Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Annu Rev Biochem ; 84: 603-29, 2015.
Article in English | MEDLINE | ID: mdl-26034891

ABSTRACT

Type IV secretion systems (T4SSs) are large multisubunit translocons, found in both gram-negative and gram-positive bacteria and in some archaea. These systems transport a diverse array of substrates from DNA and protein-DNA complexes to proteins, and play fundamental roles in both bacterial pathogenesis and bacterial adaptation to the cellular milieu in which bacteria live. This review describes the various biochemical and structural advances made toward understanding the biogenesis, architecture, and function of T4SSs.


Subject(s)
Bacteria/metabolism , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/ultrastructure , Bacteria/chemistry , Bacteria/classification , F Factor/genetics , Microscopy, Electron
2.
EMBO J ; 43(15): 3287-3306, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38886579

ABSTRACT

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.


Subject(s)
Cryoelectron Microscopy , Fimbriae, Bacterial , Type IV Secretion Systems , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Fimbriae, Bacterial/genetics , Type IV Secretion Systems/metabolism , Type IV Secretion Systems/genetics , Type IV Secretion Systems/chemistry , Models, Molecular , Conjugation, Genetic , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Escherichia coli/metabolism , Escherichia coli/genetics , Protein Conformation
3.
Nature ; 607(7917): 191-196, 2022 07.
Article in English | MEDLINE | ID: mdl-35732732

ABSTRACT

Bacterial conjugation is the fundamental process of unidirectional transfer of DNAs, often plasmid DNAs, from a donor cell to a recipient cell1. It is the primary means by which antibiotic resistance genes spread among bacterial populations2,3. In Gram-negative bacteria, conjugation is mediated by a large transport apparatus-the conjugative type IV secretion system (T4SS)-produced by the donor cell and embedded in both its outer and inner membranes. The T4SS also elaborates a long extracellular filament-the conjugative pilus-that is essential for DNA transfer4,5. Here we present a high-resolution cryo-electron microscopy (cryo-EM) structure of a 2.8 megadalton T4SS complex composed of 92 polypeptides representing 8 of the 10 essential T4SS components involved in pilus biogenesis. We added the two remaining components to the structural model using co-evolution analysis of protein interfaces, to enable the reconstitution of the entire system including the pilus. This structure describes the exceptionally large protein-protein interaction network required to assemble the many components that constitute a T4SS and provides insights on the unique mechanism by which they elaborate pili.


Subject(s)
Bacterial Proteins , Cryoelectron Microscopy , Type IV Secretion Systems , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Conjugation, Genetic , DNA/genetics , Evolution, Molecular , Fimbriae, Bacterial/metabolism , Plasmids/genetics , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/metabolism , Type IV Secretion Systems/ultrastructure
4.
PLoS Genet ; 20(3): e1011088, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38437248

ABSTRACT

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.


Subject(s)
F Factor , Type IV Secretion Systems , Type IV Secretion Systems/genetics , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/metabolism , Fimbriae Proteins/genetics , Plasmids/genetics , DNA, Bacterial , Bacterial Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34983846

ABSTRACT

Many soil-, water-, and plant-associated bacterial species from the orders Xanthomonadales, Burkholderales, and Neisseriales carry a type IV secretion system (T4SS) specialized in translocating effector proteins into other gram-negative species, leading to target cell death. These effectors, known as X-Tfes, carry a carboxyl-terminal domain of ∼120 residues, termed XVIPCD, characterized by several conserved motifs and a glutamine-rich tail. Previous studies showed that the XVIPCD is required for interaction with the T4SS coupling protein VirD4 and for T4SS-dependent translocation. However, the structural basis of the XVIPCD-VirD4 interaction is unknown. Here, we show that the XVIPCD interacts with the central all-alpha domain of VirD4 (VirD4AAD). We used solution NMR spectroscopy to solve the structure of the XVIPCD of X-TfeXAC2609 from Xanthomonas citri and to map its interaction surface with VirD4AAD Isothermal titration calorimetry and in vivo Xanthomonas citri versus Escherichia coli competition assays using wild-type and mutant X-TfeXAC2609 and X-TfeXAC3634 indicate that XVIPCDs can be divided into two regions with distinct functions: the well-folded N-terminal region contains specific conserved motifs that are responsible for interactions with VirD4AAD, while both N- and carboxyl-terminal regions are required for effective X-Tfe translocation into the target cell. The conformational stability of the N-terminal region is reduced at and below pH 7.0, a property that may facilitate X-Tfe unfolding and translocation through the more acidic environment of the periplasm.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Escherichia coli/chemistry , Type IV Secretion Systems/antagonists & inhibitors , Type IV Secretion Systems/chemistry , Xanthomonas/chemistry , Bacterial Proteins/genetics , Escherichia coli/genetics , Models, Molecular , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Domains , Structure-Activity Relationship , Type IV Secretion Systems/genetics , Xanthomonas/genetics
6.
Biochem Cell Biol ; 102(3): 226-237, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38377487

ABSTRACT

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.


Subject(s)
Adenosine Triphosphatases , Bacterial Proteins , Helicobacter pylori , Helicobacter pylori/enzymology , Humans , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Type IV Secretion Systems/metabolism , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/antagonists & inhibitors , Drug Design , Helicobacter Infections/drug therapy , Helicobacter Infections/microbiology , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Models, Molecular , Binding Sites , Structure-Activity Relationship , Cell Line, Tumor , Interleukin-8/metabolism
7.
Mol Microbiol ; 115(3): 436-452, 2021 03.
Article in English | MEDLINE | ID: mdl-33326642

ABSTRACT

Bacterial type IV secretion systems (T4SSs) are a functionally diverse translocation superfamily. They consist mainly of two large subfamilies: (i) conjugation systems that mediate interbacterial DNA transfer and (ii) effector translocators that deliver effector macromolecules into prokaryotic or eukaryotic cells. A few other T4SSs export DNA or proteins to the milieu, or import exogenous DNA. The T4SSs are defined by 6 or 12 conserved "core" subunits that respectively elaborate "minimized" systems in Gram-positive or -negative bacteria. However, many "expanded" T4SSs are built from "core" subunits plus numerous others that are system-specific, which presumptively broadens functional capabilities. Recently, there has been exciting progress in defining T4SS assembly pathways and architectures using a combination of fluorescence and cryoelectron microscopy. This review will highlight advances in our knowledge of structure-function relationships for model Gram-negative bacterial T4SSs, including "minimized" systems resembling the Agrobacterium tumefaciens VirB/VirD4 T4SS and "expanded" systems represented by the Helicobacter pylori Cag, Legionella pneumophila Dot/Icm, and F plasmid-encoded Tra T4SSs. Detailed studies of these model systems are generating new insights, some at atomic resolution, to long-standing questions concerning mechanisms of substrate recruitment, T4SS channel architecture, conjugative pilus assembly, and machine adaptations contributing to T4SS functional versatility.


Subject(s)
Conjugation, Genetic , Fimbriae, Bacterial/physiology , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/physiology , Protein Translocation Systems/metabolism , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/physiology , Agrobacterium tumefaciens/chemistry , Agrobacterium tumefaciens/physiology , Amino Acid Motifs , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/physiology , Cryoelectron Microscopy , Gram-Negative Bacteria/ultrastructure , Gram-Negative Bacterial Infections/microbiology , Helicobacter pylori/chemistry , Helicobacter pylori/physiology , Humans , Legionella pneumophila/chemistry , Legionella pneumophila/physiology , Molecular Docking Simulation , Protein Translocation Systems/chemistry , Protein Translocation Systems/ultrastructure , Structure-Activity Relationship , Type IV Secretion Systems/ultrastructure
8.
PLoS Pathog ; 15(9): e1007651, 2019 09.
Article in English | MEDLINE | ID: mdl-31513674

ABSTRACT

Bacterial type IV secretion systems (T4SS) are a highly diversified but evolutionarily related family of macromolecule transporters that can secrete proteins and DNA into the extracellular medium or into target cells. It was recently shown that a subtype of T4SS harboured by the plant pathogen Xanthomonas citri transfers toxins into target cells. Here, we show that a similar T4SS from the multi-drug-resistant opportunistic pathogen Stenotrophomonas maltophilia is proficient in killing competitor bacterial species. T4SS-dependent duelling between S. maltophilia and X. citri was observed by time-lapse fluorescence microscopy. A bioinformatic search of the S. maltophilia K279a genome for proteins containing a C-terminal domain conserved in X. citri T4SS effectors (XVIPCD) identified twelve putative effectors and their cognate immunity proteins. We selected a putative S. maltophilia effector with unknown function (Smlt3024) for further characterization and confirmed that it is indeed secreted in a T4SS-dependent manner. Expression of Smlt3024 in the periplasm of E. coli or its contact-dependent delivery via T4SS into E. coli by X. citri resulted in reduced growth rates, which could be counteracted by expression of its cognate inhibitor Smlt3025 in the target cell. Furthermore, expression of the VirD4 coupling protein of X. citri can restore the function of S. maltophilia ΔvirD4, demonstrating that effectors from one species can be recognized for transfer by T4SSs from another species. Interestingly, Smlt3024 is homologous to the N-terminal domain of large Ca2+-binding RTX proteins and the crystal structure of Smlt3025 revealed a topology similar to the iron-regulated protein FrpD from Neisseria meningitidis which has been shown to interact with the RTX protein FrpC. This work expands our current knowledge about the function of bacteria-killing T4SSs and increases the panel of effectors known to be involved in T4SS-mediated interbacterial competition, which possibly contribute to the establishment of S. maltophilia in clinical and environmental settings.


Subject(s)
Bacterial Proteins/physiology , Stenotrophomonas maltophilia/physiology , Stenotrophomonas maltophilia/pathogenicity , Type IV Secretion Systems/physiology , Amino Acid Sequence , Antibiosis/genetics , Antibiosis/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Conserved Sequence , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/growth & development , Genes, Bacterial , Gram-Negative Bacterial Infections/microbiology , Humans , Iron-Regulatory Proteins/chemistry , Iron-Regulatory Proteins/genetics , Iron-Regulatory Proteins/physiology , Models, Molecular , Opportunistic Infections/microbiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity , Stenotrophomonas maltophilia/genetics , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/genetics , Xanthomonas/genetics , Xanthomonas/growth & development
9.
EMBO Rep ; 20(2)2019 02.
Article in English | MEDLINE | ID: mdl-30602585

ABSTRACT

Conjugation is the process by which bacteria exchange genetic materials in a unidirectional manner from a donor cell to a recipient cell. The discovery of conjugation signalled the dawn of genetics and molecular biology. In Gram-negative bacteria, the process of conjugation is mediated by a large membrane-embedded machinery termed "conjugative type IV secretion (T4S) system", a large injection nanomachine, which together with a DNA-processing machinery termed "the relaxosome" and a large extracellular tube termed "pilus" orchestrates directional DNA transfer. Here, the focus is on past and latest research in the field of conjugation and T4S systems in Gram-negative bacteria, with an emphasis on the various questions and debates that permeate the field from a mechanistic perspective.


Subject(s)
Conjugation, Genetic , Gram-Negative Bacteria/physiology , Gram-Negative Bacterial Infections/microbiology , Type IV Secretion Systems/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Biological Transport , Fimbriae, Bacterial/metabolism , Humans , Models, Biological , Models, Molecular , Multiprotein Complexes , Protein Conformation , Structure-Activity Relationship , Type IV Secretion Systems/chemistry
10.
Molecules ; 26(9)2021 Apr 24.
Article in English | MEDLINE | ID: mdl-33923273

ABSTRACT

Many gram-negative bacteria use type IV secretion systems to deliver effector molecules to a wide range of target cells. These substrate proteins, which are called type IV secreted effectors (T4SE), manipulate host cell processes during infection, often resulting in severe diseases or even death of the host. Therefore, identification of putative T4SEs has become a very active research topic in bioinformatics due to its vital roles in understanding host-pathogen interactions. PSI-BLAST profiles have been experimentally validated to provide important and discriminatory evolutionary information for various protein classification tasks. In the present study, an accurate computational predictor termed iT4SE-EP was developed for identifying T4SEs by extracting evolutionary features from the position-specific scoring matrix and the position-specific frequency matrix profiles. First, four types of encoding strategies were designed to transform protein sequences into fixed-length feature vectors based on the two profiles. Then, the feature selection technique based on the random forest algorithm was utilized to reduce redundant or irrelevant features without much loss of information. Finally, the optimal features were input into a support vector machine classifier to carry out the prediction of T4SEs. Our experimental results demonstrated that iT4SE-EP outperformed most of existing methods based on the independent dataset test.


Subject(s)
Evolution, Molecular , Gram-Negative Bacteria/genetics , Host-Pathogen Interactions/genetics , Type IV Secretion Systems/genetics , Amino Acid Sequence/genetics , Bacterial Infections/drug therapy , Bacterial Infections/genetics , Bacterial Infections/microbiology , Computational Biology , Gram-Negative Bacteria/pathogenicity , Humans , Type IV Secretion Systems/chemistry
11.
J Biol Chem ; 294(13): 4806-4814, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30692196

ABSTRACT

Mycobacteria use type VII secretion systems to secrete proteins across their highly hydrophobic diderm cell envelope. Pathogenic mycobacteria, such as Mycobacterium tuberculosis and Mycobacterium marinum, have up to five of these systems, named ESX-1 to ESX-5. Most of these systems contain a set of five conserved membrane components, of which the four Ecc proteins form the core membrane-embedded secretion complex. The fifth conserved membrane protein, mycosin protease (MycP), is not part of the core complex but is essential for secretion, as it stabilizes this membrane complex. Here we investigated which MycP domains are required for this stabilization by producing hybrid constructs between MycP1 and MycP5 in M. marinum and analyzed their effect on ESX-1 and ESX-5 secretion. We found that both the protease and transmembrane domain are required for the ESX system-specific function of mycosins. In addition, we observed that the transmembrane domain strongly affects MycP protein levels. We also show that the extended loops 1 and 2 in the protease domain are probably primarily involved in MycP stability, whereas loop 3 and the MycP5-specific loop 5 are dispensable. The atypical propeptide, or N-terminal extension, is required only for MycP stability. Finally, we show that the protease domain of MycPP1, encoded by the esx-P1 locus on the pRAW plasmid, is functionally redundant to the protease domain of MycP5 These results provide the first insight into the regions of mycosins involved in interaction with and stabilization of their respective ESX complexes.


Subject(s)
Bacterial Proteins , Mycobacterium marinum , Mycobacterium tuberculosis , Subtilisins , Type IV Secretion Systems , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mycobacterium marinum/enzymology , Mycobacterium marinum/genetics , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Protein Domains , Protein Structure, Secondary , Subtilisins/chemistry , Subtilisins/genetics , Subtilisins/metabolism , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
12.
Proc Natl Acad Sci U S A ; 114(51): 13543-13548, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29203674

ABSTRACT

The type IVb secretion system (T4BSS) of Legionella pneumophila is a multiple-component apparatus that delivers ∼300 virulent effector proteins into host cells. The injected effectors modulate host cellular processes to promote bacterial infection and proliferation. IcmS and IcmW are two conserved small, acidic adaptor proteins that form a binary complex to interact with many effectors and facilitate their translocation. IcmS and IcmW can also interact with DotL, an ATPase of the type IV coupling protein complex (T4CP). However, how IcmS-IcmW recognizes effectors, and what the roles of IcmS-IcmW are in T4BSSs are unclear. In this study, we found that IcmS and IcmW form a 1:1 heterodimeric complex to bind effector substrates. Both IcmS and IcmW adopt new structural folds and have no structural similarities with known effector chaperones. IcmS has a compact global structure with an α/ß fold, while IcmW adopts a fully α-folded, relatively loose architecture. IcmS stabilizes IcmW by binding to its two C-terminal α-helices. Photocrosslinking assays revealed that the IcmS-IcmW complex binds its cognate effectors via an extended hydrophobic surface, which can also interact with the C terminus of DotL. A crystal structure of the DotL-IcmS-IcmW complex reveals extensive and highly stable interactions between DotL and IcmS-IcmW. Moreover, IcmS-IcmW recruits LvgA to DotL and assembles a unique T4CP. These data suggest that IcmS-IcmW also functions as an inseparable integral component of the DotL-T4CP complex in the bacterial inner membrane. This study provides molecular insights into the dual roles of the IcmS-IcmW complex in T4BSSs.


Subject(s)
Molecular Chaperones/chemistry , Type IV Secretion Systems/chemistry , Bacterial Proteins/metabolism , Binding Sites , Legionella pneumophila/chemistry , Legionella pneumophila/metabolism , Molecular Chaperones/metabolism , Protein Binding , Type IV Secretion Systems/metabolism , Virulence Factors/metabolism
13.
J Biol Chem ; 293(35): 13415-13426, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29976757

ABSTRACT

Many bacterial pathogens employ multicomponent protein complexes such as type IV secretion systems (T4SSs) to transfer virulence factors into host cells. Here we studied the interaction between two essential T4SS components: the very hydrophobic inner membrane protein VirB6, which may be a component of the translocation channel, and VirB10, which links the inner and outer bacterial membranes. To map the interaction site between these two T4SS components, we conducted alanine scanning and deleted six-amino acid stretches from the N-terminal periplasmic domain of VirB6 from Brucella suis Using the bacterial two-hybrid system to analyze the effects of these alterations on the VirB6-VirB10 interaction, we identified the amino acid regions 16-21 and 28-33 and Leu-18 in VirB6 as being required for this interaction. SDS-PAGE coupled with Western blotting of cell lysates and native PAGE of detergent-extracted membrane proteins revealed that the corresponding VirB6 residues in Agrobacterium tumefaciens (Phe-20 and amino acids 18-23 and 30-35) modulate the stability of both VirB6 and VirB5. However, the results from immuno-EM and super-resolution microscopy suggested that these regions and residues are not required for membrane association or for polar localization of VirB6. The six-amino acid deletions in the N terminus of VirB6 abolished pilus formation and virulence of A. tumefaciens, and the corresponding deletions in the VirB6 homolog TraD from the plasmid pKM101-T4SS abrogated plasmid transfer. Our results indicate that specific residues of the VirB6 N-terminal domain are required for VirB6 stabilization, its interaction with VirB10, and the incorporation of VirB2 and VirB5 into T-pili.


Subject(s)
Agrobacterium tumefaciens/metabolism , Bacterial Proteins/metabolism , Plant Diseases/microbiology , Protein Interaction Maps , Type IV Secretion Systems/metabolism , Agrobacterium tumefaciens/chemistry , Amino Acid Sequence , Bacterial Proteins/chemistry , Brucella suis/chemistry , Brucella suis/metabolism , Protein Interaction Domains and Motifs , Protein Stability , Sequence Alignment , Type IV Secretion Systems/chemistry
14.
Mol Microbiol ; 107(4): 455-471, 2018 02.
Article in English | MEDLINE | ID: mdl-29235173

ABSTRACT

Type IV secretion systems (T4SSs) are versatile multiprotein nanomachines spanning the entire cell envelope in Gram-negative and Gram-positive bacteria. They play important roles through the contact-dependent secretion of effector molecules into eukaryotic hosts and conjugative transfer of mobile DNA elements as well as contact-independent exchange of DNA with the extracellular milieu. In the last few years, many details on the molecular mechanisms of T4SSs have been elucidated. Exciting structures of T4SS complexes from Escherichia coli plasmids R388 and pKM101, Helicobacter pylori and Legionella pneumophila have been solved. The structure of the F-pilus was also reported and surprisingly revealed a filament composed of pilin subunits in 1:1 stoichiometry with phospholipid molecules. Many new T4SSs have been identified and characterized, underscoring the structural and functional diversity of this secretion superfamily. Complex regulatory circuits also have been shown to control T4SS machine production in response to host cell physiological status or a quorum of bacterial recipient cells in the vicinity. Here, we summarize recent advances in our knowledge of 'paradigmatic' and emerging systems, and further explore how new basic insights are aiding in the design of strategies aimed at suppressing T4SS functions in bacterial infections and spread of antimicrobial resistances.


Subject(s)
Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/metabolism , Animals , DNA, Bacterial/metabolism , DNA, Single-Stranded/metabolism , Gram-Negative Bacteria/genetics , Gram-Positive Bacteria/genetics , Host-Pathogen Interactions , Humans , Plasmids
15.
Curr Top Microbiol Immunol ; 418: 233-260, 2018.
Article in English | MEDLINE | ID: mdl-29808338

ABSTRACT

The Agrobacterium tumefaciens VirB/VirD4 translocation machine is a member of a superfamily of translocators designated as type IV secretion systems (T4SSs) that function in many species of gram-negative and gram-positive bacteria. T4SSs evolved from ancestral conjugation systems for specialized purposes relating to bacterial colonization or infection. A. tumefaciens employs the VirB/VirD4 T4SS to deliver oncogenic DNA (T-DNA) and effector proteins to plant cells, causing the tumorous disease called crown gall. This T4SS elaborates both a cell-envelope-spanning channel and an extracellular pilus for establishing target cell contacts. Recent mechanistic and structural studies of the VirB/VirD4 T4SS and related conjugation systems in Escherichia coli have defined T4SS architectures, bases for substrate recruitment, the translocation route for DNA substrates, and steps in the pilus biogenesis pathway. In this review, we provide a brief history of A. tumefaciens VirB/VirD4 T4SS from its discovery in the 1980s to its current status as a paradigm for the T4SS superfamily. We discuss key advancements in defining VirB/VirD4 T4SS function and structure, and we highlight the power of in vivo mutational analyses and chimeric systems for identifying mechanistic themes and specialized adaptations of this fascinating nanomachine.


Subject(s)
Agrobacterium tumefaciens/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mutagenesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/genetics , Agrobacterium tumefaciens/chemistry , Agrobacterium tumefaciens/metabolism , Agrobacterium tumefaciens/pathogenicity , Bacterial Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Type IV Secretion Systems/metabolism
16.
EMBO Rep ; 18(5): 726-732, 2017 05.
Article in English | MEDLINE | ID: mdl-28336774

ABSTRACT

Type IV secretion systems (T4SSs) are large macromolecular machines that translocate protein and DNA and are involved in the pathogenesis of multiple human diseases. Here, using electron cryotomography (ECT), we report the in situ structure of the Dot/Icm type IVB secretion system (T4BSS) utilized by the human pathogen Legionella pneumophila This is the first structure of a type IVB secretion system, and also the first structure of any T4SS in situ While the Dot/Icm system shares almost no sequence similarity with type IVA secretion systems (T4ASSs), its overall structure is seen here to be remarkably similar to previously reported T4ASS structures (those encoded by the R388 plasmid in Escherichia coli and the cag pathogenicity island in Helicobacter pylori). This structural similarity suggests shared aspects of mechanism. However, compared to the negative-stain reconstruction of the purified T4ASS from the R388 plasmid, the L. pneumophila Dot/Icm system is approximately twice as long and wide and exhibits several additional large densities, reflecting type-specific elaborations and potentially better structural preservation in situ.


Subject(s)
Electron Microscope Tomography/methods , Legionella pneumophila/chemistry , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Gene Expression Regulation, Bacterial , Legionella pneumophila/genetics , Legionella pneumophila/pathogenicity , Legionella pneumophila/ultrastructure , Plasmids
17.
Curr Top Microbiol Immunol ; 413: 31-60, 2017.
Article in English | MEDLINE | ID: mdl-29536354

ABSTRACT

Type IV secretion systems (T4SSs) are nanomachines that Gram-negative, Gram-positive bacteria, and some archaea use to transport macromolecules across their membranes into bacterial or eukaryotic host targets or into the extracellular milieu. They are the most versatile secretion systems, being able to deliver both proteins and nucleoprotein complexes into targeted cells. By mediating conjugation and/or competence, T4SSs play important roles in determining bacterial genome plasticity and diversity; they also play a pivotal role in the spread of antibiotic resistance within bacterial populations. T4SSs are also used by human pathogens such as Legionella pneumophila, Bordetella pertussis, Brucella sp., or Helicobacter pylori to sustain infection. Since they are essential virulence factors for these important pathogens, T4SSs might represent attractive targets for vaccines and therapeutics. The best-characterized conjugative T4SSs of Gram-negative bacteria are composed of twelve components that are conserved across many T4SSs. In this chapter, we will review our current structural knowledge on the T4SSs by describing the structures of the individual components and how they assemble into large macromolecular assemblies. With the combined efforts of X-ray crystallography, nuclear magnetic resonance (NMR), and more recently electron microscopy, structural biology of the T4SS has made spectacular progress during the past fifteen years and has unraveled the properties of unique proteins and complexes that assemble dynamically in a highly sophisticated manner.


Subject(s)
Type IV Secretion Systems/chemistry , Type IV Secretion Systems/metabolism , Bacterial Proteins , Genome, Bacterial , Gram-Negative Bacteria , Gram-Positive Bacteria , Humans
18.
J Infect Dis ; 215(10): 1580-1589, 2017 05 15.
Article in English | MEDLINE | ID: mdl-27703037

ABSTRACT

Coxiella burnetii is a gram-negative bacterium that causes acute and chronic Q fever. Because of the severe adverse effect of whole-cell vaccination, identification of immunodominant antigens of C. burnetii has become a major focus of Q fever vaccine development. We hypothesized that secreted C. burnetii type IV secretion system (T4SS) effectors may represent a major class of CD8+ T-cell antigens, owing to their cytosolic localization. Twenty-nine peptides were identified that elicited robust CD8+ T-cell interferon γ (IFN-γ) recall responses from mice infected with C. burnetii. Interestingly, 22 of 29 epitopes were derived from 17 T4SS-related proteins, none of which were identified as immunodominant antigens by using previous antibody-guided approaches. These epitopes were expressed in an attenuated Listeria monocytogenes vaccine strain. Immunization with recombinant L. monocytogenes vaccines induced a robust CD8+ T-cell response and conferred measurable protection against C. burnetii infection in mice. These data suggested that T4SS effectors represent an important class of C. burnetii antigens that can induce CD8+ T-cell responses. We also showed that attenuated L. monocytogenes vaccine vectors are an efficient antigen-delivery platform that can be used to induce robust protective CD8+ T-cell immune responses against C. burnetii infection.


Subject(s)
Antigen Presentation/immunology , Bacterial Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Coxiella burnetii/immunology , Epitopes, T-Lymphocyte/immunology , Vaccines, Attenuated/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Vaccines/chemistry , Coxiella burnetii/chemistry , Epitopes, T-Lymphocyte/chemistry , Female , Interferon-gamma Release Tests , Listeria monocytogenes/immunology , Mice , Mice, Inbred C57BL , Peptide Fragments/chemistry , Peptide Fragments/immunology , Q Fever/immunology , Q Fever/microbiology , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/immunology , Vaccines, Attenuated/chemistry
19.
J Biol Chem ; 291(45): 23817-23829, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27634044

ABSTRACT

Gram-negative bacteria use type IV secretion systems (T4SSs) for a variety of macromolecular transport processes that include the exchange of genetic material. The pKM101 plasmid encodes a T4SS similar to the well-studied model systems from Agrobacterium tumefaciens and Brucella suis Here, we studied the structure and function of TraE, a homolog of VirB8 that is an essential component of all T4SSs. Analysis by X-ray crystallography revealed a structure that is similar to other VirB8 homologs but displayed an altered dimerization interface. The dimerization interface observed in the X-ray structure was corroborated using the bacterial two-hybrid assay, biochemical characterization of the purified protein, and in vivo complementation, demonstrating that there are different modes of dimerization among VirB8 homologs. Analysis of interactions using the bacterial two-hybrid and cross-linking assays showed that TraE and its homologs from Agrobacterium, Brucella, and Helicobacter pylori form heterodimers. They also interact with heterologous VirB10 proteins, indicating a significant degree of plasticity in the protein-protein interactions of VirB8-like proteins. To further assess common features of VirB8-like proteins, we tested a series of small molecules derived from inhibitors of Brucella VirB8 dimerization. These molecules bound to TraE in vitro, docking predicted that they bind to a structurally conserved surface groove of the protein, and some of them inhibited pKM101 plasmid transfer. VirB8-like proteins thus share functionally important sites, and these can be exploited for the design of specific inhibitors of T4SS function.


Subject(s)
Bacterial Proteins/chemistry , Gram-Negative Bacteria/chemistry , Plasmids/chemistry , Type IV Secretion Systems/chemistry , Agrobacterium tumefaciens/chemistry , Agrobacterium tumefaciens/metabolism , Amino Acid Sequence , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Brucella suis/chemistry , Brucella suis/metabolism , Crystallography, X-Ray , Gram-Negative Bacteria/metabolism , Helicobacter pylori/chemistry , Helicobacter pylori/metabolism , Models, Molecular , Plasmids/antagonists & inhibitors , Plasmids/metabolism , Protein Conformation , Protein Interaction Maps , Protein Multimerization , Small Molecule Libraries/pharmacology , Type IV Secretion Systems/antagonists & inhibitors , Type IV Secretion Systems/metabolism
20.
Biochem Biophys Res Commun ; 465(4): 817-24, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26315269

ABSTRACT

The SET domain of LegAS4, a type IV secretion system effector of Legionella pneumophila, is a eukaryotic protein motif involved in histone methylation and epigenetic modulation. The SET domain of LegAS4 is involved in the modification of Lys4 of histone H3 (H3K4) in the nucleolus of the host cell, thereby enhancing heterochromatic rDNA transcription. Moreover, LegAS4 contains an ankyrin repeat domain of unknown function at its C-terminal region. Here, we report the crystal structure of LegAS4 in complex with S-adenosyl-l-methionine (SAM). Our data indicate that the ankyrin repeats interact extensively with the SET domain, especially with the SAM-binding amino acids, through conserved residues. Conserved surface analysis marks Glu159, Glu203, and Glu206 on the SET domain serve as candidate residues involved in interaction with the positively charged histone tail. Conserved surface residues on the ankyrin repeat domain surround a small pocket, which is suspected to serve as a binding site for an unknown ligand.


Subject(s)
Bacterial Proteins/chemistry , Legionella pneumophila/chemistry , Type IV Secretion Systems/chemistry , Amino Acid Sequence , Ankyrin Repeat , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Histones/genetics , Histones/metabolism , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Models, Molecular , Molecular Sequence Data , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Structural Homology, Protein , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL