(3): e1011088, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38437248

RÉSUMÉ

Bacterial type IV secretion systems (T4SSs) are a versatile family of macromolecular translocators, collectively able to recruit diverse DNA and protein substrates and deliver them to a wide range of cell types. Presently, there is little understanding of how T4SSs recognize substrate repertoires and form productive contacts with specific target cells. Although T4SSs are composed of a number of conserved subunits and adopt certain conserved structural features, they also display considerable compositional and structural diversity. Here, we explored the structural bases underlying the functional versatility of T4SSs through systematic deletion and subunit swapping between two conjugation systems encoded by the distantly-related IncF plasmids, pED208 and F. We identified several regions of intrinsic flexibility among the encoded T4SSs, as evidenced by partial or complete functionality of chimeric machines. Swapping of VirD4-like TraD type IV coupling proteins (T4CPs) yielded functional chimeras, indicative of relaxed specificity at the substrate-TraD and TraD-T4SS interfaces. Through mutational analyses, we further delineated domains of the TraD T4CPs contributing to recruitment of cognate vs heterologous DNA substrates. Remarkably, swaps of components comprising the outer membrane core complexes, a few F-specific subunits, or the TraA pilins supported DNA transfer in the absence of detectable pilus production. Among sequenced enterobacterial species in the NCBI database, we identified many strains that harbor two or more F-like plasmids and many F plasmids lacking one or more T4SS components required for self-transfer. We confirmed that host cells carrying co-resident, non-selftransmissible variants of pED208 and F elaborate chimeric T4SSs, as evidenced by transmission of both plasmids. We propose that T4SS plasticity enables the facile assembly of functional chimeras, and this intrinsic flexibility at the structural level can account for functional diversification of this superfamily over evolutionary time and, on a more immediate time-scale, to proliferation of transfer-defective MGEs in nature.


Sujet(s)
Facteur F , Systèmes de sécrétion de type IV , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/composition chimique , Systèmes de sécrétion de type IV/métabolisme , Protéines de fimbriae/génétique , Plasmides/génétique , ADN bactérien , Protéines bactériennes/métabolisme
16.
Biochem Cell Biol ; 102(3): 226-237, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38377487

RÉSUMÉ

We here describe the structure-based design of small molecule inhibitors of the type IV secretion system of Helicobacter pylori. The secretion system is encoded by the cag pathogenicity island, and we chose Cagα, a hexameric ATPase and member of the family of VirB11-like proteins, as target for inhibitor design. We first solved the crystal structure of Cagα in a complex with the previously identified small molecule inhibitor 1G2. The molecule binds at the interface between two Cagα subunits and mutagenesis of the binding site identified Cagα residues F39 and R73 as critical for 1G2 binding. Based on the inhibitor binding site we synthesized 98 small molecule derivates of 1G2 to improve binding of the inhibitor. We used the production of interleukin-8 of gastric cancer cells during H. pylori infection to screen the potency of inhibitors and we identified five molecules (1G2_1313, 1G2_1338, 1G2_2886, 1G2_2889, and 1G2_2902) that have similar or higher potency than 1G2. Differential scanning fluorimetry suggested that these five molecules bind Cagα, and enzyme assays demonstrated that some are more potent ATPase inhibitors than 1G2. Finally, scanning electron microscopy revealed that 1G2 and its derivatives inhibit the assembly of T4SS-determined extracellular pili suggesting a mechanism for their anti-virulence effect.


Sujet(s)
Adenosine triphosphatases , Protéines bactériennes , Helicobacter pylori , Helicobacter pylori/enzymologie , Humains , Adenosine triphosphatases/antagonistes et inhibiteurs , Adenosine triphosphatases/métabolisme , Adenosine triphosphatases/composition chimique , Protéines bactériennes/antagonistes et inhibiteurs , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique , Systèmes de sécrétion de type IV/métabolisme , Systèmes de sécrétion de type IV/composition chimique , Systèmes de sécrétion de type IV/antagonistes et inhibiteurs , Conception de médicament , Infections à Helicobacter/traitement médicamenteux , Infections à Helicobacter/microbiologie , Cristallographie aux rayons X , Antienzymes/pharmacologie , Antienzymes/composition chimique , Modèles moléculaires , Sites de fixation , Relation structure-activité , Lignée cellulaire tumorale , Interleukine-8/métabolisme
17.
EMBO Rep ; 25(3): 1436-1452, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38332152

RÉSUMÉ

Many bacteria kill rival species by translocating toxic effectors into target cells. Effectors are often encoded along with cognate immunity proteins that could (i) protect against "friendly-fire" (trans-intoxication) from neighboring sister cells and/or (ii) protect against internal cis-intoxication (suicide). Here, we distinguish between these two mechanisms in the case of the bactericidal Xanthomonas citri Type IV Secretion System (X-T4SS). We use a set of X. citri mutants lacking multiple effector/immunity protein (X-Tfe/X-Tfi) pairs to show that X-Tfis are not absolutely required to protect against trans-intoxication by wild-type cells. Our investigation then focused on the in vivo function of the lysozyme-like effector X-TfeXAC2609 and its cognate immunity protein X-TfiXAC2610. In the absence of X-TfiXAC2610, we observe X-TfeXAC2609-dependent and X-T4SS-independent accumulation of damage in the X. citri cell envelope, cell death, and inhibition of biofilm formation. While immunity proteins in other systems have been shown to protect against attacks by sister cells (trans-intoxication), this is an example of an antibacterial secretion system in which the immunity proteins are dedicated to protecting cells against cis-intoxication.


Sujet(s)
Protéines bactériennes , Xanthomonas , Humains , Protéines bactériennes/métabolisme , Xanthomonas/métabolisme , Systèmes de sécrétion de type IV/métabolisme , Antibactériens/métabolisme
18.
Can J Microbiol ; 70(4): 119-127, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38176008

RÉSUMÉ

Helicobacter pylori resistance to antibiotics is a growing problem and it increasingly leads to treatment failure. While the bacterium is present worldwide, the severity of clinical outcomes is highly dependent on the geographical origin and genetic characteristics of the strains. One of the major virulence factors identified in H. pylori is the cag pathogenicity island (cagPAI), which encodes a type IV secretion system (T4SS) used to translocate effectors into human cells. Here, we investigated the genetic variability of the cagPAI among 13 antibiotic-resistant H. pylori strains that were isolated from patient biopsies in Québec. Seven of the clinical strains carried the cagPAI, but only four could be readily cultivated under laboratory conditions. We observed variability of the sequences of CagA and CagL proteins that are encoded by the cagPAI. All clinical isolates induce interleukin-8 secretion and morphological changes upon co-incubation with gastric cancer cells and two of them produce extracellular T4SS pili. Finally, we demonstrate that molecule 1G2, a small molecule inhibitor of the Cagα protein from the model strain H. pylori 26695, reduces interleukin-8 secretion in one of the clinical isolates. Co-incubation with 1G2 also inhibits the assembly of T4SS pili, suggesting a mechanism for its action on T4SS function.


Sujet(s)
Infections à Helicobacter , Helicobacter pylori , Humains , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Antigènes bactériens/génétique , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Helicobacter pylori/génétique , Helicobacter pylori/métabolisme , Interleukine-8/métabolisme , Infections à Helicobacter/microbiologie
19.
Front Cell Infect Microbiol ; 13: 1255852, 2023.
Article de Anglais | MEDLINE | ID: mdl-38089815

RÉSUMÉ

Many pathogens use Type III and Type IV protein secretion systems to secrete virulence factors from the bacterial cytosol into host cells. These systems operate through a one-step mechanism. The secreted substrates (protein or nucleo-protein complexes in the case of Type IV conjugative systems) are guided to the base of the secretion channel, where they are directly delivered into the host cell in an ATP-dependent unfolded state. Despite the numerous disparities between these secretion systems, here we have focused on the structural and functional similarities between both systems. In particular, on the structural similarity shared by one of the main ATPases (EscN and VirD4 in Type III and Type IV secretion systems, respectively). Interestingly, these ATPases also exhibit a structural resemblance to F1-ATPases, which suggests a common mechanism for substrate secretion. The correlation between structure and function of essential components in both systems can provide significant insights into the molecular mechanisms involved. This approach is of great interest in the pursuit of identifying inhibitors that can effectively target these systems.


Sujet(s)
Protéines bactériennes , Systèmes de sécrétion de type IV , Systèmes de sécrétion de type IV/métabolisme , Protéines bactériennes/métabolisme , Bactéries/métabolisme , Transport des protéines , Adenosine triphosphatases , Systèmes de sécrétion de type III/métabolisme
20.
J Microbiol Biotechnol ; 33(12): 1543-1551, 2023 Dec 28.
Article de Anglais | MEDLINE | ID: mdl-37528551

RÉSUMÉ

The recently published high-resolution R388 T4SS structure provides exciting new details about the complete complex of T4SS, including the components making up the stalk and arches, numerous symmetry mismatches between regions of the complex, and an intriguing interpretation of the closed stalk and radial symmetry of the inner membrane complex, which is related to pilus biogenesis assembly. However, there are a few unidentified densities in the electron microscopy map and portions of the identified component sequences for which the structure is not yet known. It is also unclear how well this minimized DNA-transporting T4SS predicts the structure of other T4SSs, such as expanded systems and those that transport proteins rather than DNA. In this review, we evaluate what can be inferred from the recent high-resolution structure of the R388 T4SS with respect to the Cag and Dot/Icm systems. These systems were selected because, given what is currently known about these systems, we expect them to present most structural differences compared to the R388 T4SS structure. Furthermore, we discuss bacterial physiology and diversity, the T4SS structures and their variations between different bacterial species. These insights may prove beneficial for researchers who elucidate the structure and functions of T4SS in different bacterial species.


Sujet(s)
ADN , Systèmes de sécrétion de type IV , Systèmes de sécrétion de type IV/composition chimique , Systèmes de sécrétion de type IV/génétique , Systèmes de sécrétion de type IV/métabolisme , Transport biologique , Protéines bactériennes/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE