Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Nature ; 564(7734): 77-82, 2018 12.
Article in English | MEDLINE | ID: mdl-30405243

ABSTRACT

The type 9 secretion system (T9SS) is the protein export pathway of bacteria of the Gram-negative Fibrobacteres-Chlorobi-Bacteroidetes superphylum and is an essential determinant of pathogenicity in severe periodontal disease. The central element of the T9SS is a so-far uncharacterized protein-conducting translocon located in the bacterial outer membrane. Here, using cryo-electron microscopy, we provide structural evidence that the translocon is the T9SS protein SprA. SprA forms an extremely large (36-strand) single polypeptide transmembrane ß-barrel. The barrel pore is capped on the extracellular end, but has a lateral opening to the external membrane surface. Structures of SprA bound to different components of the T9SS show that partner proteins control access to the lateral opening and to the periplasmic end of the pore. Our results identify a protein transporter with a distinctive architecture that uses an alternating access mechanism in which the two ends of the protein-conducting channel are open at different times.


Subject(s)
Bacterial Secretion Systems/metabolism , Bacterial Secretion Systems/ultrastructure , Cryoelectron Microscopy , Flavobacterium , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/genetics , Flavobacterium/chemistry , Flavobacterium/genetics , Flavobacterium/metabolism , Flavobacterium/ultrastructure , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding , Protein Domains , Protein Structure, Secondary , Protein Transport
2.
EMBO J ; 38(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30877094

ABSTRACT

Bacteria have evolved macromolecular machineries that secrete effectors and toxins to survive and thrive in diverse environments. The type VI secretion system (T6SS) is a contractile machine that is related to Myoviridae phages. It is composed of a phage tail-like structure inserted in the bacterial cell envelope by a membrane complex (MC) comprising the TssJ, TssL and TssM proteins. We previously reported the low-resolution negative-stain electron microscopy structure of the enteroaggregative Escherichia coli MC and proposed a rotational 5-fold symmetry with a TssJ:TssL:TssM stoichiometry of 2:2:2. Here, cryo-electron tomography analyses of the T6SS MC confirm the 5-fold symmetry in situ and identify the regions of the structure that insert into the bacterial membranes. A high-resolution model obtained by single-particle cryo-electron microscopy highlights new features: five additional copies of TssJ, yielding a TssJ:TssL:TssM stoichiometry of 3:2:2, an 11-residue loop in TssM, protruding inside the lumen of the MC and constituting a functionally important periplasmic gate, and hinge regions. Based on these data, we propose an updated model on MC structure and dynamics during T6SS assembly and function.


Subject(s)
Type VI Secretion Systems/chemistry , Type VI Secretion Systems/metabolism , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cryoelectron Microscopy , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Protein Binding , Protein Structure, Quaternary
3.
PLoS Biol ; 17(7): e3000351, 2019 07.
Article in English | MEDLINE | ID: mdl-31260457

ABSTRACT

Type III protein-secretion machines are essential for the interactions of many pathogenic or symbiotic bacterial species with their respective eukaryotic hosts. The core component of these machines is the injectisome, a multiprotein complex that mediates the selection of substrates, their passage through the bacterial envelope, and ultimately their delivery into eukaryotic target cells. The injectisome is composed of a large cytoplasmic complex or sorting platform, a multiring base embedded in the bacterial envelope, and a needle-like filament that protrudes several nanometers from the bacterial surface and is capped at its distal end by the tip complex. A characteristic feature of these machines is that their activity is stimulated by contact with target host cells. The sensing of target cells, thought to be mediated by the distal tip of the needle filament, generates an activating signal that must be transduced to the secretion machine by the needle filament. Here, through a multidisciplinary approach, including solid-state NMR (SSNMR) and cryo electron microscopy (cryo-EM) analyses, we have identified critical residues of the needle filament protein of a Salmonella Typhimurium type III secretion system that are involved in the regulation of the activity of the secretion machine. We found that mutations in the needle filament protein result in various specific phenotypes associated with different steps in the type III secretion process. More specifically, these studies reveal an important role for a polymorphic helix of the needle filament protein and the residues that line the lumen of its central channel in the control of type III secretion.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Multiprotein Complexes/metabolism , Salmonella typhimurium/metabolism , Type III Secretion Systems/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/genetics , Cryoelectron Microscopy , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/ultrastructure , Mutation , Protein Conformation , Protein Transport/genetics , Salmonella typhimurium/genetics , Type III Secretion Systems/genetics , Type III Secretion Systems/ultrastructure
4.
Cell Microbiol ; 22(5): e13157, 2020 05.
Article in English | MEDLINE | ID: mdl-31891220

ABSTRACT

Many human pathogens use Type III, Type IV, and Type VI secretion systems to deliver effectors into their target cells. The contribution of these secretion systems to microbial virulence was the main focus of a workshop organised by the International University of Andalusia in Spain. The meeting addressed structure-function, substrate recruitment, and translocation processes, which differ widely on the different secretion machineries, as well as the nature of the translocated effectors and their roles in subverting the host cell. An excellent panel of worldwide speakers presented the state of the art of the field, highlighting the involvement of bacterial secretion in human disease and discussing mechanistic aspects of bacterial pathogenicity, which can provide the bases for the development of novel antivirulence strategies.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Bacteria/genetics , Bacterial Proteins/genetics , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/genetics , Humans , Protein Transport , Spain , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism , Virulence
5.
Proc Natl Acad Sci U S A ; 115(31): 7967-7972, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30012626

ABSTRACT

The transport of proteins across or into membranes is a vital biological process, achieved in every cell by the conserved Sec machinery. In bacteria, SecYEG combines with the SecA motor protein for secretion of preproteins across the plasma membrane, powered by ATP hydrolysis and the transmembrane proton-motive force (PMF). The activities of SecYEG and SecA are modulated by membrane lipids, particularly cardiolipin (CL), a specialized phospholipid known to associate with a range of energy-transducing machines. Here, we identify two specific CL binding sites on the Thermotoga maritima SecA-SecYEG complex, through application of coarse-grained molecular dynamics simulations. We validate the computational data and demonstrate the conserved nature of the binding sites using in vitro mutagenesis, native mass spectrometry, biochemical analysis, and fluorescence spectroscopy of Escherichia coli SecYEG. The results show that the two sites account for the preponderance of functional CL binding to SecYEG, and mediate its roles in ATPase and protein transport activity. In addition, we demonstrate an important role for CL in the conferral of PMF stimulation of protein transport. The apparent transient nature of the CL interaction might facilitate proton exchange with the Sec machinery, and thereby stimulate protein transport, by a hitherto unexplored mechanism. This study demonstrates the power of coupling the high predictive ability of coarse-grained simulation with experimental analyses, toward investigation of both the nature and functional implications of protein-lipid interactions.


Subject(s)
Bacterial Secretion Systems/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Molecular Dynamics Simulation , Proton-Motive Force , SEC Translocation Channels/chemistry , Thermotoga maritima/chemistry , Bacterial Secretion Systems/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , SEC Translocation Channels/metabolism , Thermotoga maritima/metabolism
6.
BMC Bioinformatics ; 21(1): 480, 2020 Oct 27.
Article in English | MEDLINE | ID: mdl-33109082

ABSTRACT

BACKGROUND: Classification of certain proteins with specific functions is momentous for biological research. Encoding approaches of protein sequences for feature extraction play an important role in protein classification. Many computational methods (namely classifiers) are used for classification on protein sequences according to various encoding approaches. Commonly, protein sequences keep certain labels corresponding to different categories of biological functions (e.g., bacterial type IV secreted effectors or not), which makes protein prediction a fantasy. As to protein prediction, a kernel set of protein sequences keeping certain labels certified by biological experiments should be existent in advance. However, it has been hardly ever seen in prevailing researches. Therefore, unsupervised learning rather than supervised learning (e.g. classification) should be considered. As to protein classification, various classifiers may help to evaluate the effectiveness of different encoding approaches. Besides, variable selection from an encoded feature representing protein sequences is an important issue that also needs to be considered. RESULTS: Focusing on the latter problem, we propose a new method for variable selection from an encoded feature representing protein sequences. Taking a benchmark dataset containing 1947 protein sequences as a case, experiments are made to identify bacterial type IV secreted effectors (T4SE) from protein sequences, which are composed of 399 T4SE and 1548 non-T4SE. Comparable and quantified results are obtained only using certain components of the encoded feature, i.e., position-specific scoring matix, and that indicates the effectiveness of our method. CONCLUSIONS: Certain variables other than an encoded feature they belong to do work for discrimination between different types of proteins. In addition, ensemble classifiers with an automatic assignment of different base classifiers do achieve a better classification result.


Subject(s)
Algorithms , Bacterial Proteins/chemistry , Bacterial Secretion Systems/chemistry , Amino Acid Sequence , Area Under Curve , Protein Domains , ROC Curve
7.
J Struct Biol ; 212(3): 107651, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33096229

ABSTRACT

Recent reports indicate that the Type six secretion system exported effector 8 (Tse8) is a cytoactive effector secreted by the Type VI secretion system (T6SS) of the human pathogen Pseudomonas aeruginosa. The T6SS is a nanomachine that assembles inside of the bacteria and injects effectors/toxins into target cells, providing a fitness advantage over competing bacteria and facilitating host colonisation. Here we present the first crystal structure of Tse8 revealing that it conserves the architecture of the catalytic triad Lys84-transSer162-Ser186 that characterises members of the Amidase Signature superfamily. Furthermore, using binding affinity experiments, we show that the interaction of phenylmethylsulfonyl fluoride (PMSF) to Tse8 is dependent on the putative catalytic residue Ser186, providing support for its nucleophilic reactivity. This work thus demonstrates that Tse8 belongs to the Amidase Signature (AS) superfamily. Furthermore, it highlights Tse8 similarity to two family members: the Stenotrophomonas maltophilia Peptide Amidase and the Glutamyl-tRNAGln amidotransferase subunit A from Staphylococcus aureus.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Secretion Systems/chemistry , Pseudomonas aeruginosa/chemistry , Type VI Secretion Systems/chemistry , Amidohydrolases/chemistry , RNA, Transfer/chemistry
8.
Biochem Biophys Res Commun ; 532(1): 114-119, 2020 10 29.
Article in English | MEDLINE | ID: mdl-32828535

ABSTRACT

Porphyromonas gingivalis, which is a major pathogen of the periodontal disease, secrets virulence factors such as gingipain proteases via the type IX secretion system (T9SS). T9SS consists of a trans-periplasmic core complex, the outer membrane translocon complex and the cell-surface complex attached on the outer membrane. PorM is a major component of the trans-periplasmic core complex and is believed to connect the outer membrane component with the inner membrane component. Recent structural studies have revealed that the periplasmic region of GldM, a PorM homolog of a gliding bacterium, consist of four domains and forms a dimer with a straight rod shape. However, only fragment structures are known for PorM. Moreover, one of the PorM fragment structure shows a kink. Here we show the structure of the entire structure of the periplasmic region of PorM (PorMp) at 3.7 Å resolution. PorMp is made up of four domains and forms a unique dimeric structure with an asymmetric, kinked-rod shape. The structure and the following mutational analysis revealed that R204 stabilizes the kink between the D1 and D2 domains and is essential for gingipains secretion, suggesting that the kinked structure of PorM is important for the functional T9SS formation.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Secretion Systems/chemistry , Porphyromonas gingivalis/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Secretion Systems/genetics , Crystallography, X-Ray , Genes, Bacterial , Humans , Models, Molecular , Mutation , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/pathogenicity , Protein Domains , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Quaternary , Sequence Homology, Amino Acid
9.
Mol Microbiol ; 110(1): 64-81, 2018 10.
Article in English | MEDLINE | ID: mdl-30030863

ABSTRACT

The periodontal pathogen Porphyromonas gingivalis secretes many potent virulence factors using the type IX secretion system (T9SS). T9SS cargo proteins that have been structurally determined by X-ray crystallography are composed of a signal peptide, functional domain(s), an immunoglobulin (Ig)-like domain and a C-terminal domain. Role of the Ig-like domains of cargo proteins in the T9SS has not been elucidated. Gingipain proteases, which are cargo proteins of the T9SS, were degraded when their Ig-like domains were lacking or truncated. The degradation was dependent on the activity of a quality control factor, HtrA protease. Another T9SS cargo protein, HBP35, which has a thioredoxin domain as a functional domain, was analyzed by X-ray crystallography, revealing that HBP35 has an Ig-like domain after the thioredoxin domain and that the hydrophobic regions of the thioredoxin domain and the Ig-like domain face each other. HBP35 with substitution of hydrophobic amino acids in the Ig-like domain was degraded depending on HtrA. These results suggest that the Ig-like domain mediates stability of the cargo proteins in the T9SS.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Secretion Systems/metabolism , Cysteine Endopeptidases/metabolism , Immunoglobulin Domains/physiology , Porphyromonas gingivalis/physiology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/genetics , Caseins/metabolism , Crystallography, X-Ray , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Gingipain Cysteine Endopeptidases , Immunoglobulin Domains/genetics , Muramidase/metabolism , Porphyromonas gingivalis/genetics , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Serine Proteases/chemistry , Serine Proteases/genetics , Serine Proteases/metabolism
10.
Int J Med Microbiol ; 309(7): 151322, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31262642

ABSTRACT

In Gram-negative bacteria, secretion of toxins ensure the survival of the bacterium. Such toxins are secreted by sophisticated multiprotein systems. The most conserved part in some of these secretion systems are components, called secretins, which form the outer membrane ring in these systems. Recent structural studies shed some light on the oligomeric organization of secretins. However, the mechanisms by which these proteins are targeted to the outer membrane and assemble there into ring structures are still not fully understood. This review discusses the various species-specific targeting and assembly pathways that are taken by secretins in order to form their functional oligomers.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Bacterial Outer Membrane/chemistry , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/metabolism , Gram-Negative Bacteria/chemistry , Gram-Negative Bacteria/metabolism , Models, Biological , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Multimerization , Protein Transport
11.
J Bacteriol ; 200(5)2018 03 01.
Article in English | MEDLINE | ID: mdl-29263097

ABSTRACT

Bacterial secretins are outer membrane proteins that provide a path for secreted proteins to access the cell exterior/surface. They are one of the core components of secretion machines and are found in type II and type III secretion systems (T2SS and T3SS, respectively). The secretins comprise giant ring-shaped homo-oligomers whose precise atomic organization was only recently deciphered thanks to spectacular developments in cryo-electron microscopy (cryo-EM) imaging techniques.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Secretion Systems/chemistry , Secretin/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/ultrastructure , Bacterial Secretion Systems/metabolism , Bacterial Secretion Systems/ultrastructure , Cryoelectron Microscopy/methods , Klebsiella/chemistry , Models, Molecular , Secretin/metabolism , Type II Secretion Systems/chemistry , Type II Secretion Systems/ultrastructure , Vibrio/chemistry
12.
Curr Issues Mol Biol ; 25: 1-42, 2018.
Article in English | MEDLINE | ID: mdl-28875938

ABSTRACT

In this review we examine the use of secretion systems by bacteria to subvert host functions. Bacteria have evolved multiple systems to interact with and overcome their eukaryotic host and other prokaryotes. Secretion systems are required for the release of several effectors through the bacterial membrane(s) into the extracellular space or directly into the cytoplasm of the host. We review the secretion systems of Gram-positive and Gram-negative bacteria and describe briefly the structural composition of the seven secretion systems that have been associated with increased virulence through subversion of host functions. Some of the effects of such systems on eukaryotic host processes have been studied extensively. We also describe the best-characterized effectors of each secretion system to give an overview of the molecular mechanisms employed by bacteria to hide from the immune system and convert eukaryotic cells into optimal ecological niches for their replication.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Gene Expression Regulation, Bacterial , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/classification , Bacterial Secretion Systems/genetics , Eukaryotic Cells/microbiology , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/pathogenicity , Host-Pathogen Interactions , Humans , Models, Molecular , Periplasm/metabolism , Protein Structure, Secondary , Protein Transport , Virulence , Virulence Factors/genetics
13.
J Biol Chem ; 291(50): 25837-25852, 2016 Dec 09.
Article in English | MEDLINE | ID: mdl-27770024

ABSTRACT

Like many Gram-negative pathogens, Shigella rely on a complex type III secretion system (T3SS) to inject effector proteins into host cells, take over host functions, and ultimately establish infection. Despite these critical roles, the energetics and regulatory mechanisms controlling the T3SS and pathogen virulence remain largely unclear. In this study, we present a series of high resolution crystal structures of Spa47 and use the structures to model an activated Spa47 oligomer, finding that ATP hydrolysis may be supported by specific side chain contributions from adjacent protomers within the complex. Follow-up mutagenesis experiments targeting the predicted active site residues validate the oligomeric model and determined that each of the tested residues are essential for Spa47 ATPase activity, although they are not directly responsible for stable oligomer formation. Although N-terminal domain truncation was necessary for crystal formation, it resulted in strictly monomeric Spa47 that is unable to hydrolyze ATP, despite maintaining the canonical ATPase core structure and active site residues. Coupled with studies of ATPase inactive full-length Spa47 point mutants, we find that Spa47 oligomerization and ATP hydrolysis are needed for complete T3SS apparatus formation, a proper translocator secretion profile, and Shigella virulence. This work represents the first structure-function characterization of Spa47, uniquely complementing the multitude of included Shigella T3SS phenotype assays and providing a more complete understanding of T3SS ATPase-mediated pathogen virulence. Additionally, these findings provide a strong platform for follow-up studies evaluating regulation of Spa47 oligomerization in vivo as a much needed means of treating and perhaps preventing shigellosis.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacterial Secretion Systems/metabolism , Point Mutation , Protein Multimerization , Shigella flexneri/metabolism , Shigella flexneri/pathogenicity , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/genetics , Humans , Structure-Activity Relationship
14.
J Biol Chem ; 291(30): 15767-77, 2016 07 22.
Article in English | MEDLINE | ID: mdl-27226543

ABSTRACT

Legionella pneumophila is a causative agent of a severe pneumonia, known as Legionnaires' disease. Legionella pathogenicity is mediated by specific virulence factors, called bacterial effectors, which are injected into the invaded host cell by the bacterial type IV secretion system. Bacterial effectors are involved in complex interactions with the components of the host cell immune and signaling pathways, which eventually lead to bacterial survival and replication inside the mammalian cell. Structural and functional studies of bacterial effectors are, therefore, crucial for elucidating the mechanisms of Legionella virulence. Here we describe the crystal structure of the LpiR1 (Lpg0634) effector protein and investigate the effects of its overexpression in mammalian cells. LpiR1 is an α-helical protein that consists of two similar domains aligned in an antiparallel fashion. The hydrophilic cleft between the domains might serve as a binding site for a potential host cell interaction partner. LpiR1 binds the phosphate group at a conserved site and is stabilized by Mn(2+), Ca(2+), or Mg(2+) ions. When overexpressed in mammalian cells, a GFP-LpiR1 fusion protein is localized in the cytoplasm. Intracellular signaling antibody array analysis revealed small changes in the phosphorylation state of several components of the Akt signaling pathway in HEK293T cells overexpressing LpiR1.


Subject(s)
Bacterial Secretion Systems , Legionella pneumophila , Legionnaires' Disease , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Virulence Factors , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/metabolism , HEK293 Cells , Humans , Legionella pneumophila/chemistry , Legionella pneumophila/genetics , Legionella pneumophila/metabolism , Legionella pneumophila/pathogenicity , Legionnaires' Disease/genetics , Legionnaires' Disease/metabolism , Metals/chemistry , Metals/metabolism , Protein Domains , Structure-Activity Relationship , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism
15.
Mol Microbiol ; 100(5): 912-21, 2016 06.
Article in English | MEDLINE | ID: mdl-26915347

ABSTRACT

Bacterial conjugation is the main mechanism responsible for the dissemination of antibiotic resistance genes. Hence, the search for specific conjugation inhibitors is paramount in the fight against the spread of these genes. In this pursuit, unsaturated fatty acids have been found to specifically inhibit bacterial conjugation. Despite the growing interest on these compounds, their mode of action and their specific target remain unknown. Here, we identified TrwD, a Type IV secretion traffic ATPase, as the molecular target for fatty acid-mediated inhibition of conjugation. Moreover, 2-alkynoic fatty acids, which are also potent inhibitors of bacterial conjugation, are also powerful inhibitors of the ATPase activity of TrwD. Characterization of the kinetic parameters of ATPase inhibition has led us to identify the catalytic mechanism by which fatty acids exert their activity. These results open a new avenue for the rational design of inhibitors of bacterial conjugation in the fight against the dissemination of antibiotic resistance genes.


Subject(s)
Adenosine Triphosphatases/metabolism , Conjugation, Genetic/drug effects , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli/genetics , Fatty Acids, Unsaturated/pharmacology , Linoleic Acid/pharmacology , Bacterial Proteins/genetics , Bacterial Secretion Systems/chemistry , Fatty Acids, Unsaturated/chemical synthesis , Kinetics , Molecular Docking Simulation , Plasmids
16.
Microb Pathog ; 110: 225-231, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28687320

ABSTRACT

Chronic burn wound infections caused by Stapyhylococcus aureus (S. aureus) are largely associated with biofilm formation. However, the mechanism by which S. aureus form biofilm in clinical environments is far less understood. In the present study we addressed the association between biofilm formation and membrane vesicle (MV) secretion of S. aureus during vancomycin treatment. A representative methicillin-resistant S. aureus (MRSA) strain BWMR22 obtained from a chronic burn wound was used in this study. Transmission electron microscope was used to observe the MV secretion. Fourier transform infrared spectroscopy was used to analyze the chemical component of MV. Biofilm formation was assayed under conditions of sub-inhibitory concentrations of vancomycin. Functional potencies of MV in surface adhesion and auto-aggregation were assayed in the presence of additional purified MVs. Biofilm formation by S. aureus BWMR22 was enhanced in the presence of sub-inhibitory concentration of vancomycin. Vancomycin treatment caused an increase in the chemical composition of protein relative to carbohydrates of secreted MVs, a property which was highly associated with bacterial hydrophobicity, surface adhesion, and intercellular aggregation. These findings suggest that MV secretion is correlated with biofilm formation by MRSA especially under clinical conditions with improper vancomycin chemotherapy. This study first demonstrates a potential role of MVs in the biofilm formation by S. aureus, stresses on the importance of avoiding low dose of antibiotic therapy in controlling of S. aureus infections, and provides further information to reveal the mechanisms behind MRSA infections.


Subject(s)
Bacterial Secretion Systems/drug effects , Biofilms/drug effects , Biofilms/growth & development , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/physiology , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Adhesion/drug effects , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/metabolism , Bacterial Secretion Systems/ultrastructure , Cell Aggregation/drug effects , Cell Communication/drug effects , Cell Culture Techniques , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Vancomycin/administration & dosage
17.
J Comput Aided Mol Des ; 31(11): 1029-1038, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29127583

ABSTRACT

Various bacterial pathogens can deliver their secreted substrates also called as effectors through type IV secretion systems (T4SSs) into host cells and cause diseases. Since T4SS secreted effectors (T4SEs) play important roles in pathogen-host interactions, identifying them is crucial to our understanding of the pathogenic mechanisms of T4SSs. A few computational methods using machine learning algorithms for T4SEs prediction have been developed by using features of C-terminal residues. However, recent studies have shown that targeting information can also be encoded in the N-terminal region of at least some T4SEs. In this study, we present an effective method for T4SEs prediction by novelly integrating both N-terminal and C-terminal sequence information. First, we collected a comprehensive dataset across multiple bacterial species of known T4SEs and non-T4SEs from literatures. Then, three types of distinctive features, namely amino acid composition, composition, transition and distribution and position-specific scoring matrices were calculated for 50 N-terminal and 100 C-terminal residues. After that, we employed information gain represent to rank the importance score of the 150 different position residues for T4SE secretion signaling. At last, 125 distinctive position residues were singled out for the prediction model to classify T4SEs and non-T4SEs. The support vector machine model yields a high receiver operating curve of 0.916 in the fivefold cross-validation and an accuracy of 85.29% for the independent test set.


Subject(s)
Bacteria/chemistry , Bacterial Proteins/chemistry , Bacterial Secretion Systems/chemistry , Models, Molecular , Algorithms , Amino Acids/chemistry , Binding Sites , Computer Simulation , Databases, Factual , Drug Design , Host-Pathogen Interactions , Protein Binding , Protein Conformation , Support Vector Machine
18.
Nat Microbiol ; 9(4): 1089-1102, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38538833

ABSTRACT

Secretion systems are protein export machines that enable bacteria to exploit their environment through the release of protein effectors. The Type 9 Secretion System (T9SS) is responsible for protein export across the outer membrane (OM) of bacteria of the phylum Bacteroidota. Here we trap the T9SS of Flavobacterium johnsoniae in the process of substrate transport by disrupting the T9SS motor complex. Cryo-EM analysis of purified substrate-bound T9SS translocons reveals an extended translocon structure in which the previously described translocon core is augmented by a periplasmic structure incorporating the proteins SprE, PorD and a homologue of the canonical periplasmic chaperone Skp. Substrate proteins bind to the extracellular loops of a carrier protein within the translocon pore. As transport intermediates accumulate on the translocon when energetic input is removed, we deduce that release of the substrate-carrier protein complex from the translocon is the energy-requiring step in T9SS transport.


Subject(s)
Bacterial Proteins , Bacterial Secretion Systems , Bacterial Proteins/metabolism , Bacterial Secretion Systems/chemistry , Protein Transport , Carrier Proteins/metabolism
19.
Open Biol ; 14(6): 230448, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862016

ABSTRACT

Gram-negative bacteria from the Bacteroidota phylum possess a type-IX secretion system (T9SS) for protein secretion, which requires cargoes to have a C-terminal domain (CTD). Structurally analysed CTDs are from Porphyromonas gingivalis proteins RgpB, HBP35, PorU and PorZ, which share a compact immunoglobulin-like antiparallel 3+4 ß-sandwich (ß1-ß7). This architecture is essential as a P. gingivalis strain with a single-point mutant of RgpB disrupting the interaction of the CTD with its preceding domain prevented secretion of the protein. Next, we identified the C-terminus ('motif C-t.') and the loop connecting strands ß3 and ß4 ('motif Lß3ß4') as conserved. We generated two strains with insertion and replacement mutants of PorU, as well as three strains with ablation and point mutants of RgpB, which revealed both motifs to be relevant for T9SS function. Furthermore, we determined the crystal structure of the CTD of mirolase, a cargo of the Tannerella forsythia T9SS, which shares the same general topology as in Porphyromonas CTDs. However, motif Lß3ß4 was not conserved. Consistently, P. gingivalis could not properly secrete a chimaeric protein with the CTD of peptidylarginine deiminase replaced with this foreign CTD. Thus, the incompatibility of the CTDs between these species prevents potential interference between their T9SSs.


Subject(s)
Bacterial Proteins , Bacterial Secretion Systems , Porphyromonas gingivalis , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Secretion Systems/metabolism , Bacterial Secretion Systems/genetics , Bacterial Secretion Systems/chemistry , Models, Molecular , Crystallography, X-Ray , Amino Acid Sequence , Protein Sorting Signals , Protein Domains , Bacteroidetes/metabolism , Bacteroidetes/genetics , Tannerella forsythia/metabolism , Tannerella forsythia/genetics , Tannerella forsythia/chemistry , Structure-Activity Relationship , Protein Conformation
20.
Microbiol Spectr ; 10(1): e0160221, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35019767

ABSTRACT

The type IX secretion system (T9SS) transports cargo proteins through the outer membrane of Bacteroidetes and attaches them to the cell surface for functions including pathogenesis, gliding motility, and degradation of carbon sources. The T9SS comprises at least 20 different proteins and includes several modules: the trans-envelope core module comprising the PorL/M motor and the PorK/N ring, the outer membrane Sov translocon, and the cell attachment complex. However, the spatial organization of these modules is unknown. We have characterized the protein interactome of the Sov translocon in Porphyromonas gingivalis and identified Sov-PorV-PorA as well as Sov-PorW-PorN-PorK to be novel networks. PorW also interacted with PGN_1783 (PorD), which was required for maximum secretion efficiency. The identification of PorW as the missing link completes a continuous interaction network from the PorL/M motor to the Sov translocon, providing a pathway for cargo delivery and energy transduction from the inner membrane to the secretion pore. IMPORTANCE The T9SS is a newly identified protein secretion system of the Fibrobacteres-Chlorobi-Bacteroidetes superphylum used by pathogens associated with diseases of humans, fish, and poultry for the secretion and cell surface attachment of virulence factors. The T9SS comprises three known modules: (i) the trans-envelope core module comprising the PorL/M motor and the PorK/N ring, (ii) the outer membrane Sov translocon, and (iii) the cell surface attachment complex. The spatial organization and interaction of these modules have been a mystery. Here, we describe the protein interactome of the Sov translocon in the human pathogen Porphyromonas gingivalis and have identified PorW as the missing link which bridges PorN with Sov and so completes a continuous interaction network from the PorL/M motor to the Sov translocon, providing, for the first time, a pathway for cargo delivery and energy transduction from the inner membrane to the secretion pore.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Porphyromonas gingivalis/metabolism , Amino Acid Sequence , Bacterial Outer Membrane/chemistry , Bacterial Outer Membrane/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Secretion Systems/chemistry , Bacterial Secretion Systems/genetics , Porphyromonas gingivalis/chemistry , Porphyromonas gingivalis/genetics , Protein Binding , Protein Transport , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL