Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Nat Struct Mol Biol ; 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38388830

ABSTRACT

The RAS-MAPK pathway regulates cell proliferation, differentiation and survival, and its dysregulation is associated with cancer development. The pathway minimally comprises the small GTPase RAS and the kinases RAF, MEK and ERK. Activation of RAF by RAS is notoriously intricate and remains only partially understood. There are three RAF isoforms in mammals (ARAF, BRAF and CRAF) and two related pseudokinases (KSR1 and KSR2). RAS-mediated activation of RAF depends on an allosteric mechanism driven by the dimerization of its kinase domain. Recent work on human RAFs showed that MEK binding to KSR1 promotes KSR1-BRAF heterodimerization, which leads to the phosphorylation of free MEK molecules by BRAF. Similar findings were made with the single Drosophila RAF homolog. Here we show that the fly scaffold proteins CNK and HYP stabilize the KSR-MEK interaction, which in turn enhances RAF-KSR heterodimerization and RAF activation. The cryogenic electron microscopy structure of the minimal KSR-MEK-CNK-HYP complex reveals a ring-like arrangement of the CNK-HYP complex allowing CNK to simultaneously engage KSR and MEK, thus stabilizing the binary interaction. Together, these results illuminate how CNK contributes to RAF activation by stimulating the allosteric function of KSR and highlight the diversity of mechanisms impacting RAF dimerization as well as the regulatory potential of the KSR-MEK interaction.

2.
Nat Commun ; 14(1): 7143, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37932265

ABSTRACT

Type 4 pili (T4P) are important virulence factors, which belong to a superfamily of nanomachines ubiquitous in prokaryotes, called type 4 filaments (T4F). T4F are defined as helical polymers of type 4 pilins. Recent advances in cryo-electron microscopy (cryo-EM) led to structures of several T4F, revealing that the long N-terminal α-helix (α1) - the trademark of pilins - packs in the centre of the filaments to form a hydrophobic core. In diderm bacteria - all available bacterial T4F structures are from diderm species - a portion of α1 is melted (unfolded). Here we report that this architecture is conserved in phylogenetically distant monoderm species by determining the structure of Streptococcus sanguinis T4P. Our 3.7 Å resolution cryo-EM structure of S. sanguinis heteropolymeric T4P and the resulting full atomic model including all minor pilins highlight universal features of bacterial T4F and have widespread implications in understanding T4F biology.


Subject(s)
Fimbriae Proteins , Fimbriae, Bacterial , Fimbriae Proteins/chemistry , Cryoelectron Microscopy/methods , Fimbriae, Bacterial/chemistry , Bacteria , Polymers
3.
Microbiol Spectr ; 11(6): e0147823, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37800964

ABSTRACT

IMPORTANCE: The type VI secretion system (T6SS) is a bacterial contractile injection system involved in bacterial competition by the delivery of antibacterial toxins. The T6SS consists of an envelope-spanning complex that recruits the baseplate, allowing the polymerization of a contractile tail structure. The tail is a tube wrapped by a sheath and topped by the tip of the system, the VgrG spike/PAAR complex. Effectors loaded onto the puncturing tip or into the tube are propelled in the target cells upon sheath contraction. The PAAR protein tips and sharpens the VgrG spike. However, the importance and the function of this protein remain unclear. Here, we provide evidence for association of PAAR at the tip of the VgrG spike. We also found that the PAAR protein is a T6SS critical component required for baseplate and sheath assembly.


Subject(s)
Type VI Secretion Systems , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism , Bacterial Proteins/metabolism
4.
PLoS Pathog ; 19(5): e1011368, 2023 05.
Article in English | MEDLINE | ID: mdl-37155700

ABSTRACT

The bacterial human pathogen Helicobacter pylori produces a type IV secretion system (cagT4SS) to inject the oncoprotein CagA into gastric cells. The cagT4SS external pilus mediates attachment of the apparatus to the target cell and the delivery of CagA. While the composition of the pilus is unclear, CagI is present at the surface of the bacterium and required for pilus formation. Here, we have investigated the properties of CagI by an integrative structural biology approach. Using Alpha Fold 2 and Small Angle X-ray scattering, it was found that CagI forms elongated dimers mediated by rod-shape N-terminal domains (CagIN) prolonged by globular C-terminal domains (CagIC). Three Designed Ankyrin Repeat Proteins (DARPins) K2, K5 and K8 selected against CagI interacted with CagIC with subnanomolar affinities. The crystal structures of the CagI:K2 and CagI:K5 complexes were solved and identified the interfaces between the molecules, thereby providing a structural explanation for the difference in affinity between the two binders. Purified CagI and CagIC were found to interact with adenocarcinoma gastric (AGS) cells, induced cell spreading and the interaction was inhibited by K2. The same DARPin inhibited CagA translocation by up to 65% in AGS cells while inhibition levels were 40% and 30% with K8 and K5, respectively. Our study suggests that CagIC plays a key role in cagT4SS-mediated CagA translocation and that DARPins targeting CagI represent potent inhibitors of the cagT4SS, a crucial risk factor for gastric cancer development.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Humans , Bacterial Proteins/metabolism , Antigens, Bacterial/metabolism , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Designed Ankyrin Repeat Proteins , Helicobacter pylori/metabolism , Helicobacter Infections/microbiology
5.
Nucleic Acids Res ; 51(6): 2800-2817, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36806960

ABSTRACT

RecA-mediated homologous recombination (HR) is a key mechanism for genome maintenance and plasticity in bacteria. It proceeds through RecA assembly into a dynamic filament on ssDNA, the presynaptic filament, which mediates DNA homology search and ordered DNA strand exchange. Here, we combined structural, single molecule and biochemical approaches to characterize the ATP-dependent assembly mechanism of the presynaptic filament of RecA from Streptococcus pneumoniae (SpRecA), in comparison to the Escherichia coli RecA (EcRecA) paradigm. EcRecA polymerization on ssDNA is assisted by the Single-Stranded DNA Binding (SSB) protein, which unwinds ssDNA secondary structures that block EcRecA nucleofilament growth. We report by direct microscopic analysis of SpRecA filamentation on ssDNA that neither of the two paralogous pneumococcal SSBs could assist the extension of SpRecA nucleopolymers. Instead, we found that the conserved RadA helicase promotes SpRecA nucleofilamentation in an ATP-dependent manner. This allowed us to solve the atomic structure of such a long native SpRecA nucleopolymer by cryoEM stabilized with ATPγS. It was found to be equivalent to the crystal structure of the EcRecA filament with a marked difference in how RecA mediates nucleotide orientation in the stretched ssDNA. Then, our results show that SpRecA and EcRecA HR activities are different, in correlation with their distinct ATP-dependent ssDNA binding modes.


Subject(s)
Rec A Recombinases , Streptococcus pneumoniae , Adenosine Triphosphate/metabolism , DNA/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Rec A Recombinases/metabolism , Rec A Recombinases/ultrastructure , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Cryoelectron Microscopy
6.
mBio ; 13(5): e0013422, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36154281

ABSTRACT

Type VIIb secretion systems (T7SSb) were recently proposed to mediate different aspects of Firmicutes physiology, including bacterial pathogenicity and competition. However, their architecture and mechanism of action remain largely obscure. Here, we present a detailed analysis of the T7SSb-mediated bacterial competition in Bacillus subtilis, using the effector YxiD as a model for the LXG secreted toxins. By systematically investigating protein-protein interactions, we reveal that the membrane subunit YukC contacts all T7SSb components, including the WXG100 substrate YukE and the LXG effector YxiD. YukC's crystal structure shows unique features, suggesting an intrinsic flexibility that is required for T7SSb antibacterial activity. Overall, our results shed light on the role and molecular organization of the T7SSb and demonstrate the potential of B. subtilis as a model system for extensive structure-function studies of these secretion machineries. IMPORTANCE Type VII secretion systems mediate protein extrusion from Gram-positive bacteria and are classified as T7SSa and T7SSb in Actinobacteria and in Firmicutes, respectively. Despite the genetic divergence of T7SSa and T7SSb, the high degree of structural similarity of their WXG100 substrates suggests similar secretion mechanisms. Recent advances revealed the structures of several T7SSa cytoplasmic membrane complexes, but the molecular mechanism of secretion and the T7SSb architecture remain obscure. Here, we provide hints on the organization of T7SSb in B. subtilis and a high-resolution structure of its central pseudokinase subunit, opening new perspectives for the understanding of the T7SSb secretion mechanism by using B. subtilis as an amenable bacterial model.


Subject(s)
Type VII Secretion Systems , Type VII Secretion Systems/metabolism , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Anti-Bacterial Agents/metabolism
7.
Nat Commun ; 12(1): 6998, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853317

ABSTRACT

Bacteria have evolved toxins to outcompete other bacteria or to hijack host cell pathways. One broad family of bacterial polymorphic toxins gathers multidomain proteins with a modular organization, comprising a C-terminal toxin domain fused to a N-terminal domain that adapts to the delivery apparatus. Polymorphic toxins include bacteriocins, contact-dependent growth inhibition systems, and specialized Hcp, VgrG, PAAR or Rhs Type VI secretion (T6SS) components. We recently described and characterized Tre23, a toxin domain fused to a T6SS-associated Rhs protein in Photorhabdus laumondii, Rhs1. Here, we show that Rhs1 forms a complex with the T6SS spike protein VgrG and the EagR chaperone. Using truncation derivatives and cross-linking mass spectrometry, we demonstrate that VgrG-EagR-Rhs1 complex formation requires the VgrG C-terminal ß-helix and the Rhs1 N-terminal region. We then report the cryo-electron-microscopy structure of the Rhs1-EagR complex, demonstrating that the Rhs1 central region forms a ß-barrel cage-like structure that encapsulates the C-terminal toxin domain, and provide evidence for processing of the Rhs1 protein through aspartyl autoproteolysis. We propose a model for Rhs1 loading on the T6SS, transport and delivery into the target cell.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Photorhabdus/metabolism , Type VI Secretion Systems/chemistry , Adaptation, Physiological , Bacterial Proteins/chemistry , Bacterial Toxins/classification , Bacterial Toxins/genetics , Bacteriocins/chemistry , Contact Inhibition , Cryoelectron Microscopy , Mass Spectrometry , Models, Molecular , Photorhabdus/genetics , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism
8.
Sci Adv ; 7(10)2021 03.
Article in English | MEDLINE | ID: mdl-33674316

ABSTRACT

Mycoplasma immunoglobulin binding (MIB) and mycoplasma immunoglobulin protease (MIP) are surface proteins found in the majority of mycoplasma species, acting sequentially to capture antibodies and cleave off their VH domains. Cryo-electron microscopy structures show how MIB and MIP bind to a Fab fragment in a "hug of death" mechanism. As a result, the orientation of the VL and VH domains is twisted out of alignment, disrupting the antigen binding site. We also show that MIB-MIP has the ability to promote the dissociation of the antibody-antigen complex. This system is functional in cells and protects mycoplasmas from antibody-mediated agglutination. These results highlight the key role of the MIB-MIP system in immunity evasion by mycoplasmas through an unprecedented mechanism, and open exciting perspectives to use these proteins as potential tools in the antibody field.


Subject(s)
Mycoplasma , Antigen-Antibody Complex , Cryoelectron Microscopy , Endopeptidases , Immunoglobulin Fab Fragments , Membrane Proteins , Peptide Hydrolases
9.
Science ; 369(6505): 793-799, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32792392

ABSTRACT

Monoclonal antibodies (mAbs) targeting human antigen CD20 (cluster of differentiation 20) constitute important immunotherapies for the treatment of B cell malignancies and autoimmune diseases. Type I and II therapeutic mAbs differ in B cell binding properties and cytotoxic effects, reflecting differential interaction mechanisms with CD20. Here we present 3.7- to 4.7-angstrom cryo-electron microscopy structures of full-length CD20 in complexes with prototypical type I rituximab and ofatumumab and type II obinutuzumab. The structures and binding thermodynamics demonstrate that upon binding to CD20, type II mAbs form terminal complexes that preclude recruitment of additional mAbs and complement components, whereas type I complexes act as molecular seeds to increase mAb local concentration for efficient complement activation. Among type I mAbs, ofatumumab complexes display optimal geometry for complement recruitment. The uncovered mechanisms should aid rational design of next-generation immunotherapies targeting CD20.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Antigen-Antibody Complex/chemistry , Antigens, CD20/chemistry , Antineoplastic Agents/chemistry , Immunotherapy , Lymphoma, B-Cell/therapy , Rituximab/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antigen-Antibody Complex/immunology , Antigens, CD20/immunology , Antineoplastic Agents/immunology , B-Lymphocytes/immunology , Complement Activation , Cryoelectron Microscopy , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Protein Binding , Protein Conformation , Rituximab/immunology , Rituximab/therapeutic use
10.
EMBO J ; 39(11): e104129, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32350888

ABSTRACT

The bacterial type VI secretion system (T6SS) is a macromolecular machine that injects effectors into prokaryotic and eukaryotic cells. The mode of action of the T6SS is similar to contractile phages: the contraction of a sheath structure pushes a tube topped by a spike into target cells. Effectors are loaded onto the spike or confined into the tube. In enteroaggregative Escherichia coli, the Tle1 phospholipase binds the C-terminal extension of the VgrG trimeric spike. Here, we purify the VgrG-Tle1 complex and show that a VgrG trimer binds three Tle1 monomers and inhibits their activity. Using covalent cross-linking coupled to high-resolution mass spectrometry, we provide information on the sites of contact and further identify the requirement for a Tle1 N-terminal secretion sequence in complex formation. Finally, we report the 2.6-Å-resolution cryo-electron microscopy tri-dimensional structure of the (VgrG)3 -(Tle1)3 complex revealing how the effector binds its cargo, and how VgrG inhibits Tle1 phospholipase activity. The inhibition of Tle1 phospholipase activity once bound to VgrG suggests that Tle1 dissociation from VgrG is required upon delivery.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Phospholipases/metabolism , Type VI Secretion Systems/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Phospholipases/genetics , Type VI Secretion Systems/genetics
11.
Nat Commun ; 11(1): 1426, 2020 03 18.
Article in English | MEDLINE | ID: mdl-32188856

ABSTRACT

Acetaldehyde-alcohol dehydrogenase (AdhE) enzymes are a key metabolic enzyme in bacterial physiology and pathogenicity. They convert acetyl-CoA to ethanol via an acetaldehyde intermediate during ethanol fermentation in an anaerobic environment. This two-step reaction is associated to NAD+ regeneration, essential for glycolysis. The bifunctional AdhE enzyme is conserved in all bacterial kingdoms but also in more phylogenetically distant microorganisms such as green microalgae. It is found as an oligomeric form called spirosomes, for which the function remains elusive. Here, we use cryo-electron microscopy to obtain structures of Escherichia coli spirosomes in different conformational states. We show that spirosomes contain active AdhE monomers, and that AdhE filamentation is essential for its activity in vitro and function in vivo. The detailed analysis of these structures provides insight showing that AdhE filamentation is essential for substrate channeling within the filament and for the regulation of enzyme activity.


Subject(s)
Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/metabolism , Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/metabolism , Aldehydes/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Ethanol/metabolism , Alcohol Dehydrogenase/genetics , Aldehyde Oxidoreductases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Models, Molecular
12.
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
13.
Biochim Biophys Acta Biomembr ; 1861(2): 466-477, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30444973

ABSTRACT

Membrane protein (MP) complexes play key roles in all living cells. Their structural characterisation is hampered by difficulties in purifying and crystallising them. Recent progress in electron microscopy (EM) have revolutionised the field, not only by providing higher-resolution structures for previously characterised MPs but also by yielding first glimpses into the structure of larger and more challenging complexes, such as bacterial secretion systems. However, the resolution of pioneering EM structures may be difficult and their interpretation requires clues regarding the overall organisation of the complexes. In this context, we present BAmSA, a new method for localising transmembrane (TM) regions in MP complexes, using a general procedure that allows tagging them without resorting to neither genetic nor chemical modification. Labels bound to TM regions can be visualised directly on raw negative-stain EM images, on class averages, or on three-dimensional reconstructions, providing a novel strategy to explore the organisation of MP complexes.


Subject(s)
Cell Membrane/ultrastructure , Membrane Proteins/ultrastructure , Microscopy, Electron , Polymers/chemistry , Streptavidin/chemistry , Animals , Biotinylation , Cattle , Electron Transport Complex III/metabolism , Escherichia coli Proteins/metabolism , Lipoproteins/metabolism , Models, Molecular , Negative Staining
14.
Nat Microbiol ; 3(12): 1404-1416, 2018 12.
Article in English | MEDLINE | ID: mdl-30323254

ABSTRACT

To support their growth in a competitive environment and cause pathogenesis, bacteria have evolved a broad repertoire of macromolecular machineries to deliver specific effectors and toxins. Among these multiprotein complexes, the type VI secretion system (T6SS) is a contractile nanomachine that targets both prokaryotic and eukaryotic cells. The T6SS comprises two functional subcomplexes: a bacteriophage-related tail structure anchored to the cell envelope by a membrane complex. As in other contractile injection systems, the tail is composed of an inner tube wrapped by a sheath and built on the baseplate. In the T6SS, the baseplate is not only the tail assembly platform, but also docks the tail to the membrane complex and hence serves as an evolutionary adaptor. Here we define the biogenesis pathway and report the cryo-electron microscopy (cryo-EM) structure of the wedge protein complex of the T6SS from enteroaggregative Escherichia coli (EAEC). Using an integrative approach, we unveil the molecular architecture of the whole T6SS baseplate and its interaction with the tail sheath, offering detailed insights into its biogenesis and function. We discuss architectural and mechanistic similarities but also reveal key differences with the T4 phage and Mu phage baseplates.


Subject(s)
Bacteriophages/metabolism , Escherichia coli/metabolism , Multiprotein Complexes/chemistry , Type VI Secretion Systems/chemistry , Type VI Secretion Systems/physiology , Cell Membrane , Cryoelectron Microscopy , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Models, Molecular , Protein Conformation , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Type VI Secretion Systems/genetics
15.
Annu Rev Microbiol ; 72: 231-254, 2018 Sep 08.
Article in English | MEDLINE | ID: mdl-30004822

ABSTRACT

Bacterial secretion systems are responsible for releasing macromolecules to the extracellular milieu or directly into other cells. These membrane complexes are associated with pathogenicity and bacterial fitness. Understanding of these large assemblies has exponentially increased in the last few years thanks to electron microscopy. In fact, a revolution in this field has led to breakthroughs in characterizing the structures of secretion systems and other macromolecular machineries so as to obtain high-resolution images of complexes that could not be crystallized. In this review, we give a brief overview of structural advancements in the understanding of secretion systems, focusing in particular on cryo-electron microscopy, whether tomography or single-particle analysis. We describe how such techniques have contributed to knowledge of the mechanism of macromolecule secretion in bacteria and the impact they will have in the future.


Subject(s)
Bacteria/enzymology , Bacterial Secretion Systems/ultrastructure , Cryoelectron Microscopy/methods , Cryoelectron Microscopy/trends
16.
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
17.
Nat Commun ; 8(1): 2065, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234007

ABSTRACT

Secreted exopolysaccharides present important determinants for bacterial biofilm formation, survival, and virulence. Cellulose secretion typically requires the concerted action of a c-di-GMP-responsive inner membrane synthase (BcsA), an accessory membrane-anchored protein (BcsB), and several additional Bcs components. Although the BcsAB catalytic duo has been studied in great detail, its interplay with co-expressed subunits remains enigmatic. Here we show that E. coli Bcs proteins partake in a complex protein interaction network. Electron microscopy reveals a stable, megadalton-sized macromolecular assembly, which encompasses most of the inner membrane and cytosolic Bcs components and features a previously unobserved asymmetric architecture. Heterologous reconstitution and mutational analyses point toward a structure-function model, where accessory proteins regulate secretion by affecting both the assembly and stability of the system. Altogether, these results lay the foundation for more comprehensive models of synthase-dependent exopolysaccharide secretion in biofilms and add a sophisticated secretory nanomachine to the diverse bacterial arsenal for virulence and adaptation.


Subject(s)
Bacterial Secretion Systems/metabolism , Cellulose/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Membrane Proteins/metabolism , Adaptation, Physiological/physiology , Bacterial Secretion Systems/chemistry , Biofilms , Cyclic GMP/metabolism , DNA Mutational Analysis , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/ultrastructure , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Membrane Proteins/ultrastructure , Microscopy, Electron , Models, Biological , Protein Binding , Protein Domains/physiology , Protein Interaction Maps/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Software , Structure-Activity Relationship
18.
FEBS J ; 284(23): 4143-4157, 2017 12.
Article in English | MEDLINE | ID: mdl-29055076

ABSTRACT

The more severe strains of the bacterial human pathogen Helicobacter pylori produce a type IV secretion system (cagT4SS) to inject the oncoprotein cytotoxin-associated gene A (CagA) into gastric cells. This syringe-like molecular apparatus is prolonged by an external pilus that exploits integrins as receptors to mediate the injection of CagA. The molecular determinants of the interaction of the cagT4SS pilus with the integrin ectodomain are still poorly understood. In this study, we have used surface plasmon resonance (SPR) to generate a comprehensive analysis of the protein-protein interactions between purified CagA, CagL, CagI, CagY repeat domain II (CagYRRII ), CagY C-terminal domain (CagYB10 ) and integrin α5ß1 ectodomain (α5ß1E ) or headpiece domain (α5ß1HP ). We found that CagI, CagA, CagL and CagYB10 but not CagYRRII were able to interact with α5ß1E with affinities similar to the one observed for α5ß1E interaction with its physiological ligand fibronectin. We further showed that integrin activation and its associated conformational change increased CagA, CagL and CagYB10 affinities for the receptor. Furthermore, CagI did not interact with integrin unless the receptor was in open conformation. CagI, CagA but not CagL and CagYB10 interacted with the α5ß1HP . Our SPR study also revealed novel interactions between CagA and CagL, CagA and CagYB10 , and CagA and CagI. Altogether, our data map the network of interactions between host-cell α5ß1 integrin and the cagT4SS proteins and suggest that activation of the receptor promotes interactions with the secretion apparatus and possibly CagA injection.


Subject(s)
Bacterial Proteins/metabolism , Helicobacter pylori/metabolism , Integrin alpha5beta1/metabolism , Protein Interaction Mapping/methods , Type IV Secretion Systems/metabolism , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , CHO Cells , Cricetinae , Cricetulus , Helicobacter pylori/genetics , Humans , Integrin alpha5beta1/chemistry , Integrin alpha5beta1/genetics , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Scattering, Small Angle , Surface Plasmon Resonance , Type IV Secretion Systems/chemistry , Type IV Secretion Systems/genetics , X-Ray Diffraction
19.
Methods Mol Biol ; 1635: 125-138, 2017.
Article in English | MEDLINE | ID: mdl-28755367

ABSTRACT

Localization of specific subunits or domains of interest inside protein complexes can be challenging, especially for membrane machineries. The amphipatic nature of their subunits and their modular organization results in difficult genetic manipulation and instability upon purification. Here, we present different labeling approaches that have been demonstrated successful in the structural characterization of large membrane complexes.


Subject(s)
Staining and Labeling/methods , Type IV Secretion Systems/chemistry , Type VI Secretion Systems/chemistry , Bacterial Proteins/chemistry , Gold , Membrane Proteins/chemistry , Microscopy, Electron
20.
Mol Microbiol ; 105(5): 741-754, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28618091

ABSTRACT

Pneumococcal natural transformation contributes to genomic plasticity, antibiotic resistance development and vaccine escape. Streptococcus pneumoniae, like many other naturally transformable species, has evolved sophisticated protein machinery for the binding and uptake of DNA. Two proteins encoded by the comF operon, ComFA and ComFC, are involved in transformation but their exact molecular roles remain unknown. In this study, we provide experimental evidence that ComFA binds to single stranded DNA (ssDNA) and has ssDNA-dependent ATPase activity. We show that both ComFA and ComFC are essential for the transformation process in pneumococci. Moreover, we show that these proteins interact with each other and with other proteins involved in homologous recombination, such as DprA, thus placing the ComFA-ComFC duo at the interface between DNA uptake and DNA recombination during transformation.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/metabolism , Transformation, Bacterial/physiology , Adenosine Triphosphatases/genetics , Bacterial Proteins/metabolism , DNA/metabolism , DNA, Single-Stranded/metabolism , Homologous Recombination , Membrane Proteins/metabolism , Protein Binding , Rec A Recombinases/metabolism , Recombination, Genetic , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Transformation, Bacterial/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...