Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.028
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 86: 873-896, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28426242

ABSTRACT

Electron cryotomography (ECT) provides three-dimensional views of macromolecular complexes inside cells in a native frozen-hydrated state. Over the last two decades, ECT has revealed the ultrastructure of cells in unprecedented detail. It has also allowed us to visualize the structures of macromolecular machines in their native context inside intact cells. In many cases, such machines cannot be purified intact for in vitro study. In other cases, the function of a structure is lost outside the cell, so that the mechanism can be understood only by observation in situ. In this review, we describe the technique and its history and provide examples of its power when applied to cell biology. We also discuss the integration of ECT with other techniques, including lower-resolution fluorescence imaging and higher-resolution atomic structure determination, to cover the full scale of cellular processes.


Subject(s)
Cryoelectron Microscopy/methods , Electron Microscope Tomography/methods , Fimbriae, Bacterial/ultrastructure , Nuclear Pore/chemistry , Optical Imaging/methods , Prokaryotic Cells/ultrastructure , Archaea/metabolism , Archaea/ultrastructure , Bacteria/metabolism , Bacteria/ultrastructure , Bacterial Secretion Systems/metabolism , Bacterial Secretion Systems/ultrastructure , Cryoelectron Microscopy/history , Cryoelectron Microscopy/instrumentation , Electron Microscope Tomography/history , Electron Microscope Tomography/instrumentation , Fimbriae, Bacterial/metabolism , Flagella/metabolism , Flagella/ultrastructure , History, 20th Century , History, 21st Century , Models, Molecular , Nuclear Pore/metabolism , Nuclear Pore/ultrastructure , Optical Imaging/history , Optical Imaging/instrumentation , Prokaryotic Cells/metabolism , Protein Domains , Protein Structure, Secondary
2.
Nature ; 609(7926): 335-340, 2022 09.
Article in English | MEDLINE | ID: mdl-35853476

ABSTRACT

Adhesive pili assembled through the chaperone-usher pathway are hair-like appendages that mediate host tissue colonization and biofilm formation of Gram-negative bacteria1-3. Archaic chaperone-usher pathway pili, the most diverse and widespread chaperone-usher pathway adhesins, are promising vaccine and drug targets owing to their prevalence in the most troublesome multidrug-resistant pathogens1,4,5. However, their architecture and assembly-secretion process remain unknown. Here, we present the cryo-electron microscopy structure of the prototypical archaic Csu pilus that mediates biofilm formation of Acinetobacter baumannii-a notorious multidrug-resistant nosocomial pathogen. In contrast to the thick helical tubes of the classical type 1 and P pili, archaic pili assemble into an ultrathin zigzag architecture secured by an elegant clinch mechanism. The molecular clinch provides the pilus with high mechanical stability as well as superelasticity, a property observed for the first time, to our knowledge, in biomolecules, while enabling a more economical and faster pilus production. Furthermore, we demonstrate that clinch formation at the cell surface drives pilus secretion through the outer membrane. These findings suggest that clinch-formation inhibitors might represent a new strategy to fight multidrug-resistant bacterial infections.


Subject(s)
Acinetobacter baumannii , Cryoelectron Microscopy , Fimbriae, Bacterial , Molecular Chaperones , Acinetobacter baumannii/cytology , Acinetobacter baumannii/ultrastructure , Elasticity , Fimbriae Proteins/chemistry , Fimbriae Proteins/metabolism , Fimbriae Proteins/ultrastructure , Fimbriae, Bacterial/chemistry , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Chaperones/ultrastructure
3.
EMBO J ; 40(17): e106887, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34031903

ABSTRACT

Bacillus cereus sensu lato is a group of Gram-positive endospore-forming bacteria with high ecological diversity. Their endospores are decorated with micrometer-long appendages of unknown identity and function. Here, we isolate endospore appendages (Enas) from the food poisoning outbreak strain B. cereus NVH 0075-95 and find proteinaceous fibers of two main morphologies: S- and L-Ena. By using cryoEM and 3D helical reconstruction of S-Enas, we show these to represent a novel class of Gram-positive pili. S-Enas consist of single domain subunits with jellyroll topology that are laterally stacked by ß-sheet augmentation. S-Enas are longitudinally stabilized by disulfide bonding through N-terminal connector peptides that bridge the helical turns. Together, this results in flexible pili that are highly resistant to heat, drought, and chemical damage. Phylogenomic analysis reveals a ubiquitous presence of the ena-gene cluster in the B. cereus group, which include species of clinical, environmental, and food importance. We propose Enas to represent a new class of pili specifically adapted to the harsh conditions encountered by bacterial spores.


Subject(s)
Bacillus cereus/ultrastructure , Bacterial Proteins/chemistry , Fimbriae, Bacterial/ultrastructure , Bacillus cereus/genetics , Bacterial Proteins/genetics , Cryoelectron Microscopy , Fimbriae, Bacterial/chemistry , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Stability
4.
Nature ; 562(7727): 444-447, 2018 10.
Article in English | MEDLINE | ID: mdl-30283140

ABSTRACT

Pathogenic bacteria such as Escherichia coli assemble surface structures termed pili, or fimbriae, to mediate binding to host-cell receptors1. Type 1 pili are assembled via the conserved chaperone-usher pathway2-5. The outer-membrane usher FimD recruits pilus subunits bound by the chaperone FimC via the periplasmic N-terminal domain of the usher. Subunit translocation through the ß-barrel channel of the usher occurs at the two C-terminal domains (which we label CTD1 and CTD2) of this protein. How the chaperone-subunit complex bound to the N-terminal domain is handed over to the C-terminal domains, as well as the timing of subunit polymerization into the growing pilus, have previously been unclear. Here we use cryo-electron microscopy to capture a pilus assembly intermediate (FimD-FimC-FimF-FimG-FimH) in a conformation in which FimD is in the process of handing over the chaperone-bound end of the growing pilus to the C-terminal domains. In this structure, FimF has already polymerized with FimG, and the N-terminal domain of FimD swings over to bind CTD2; the N-terminal domain maintains contact with FimC-FimF, while at the same time permitting access to the C-terminal domains. FimD has an intrinsically disordered N-terminal tail that precedes the N-terminal domain. This N-terminal tail folds into a helical motif upon recruiting the FimC-subunit complex, but reorganizes into a loop to bind CTD2 during handover. Because both the N-terminal and C-terminal domains of FimD are bound to the end of the growing pilus, the structure further suggests a mechanism for stabilizing the assembly intermediate to prevent the pilus fibre diffusing away during the incorporation of thousands of subunits.


Subject(s)
Cryoelectron Microscopy , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , Escherichia coli/metabolism , Escherichia coli/ultrastructure , Fimbriae Proteins/metabolism , Fimbriae Proteins/ultrastructure , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Adhesins, Escherichia coli/chemistry , Adhesins, Escherichia coli/metabolism , Adhesins, Escherichia coli/ultrastructure , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Fimbriae Proteins/chemistry , Fimbriae, Bacterial/chemistry , Models, Molecular , Molecular Chaperones/metabolism , Protein Binding , Protein Domains , Protein Stability , Protein Subunits/chemistry , Protein Subunits/metabolism
5.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34725157

ABSTRACT

Neisseria meningitidis utilizes type IV pili (T4P) to adhere to and colonize host endothelial cells, a process at the heart of meningococcal invasive diseases leading to meningitis and sepsis. T4P are polymers of an antigenically variable major pilin building block, PilE, plus several core minor pilins that initiate pilus assembly and are thought to be located at the pilus tip. Adhesion of N. meningitidis to human endothelial cells requires both PilE and a conserved noncore minor pilin PilV, but the localization of PilV and its precise role in this process remains to be clarified. Here, we show that both PilE and PilV promote adhesion to endothelial vessels in vivo. The substantial adhesion defect observed for pilV mutants suggests it is the main adhesin. Consistent with this observation, superresolution microscopy showed the abundant distribution of PilV throughout the pilus. We determined the crystal structure of PilV and modeled it within the pilus filament. The small size of PilV causes it to be recessed relative to adjacent PilE subunits, which are dominated by a prominent hypervariable loop. Nonetheless, we identified a conserved surface-exposed adhesive loop on PilV by alanine scanning mutagenesis. Critically, antibodies directed against PilV inhibit N. meningitidis colonization of human skin grafts. These findings explain how N. meningitidis T4P undergo antigenic variation to evade the humoral immune response while maintaining their adhesive function and establish the potential of this highly conserved minor pilin as a vaccine and therapeutic target for the prevention and treatment of N. meningitidis infections.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/physiology , Fimbriae, Bacterial/physiology , Neisseria meningitidis/physiology , Animals , Antibodies/therapeutic use , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Cell Line , Drug Evaluation, Preclinical , Female , Fimbriae, Bacterial/chemistry , Fimbriae, Bacterial/ultrastructure , Humans , Meningococcal Infections/drug therapy , Mice, SCID
6.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34011607

ABSTRACT

Escherichia coli express adhesion pili that mediate attachment to host cell surfaces and are exposed to body fluids in the urinary and gastrointestinal tracts. Pilin subunits are organized into helical polymers, with a tip adhesin for specific host binding. Pili can elastically unwind when exposed to fluid flow forces, reducing the adhesin load, thereby facilitating sustained attachment. Here we investigate biophysical and structural differences of pili commonly expressed on bacteria that inhabit the urinary and intestinal tracts. Optical tweezers measurements reveal that class 1a pili of uropathogenic E. coli (UPEC), as well as class 1b of enterotoxigenic E. coli (ETEC), undergo an additional conformational change beyond pilus unwinding, providing significantly more elasticity to their structure than ETEC class 5 pili. Examining structural and steered molecular dynamics simulation data, we find that this difference in class 1 pili subunit behavior originates from an α-helical motif that can unfold when exposed to force. A disulfide bond cross-linking ß-strands in class 1 pili stabilizes subunits, allowing them to tolerate higher forces than class 5 pili that lack this covalent bond. We suggest that these extra contributions to pilus resiliency are relevant for the UPEC niche, since resident bacteria are exposed to stronger, more transient drag forces compared to those experienced by ETEC bacteria in the mucosa of the intestinal tract. Interestingly, class 1b ETEC pili include the same structural features seen in UPEC pili, while requiring lower unwinding forces that are more similar to those of class 5 ETEC pili.


Subject(s)
Adhesins, Escherichia coli/chemistry , Enterotoxigenic Escherichia coli/ultrastructure , Fimbriae Proteins/chemistry , Fimbriae, Bacterial/ultrastructure , Uropathogenic Escherichia coli/ultrastructure , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Bacterial Adhesion , Binding Sites , Biomechanical Phenomena , Cysteine/chemistry , Cysteine/metabolism , Disulfides/chemistry , Disulfides/metabolism , Enterotoxigenic Escherichia coli/genetics , Enterotoxigenic Escherichia coli/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Gene Expression , Kinetics , Molecular Dynamics Simulation , Optical Tweezers , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Thermodynamics , Uropathogenic Escherichia coli/genetics , Uropathogenic Escherichia coli/metabolism
7.
Cell ; 133(4): 640-52, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18485872

ABSTRACT

Gram-negative pathogens commonly exhibit adhesive pili on their surfaces that mediate specific attachment to the host. A major class of pili is assembled via the chaperone/usher pathway. Here, the structural basis for pilus fiber assembly and secretion performed by the outer membrane assembly platform--the usher--is revealed by the crystal structure of the translocation domain of the P pilus usher PapC and single particle cryo-electron microscopy imaging of the FimD usher bound to a translocating type 1 pilus assembly intermediate. These structures provide molecular snapshots of a twinned-pore translocation machinery in action. Unexpectedly, only one pore is used for secretion, while both usher protomers are used for chaperone-subunit complex recruitment. The translocating pore itself comprises 24 beta strands and is occluded by a folded plug domain, likely gated by a conformationally constrained beta-hairpin. These structures capture the secretion of a virulence factor across the outer membrane of gram-negative bacteria.


Subject(s)
Biosynthetic Pathways , Escherichia coli/chemistry , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/ultrastructure , Molecular Chaperones/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , Fimbriae Proteins/chemistry , Fimbriae Proteins/ultrastructure , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Porins/chemistry , Porins/metabolism
8.
Nature ; 541(7635): 112-116, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27992877

ABSTRACT

Packaging of the genome into a protein capsid and its subsequent delivery into a host cell are two fundamental processes in the life cycle of a virus. Unlike double-stranded DNA viruses, which pump their genome into a preformed capsid, single-stranded RNA (ssRNA) viruses, such as bacteriophage MS2, co-assemble their capsid with the genome; however, the structural basis of this co-assembly is poorly understood. MS2 infects Escherichia coli via the host 'sex pilus' (F-pilus); it was the first fully sequenced organism and is a model system for studies of translational gene regulation, RNA-protein interactions, and RNA virus assembly. Its positive-sense ssRNA genome of 3,569 bases is enclosed in a capsid with one maturation protein monomer and 89 coat protein dimers arranged in a T = 3 icosahedral lattice. The maturation protein is responsible for attaching the virus to an F-pilus and delivering the viral genome into the host during infection, but how the genome is organized and delivered is not known. Here we describe the MS2 structure at 3.6 Å resolution, determined by electron-counting cryo-electron microscopy (cryoEM) and asymmetric reconstruction. We traced approximately 80% of the backbone of the viral genome, built atomic models for 16 RNA stem-loops, and identified three conserved motifs of RNA-coat protein interactions among 15 of these stem-loops with diverse sequences. The stem-loop at the 3' end of the genome interacts extensively with the maturation protein, which, with just a six-helix bundle and a six-stranded ß-sheet, forms a genome-delivery apparatus and joins 89 coat protein dimers to form a capsid. This atomic description of genome-capsid interactions in a spherical ssRNA virus provides insight into genome delivery via the host sex pilus and mechanisms underlying ssRNA-capsid co-assembly, and inspires speculation about the links between nucleoprotein complexes and the origins of viruses.


Subject(s)
Capsid/ultrastructure , Cryoelectron Microscopy , Genome, Viral/physiology , Levivirus/metabolism , Levivirus/ultrastructure , RNA, Viral/ultrastructure , Virus Assembly , Capsid/chemistry , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Fimbriae, Bacterial/chemistry , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Levivirus/chemistry , Levivirus/genetics , Models, Molecular , Molecular Conformation , Protein Multimerization , RNA, Viral/chemistry , RNA, Viral/metabolism
9.
PLoS Biol ; 17(10): e3000379, 2019 10.
Article in English | MEDLINE | ID: mdl-31658249

ABSTRACT

Recent work has revealed that Clostridioides difficile, a major cause of nosocomial diarrheal disease, exhibits phenotypic heterogeneity within a clonal population as a result of phase variation. Many C. difficile strains representing multiple ribotypes develop two colony morphotypes, termed rough and smooth, but the biological implications of this phenomenon have not been explored. Here, we examine the molecular basis and physiological relevance of the distinct colony morphotypes produced by this bacterium. We show that C. difficile reversibly differentiates into rough and smooth colony morphologies and that bacteria derived from the isolates display discrete motility behaviors. We identified an atypical phase-variable signal transduction system consisting of a histidine kinase and two response regulators, named herein colony morphology regulators RST (CmrRST), which mediates the switch in colony morphology and motility behaviors. The CmrRST-regulated surface motility is independent of flagella and type IV pili, suggesting a novel mechanism of cell migration in C. difficile. Microscopic analysis of cell and colony structure indicates that CmrRST promotes the formation of elongated bacteria arranged in bundled chains, which may contribute to bacterial migration on surfaces. In a hamster model of acute C. difficile disease, the CmrRST system is required for disease development. Furthermore, we provide evidence that CmrRST phase varies during infection, suggesting that the intestinal environment impacts the proportion of CmrRST-expressing C. difficile. Our findings indicate that C. difficile employs phase variation of the CmrRST signal transduction system to generate phenotypic heterogeneity during infection, with concomitant effects on bacterial physiology and pathogenesis.


Subject(s)
Bacterial Proteins/genetics , Clostridioides difficile/metabolism , Gene Expression Regulation, Bacterial , Histidine Kinase/genetics , Signal Transduction/genetics , Animals , Bacterial Proteins/metabolism , Clone Cells , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Clostridioides difficile/ultrastructure , Clostridium Infections/microbiology , Clostridium Infections/pathology , Cricetulus , Disease Models, Animal , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Flagella/metabolism , Flagella/ultrastructure , Histidine Kinase/metabolism , Humans , Movement , Phenotype , Ribotyping
10.
Proc Natl Acad Sci U S A ; 116(28): 14222-14227, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31239340

ABSTRACT

Bacterial conjugation systems are members of the large type IV secretion system (T4SS) superfamily. Conjugative transfer of F plasmids residing in the Enterobacteriaceae was first reported in the 1940s, yet the architecture of F plasmid-encoded transfer channel and its physical relationship with the F pilus remain unknown. We visualized F-encoded structures in the native bacterial cell envelope by in situ cryoelectron tomography (CryoET). Remarkably, F plasmids encode four distinct structures, not just the translocation channel or channel-pilus complex predicted by prevailing models. The F1 structure is composed of distinct outer and inner membrane complexes and a connecting cylinder that together house the envelope-spanning translocation channel. The F2 structure is essentially the F1 complex with the F pilus attached at the outer membrane (OM). Remarkably, the F3 structure consists of the F pilus attached to a thin, cell envelope-spanning stalk, whereas the F4 structure consists of the pilus docked to the OM without an associated periplasmic density. The traffic ATPase TraC is configured as a hexamer of dimers at the cytoplasmic faces of the F1 and F2 structures, where it respectively regulates substrate transfer and F pilus biogenesis. Together, our findings present architectural renderings of the DNA conjugation or "mating" channel, the channel-pilus connection, and unprecedented pilus basal structures. These structural snapshots support a model for biogenesis of the F transfer system and allow for detailed comparisons with other structurally characterized T4SSs.


Subject(s)
Cell Membrane/ultrastructure , Escherichia coli/ultrastructure , F Factor/ultrastructure , Fimbriae, Bacterial/ultrastructure , Adenosine Triphosphatases/genetics , Bacterial Proteins/genetics , Cell Membrane/genetics , Conjugation, Genetic/genetics , Cryoelectron Microscopy , Cytoplasm/genetics , Cytoplasm/ultrastructure , DNA, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , F Factor/genetics , Fimbriae, Bacterial/genetics , Type IV Secretion Systems/genetics
11.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34830248

ABSTRACT

The endospores (spores) of many Bacillus cereus sensu lato species are decorated with multiple hair/pilus-like appendages. Although they have been observed for more than 50 years, all efforts to characterize these fibers in detail have failed until now, largely due to their extraordinary resilience to proteolytic digestion and chemical solubilization. A recent structural analysis of B. cereus endospore appendages (Enas) using cryo-electron microscopy has revealed the structure of two distinct fiber morphologies: the longer and more abundant "Staggered-type" (S-Ena) and the shorter "Ladder-like" type (L-Ena), which further enabled the identification of the genes encoding the S-Ena. Ena homologs are widely and uniquely distributed among B. cereus sensu lato species, suggesting that appendages play important functional roles in these species. The discovery of ena genes is expected to facilitate functional studies involving Ena-depleted mutant spores to explore the role of Enas in the interaction between spores and their environment. Given the importance of B. cereus spores for the food industry and in medicine, there is a need for a better understanding of their biological functions and physicochemical properties. In this review, we discuss the current understanding of the Ena structure and the potential roles these remarkable fibers may play in the adhesion of spores to biotic and abiotic surfaces, aggregation, and biofilm formation.


Subject(s)
Bacillus cereus/ultrastructure , Bacterial Proteins/chemistry , Fimbriae, Bacterial/ultrastructure , Spores, Bacterial/ultrastructure , Bacillus cereus/genetics , Bacillus cereus/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biofilms/growth & development , Cryoelectron Microscopy , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Gene Expression Regulation, Bacterial , Models, Molecular , Protein Binding , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Spores, Bacterial/genetics , Spores, Bacterial/metabolism
12.
Mol Microbiol ; 111(3): 732-749, 2019 03.
Article in English | MEDLINE | ID: mdl-30561149

ABSTRACT

Type 4a pili (T4aP) are long, thin and dynamic fibres displayed on the surface of diverse bacteria promoting adherence, motility and transport functions. Genomes of many Enterobacteriaceae contain conserved gene clusters encoding putative T4aP assembly systems. However, their expression has been observed only in few strains including Enterohaemorrhagic Escherichia coli (EHEC) and their inducers remain unknown. Here we used EHEC genomic DNA as a template to amplify and assemble an artificial operon composed of four gene clusters encoding 13 pilus assembly proteins. Controlled expressions of this operon in nonpathogenic E. coli strains led to efficient assembly of T4aP composed of the major pilin PpdD, as shown by shearing assays and immunofluorescence microscopy. When compared with PpdD pili assembled in a heterologous Klebsiella T2SS type 2 secretion system (T2SS) by using cryo-electron microscopy (cryoEM), these pili showed indistinguishable helical parameters, emphasizing that major pilins are the principal determinants of the fibre structure. Bacterial two-hybrid analysis identified several interactions of PpdD with T4aP assembly proteins, and with components of the T2SS that allow for heterologous fibre assembly. These studies lay ground for further characterization of the T4aP structure, function and biogenesis in enterobacteria.


Subject(s)
Enterohemorrhagic Escherichia coli/metabolism , Fimbriae, Bacterial/metabolism , Type IV Secretion Systems/metabolism , Cryoelectron Microscopy , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/ultrastructure , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Klebsiella/genetics , Klebsiella/metabolism , Microscopy, Fluorescence , Protein Binding , Protein Interaction Mapping , Protein Multimerization , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Two-Hybrid System Techniques , Type IV Secretion Systems/genetics , Type IV Secretion Systems/ultrastructure
13.
J Biomol NMR ; 73(6-7): 293-303, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31124002

ABSTRACT

Secretion pili, bacterial fibers responsible for transporting proteins to the extracellular milieu in some secretion systems, are very strong structures but at the same time highly flexible. Their flexibility and helical symmetry make structure determination at atomic resolution a challenging task. We have previously used an integrative structural biology approach including liquid-state NMR, cryo-electron microscopy (cryo-EM), and modeling to determine the pseudo-atomic resolution structure of the type 2 secretion system pseudopilus in a mutant form, where we employed NMR to determine the high resolution structure of the pilin (the monomer building block of the pilus). In this work, we determine the pseudo-atomic structure of the wild type pilus, and compare the dynamics of wild type and mutant pili by normal mode analysis. We present a detailed NMR analysis of the dynamics of the pilin in isolation, and compare dynamics and solvent accessibility of isolated and assembled pilins by Hydrogen/Deuterium eXchange Mass Spectrometry (HDX-MS). These complementary approaches provide a comprehensive view of internal and overall dynamics of pili, crucial for their function.


Subject(s)
Bacterial Proteins/chemistry , Fimbriae, Bacterial/chemistry , Models, Molecular , Type II Secretion Systems , Bacterial Proteins/metabolism , Cryoelectron Microscopy , Fimbriae, Bacterial/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Solvents/chemistry
14.
J Biol Chem ; 292(34): 14134-14146, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28659339

ABSTRACT

Type IV pili are important virulence factors on the surface of many pathogenic bacteria and have been implicated in a wide range of diverse functions, including attachment, twitching motility, biofilm formation, and horizontal gene transfer. The respiratory pathogen Streptococcus pneumoniae deploys type IV pili to take up DNA during transformation. These "competence pili" are composed of the major pilin protein ComGC and exclusively assembled during bacterial competence, but their biogenesis remains unclear. Here, we report the high resolution NMR structure of N-terminal truncated ComGC revealing a highly flexible and structurally divergent type IV pilin. It consists of only three α-helical segments forming a well-defined electronegative cavity and confined electronegative and hydrophobic patches. The structure is particularly flexible between the first and second α-helix with the first helical part exhibiting slightly slower dynamics than the rest of the pilin, suggesting that the first helix is involved in forming the pilus structure core and that parts of helices two and three are primarily surface-exposed. Taken together, our results provide the first structure of a type IV pilin protein involved in the formation of competence-induced pili in Gram-positive bacteria and corroborate the remarkable structural diversity among type IV pilin proteins.


Subject(s)
Fimbriae Proteins/chemistry , Fimbriae, Bacterial/ultrastructure , Models, Molecular , Streptococcus pneumoniae/physiology , Virulence Factors/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cryoelectron Microscopy , Dimerization , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Gene Deletion , Hydrophobic and Hydrophilic Interactions , Kinetics , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Microscopy, Electron, Transmission , Nuclear Magnetic Resonance, Biomolecular , Operon , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Conformation, alpha-Helical , Recombinant Fusion Proteins , Solubility , Streptococcus pneumoniae/ultrastructure , Trans-Activators/genetics , Trans-Activators/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
15.
PLoS Pathog ; 12(12): e1006109, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27992883

ABSTRACT

Type IV pilus (T4P) systems are complex molecular machines that polymerize major pilin proteins into thin filaments displayed on bacterial surfaces. Pilus functions require rapid extension and depolymerization of the pilus, powered by the assembly and retraction ATPases, respectively. A set of low abundance minor pilins influences pilus dynamics by unknown mechanisms. The Vibrio cholerae toxin-coregulated pilus (TCP) is among the simplest of the T4P systems, having a single minor pilin TcpB and lacking a retraction ATPase. Here we show that TcpB, like its homolog CofB, initiates pilus assembly. TcpB co-localizes with the pili but at extremely low levels, equivalent to one subunit per pilus. We used a micropillars assay to demonstrate that TCP are retractile despite the absence of a retraction ATPase, and that retraction relies on TcpB, as a V. cholerae tcpB Glu5Val mutant is fully piliated but does not induce micropillars movements. This mutant is impaired in TCP-mediated autoagglutination and TcpF secretion, consistent with retraction being required for these functions. We propose that TcpB initiates pilus retraction by incorporating into the growing pilus in a Glu5-dependent manner, which stalls assembly and triggers processive disassembly. These results provide a framework for understanding filament dynamics in more complex T4P systems and the closely related Type II secretion system.


Subject(s)
Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Vibrio cholerae/metabolism , Fimbriae, Bacterial/ultrastructure , Immunoblotting , Immunohistochemistry , Microscopy, Electron, Transmission , Vibrio cholerae/ultrastructure
16.
PLoS Pathog ; 12(5): e1005616, 2016 05.
Article in English | MEDLINE | ID: mdl-27159323

ABSTRACT

Enteropathogenic Escherichia coli (EPEC) represents a major causative agent of infant diarrhea associated with significant morbidity and mortality in developing countries. Although studied extensively in vitro, the investigation of the host-pathogen interaction in vivo has been hampered by the lack of a suitable small animal model. Using RT-PCR and global transcriptome analysis, high throughput 16S rDNA sequencing as well as immunofluorescence and electron microscopy, we characterize the EPEC-host interaction following oral challenge of newborn mice. Spontaneous colonization of the small intestine and colon of neonate mice that lasted until weaning was observed. Intimate attachment to the epithelial plasma membrane and microcolony formation were visualized only in the presence of a functional bundle forming pili (BFP) and type III secretion system (T3SS). Similarly, a T3SS-dependent EPEC-induced innate immune response, mediated via MyD88, TLR5 and TLR9 led to the induction of a distinct set of genes in infected intestinal epithelial cells. Infection-induced alterations of the microbiota composition remained restricted to the postnatal period. Although EPEC colonized the adult intestine in the absence of a competing microbiota, no microcolonies were observed at the small intestinal epithelium. Here, we introduce the first suitable mouse infection model and describe an age-dependent, virulence factor-dependent attachment of EPEC to enterocytes in vivo.


Subject(s)
Disease Models, Animal , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Infections/microbiology , Host-Pathogen Interactions/physiology , Animals , Animals, Newborn , Disease Susceptibility/microbiology , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Fimbriae, Bacterial/ultrastructure , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Oligonucleotide Array Sequence Analysis , Type III Secretion Systems/metabolism , Virulence Factors/metabolism
17.
J Biol Chem ; 291(20): 10602-14, 2016 May 13.
Article in English | MEDLINE | ID: mdl-26966177

ABSTRACT

N-Glycosylation is a post-translational modification common to all three domains of life. In many archaea, the oligosacharyltransferase (AglB)-dependent N-glycosylation of flagellins is required for flagella assembly. However, whether N-glycosylation is required for the assembly and/or function of the structurally related archaeal type IV pili is unknown. Here, we show that of six Haloferax volcanii adhesion pilins, PilA1 and PilA2, the most abundant pilins in pili of wild-type and ΔaglB strains, are modified under planktonic conditions in an AglB-dependent manner by the same pentasaccharide detected on H. volcanii flagellins. However, unlike wild-type cells, which have surfaces decorated with discrete pili and form a dispersed layer of cells on a plastic surface, ΔaglB cells have thick pili bundles and form microcolonies. Moreover, expressing PilA1, PilA2, or PilA6 in ΔpilA[1-6]ΔaglB stimulates microcolony formation compared with their expression in ΔpilA[1-6]. Conversely, expressing PilA3 or PilA4 in ΔpilA[1-6] cells results in strong surface adhesion, but not microcolony formation, and neither pilin stimulates surface adhesion in ΔpilA[1-6]ΔaglB cells. Although PilA4 assembles into pili in the ΔpilA[1-6]ΔaglB cells, these pili are, unlike wild-type pili, curled, perhaps rendering them non-functional. To our knowledge, this is the first demonstration of a differential effect of glycosylation on pilus assembly and function of paralogous pilins. The growth of wild-type cells in low salt media, a condition that decreases AglB glycosylation, also stimulates microcolony formation and inhibits motility, supporting our hypothesis that N-glycosylation plays an important role in regulating the transition between planktonic to sessile cell states as a response to stress.


Subject(s)
Archaeal Proteins/metabolism , Fimbriae Proteins/metabolism , Haloferax volcanii/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Cell Adhesion/physiology , Fimbriae Proteins/chemistry , Fimbriae Proteins/genetics , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Glycosylation , Haloferax volcanii/cytology , Haloferax volcanii/genetics , Polysaccharides/metabolism , Protein Multimerization , Protein Processing, Post-Translational , Tandem Mass Spectrometry
18.
PLoS Pathog ; 11(4): e1004835, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25876066

ABSTRACT

The success of S. pneumoniae as a major human pathogen is largely due to its remarkable genomic plasticity, allowing efficient escape from antimicrobials action and host immune response. Natural transformation, or the active uptake and chromosomal integration of exogenous DNA during the transitory differentiated state competence, is the main mechanism for horizontal gene transfer and genomic makeover in pneumococci. Although transforming DNA has been proposed to be captured by Type 4 pili (T4P) in Gram-negative bacteria, and a competence-inducible comG operon encoding proteins homologous to T4P-biogenesis components is present in transformable Gram-positive bacteria, a prevailing hypothesis has been that S. pneumoniae assembles only short pseudopili to destabilize the cell wall for DNA entry. We recently identified a micrometer-sized T4P-like pilus on competent pneumococci, which likely serves as initial DNA receptor. A subsequent study, however, visualized a different structure--short, 'plaited' polymers--released in the medium of competent S. pneumoniae. Biochemical observation of concurrent pilin secretion led the authors to propose that the 'plaited' structures correspond to transformation pili acting as peptidoglycan drills that leave DNA entry pores upon secretion. Here we show that the 'plaited' filaments are not related to natural transformation as they are released by non-competent pneumococci, as well as by cells with disrupted pilus biogenesis components. Combining electron microscopy visualization with structural, biochemical and proteomic analyses, we further identify the 'plaited' polymers as spirosomes: macromolecular assemblies of the fermentative acetaldehyde-alcohol dehydrogenase enzyme AdhE that is well conserved in a broad range of Gram-positive and Gram-negative bacteria.


Subject(s)
Fimbriae, Bacterial/ultrastructure , Streptococcus pneumoniae/ultrastructure , Gene Transfer, Horizontal , Macromolecular Substances/ultrastructure , Microscopy, Electron, Transmission , Polymerase Chain Reaction , Proteomics , Streptococcus pneumoniae/genetics , Transformation, Bacterial/genetics
19.
PLoS Pathog ; 11(9): e1005162, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26367394

ABSTRACT

The ability of pathogens to cause disease depends on their aptitude to escape the immune system. Type IV pili are extracellular filamentous virulence factors composed of pilin monomers and frequently expressed by bacterial pathogens. As such they are major targets for the host immune system. In the human pathogen Neisseria meningitidis, strains expressing class I pilins contain a genetic recombination system that promotes variation of the pilin sequence and is thought to aid immune escape. However, numerous hypervirulent clinical isolates express class II pilins that lack this property. This raises the question of how they evade immunity targeting type IV pili. As glycosylation is a possible source of antigenic variation it was investigated using top-down mass spectrometry to provide the highest molecular precision on the modified proteins. Unlike class I pilins that carry a single glycan, we found that class II pilins display up to 5 glycosylation sites per monomer on the pilus surface. Swapping of pilin class and genetic background shows that the pilin primary structure determines multisite glycosylation while the genetic background determines the nature of the glycans. Absence of glycosylation in class II pilins affects pilus biogenesis or enhances pilus-dependent aggregation in a strain specific fashion highlighting the extensive functional impact of multisite glycosylation. Finally, molecular modeling shows that glycans cover the surface of class II pilins and strongly decrease antibody access to the polypeptide chain. This strongly supports a model where strains expressing class II pilins evade the immune system by changing their sugar structure rather than pilin primary structure. Overall these results show that sequence invariable class II pilins are cloaked in glycans with extensive functional and immunological consequences.


Subject(s)
Endothelium, Vascular/microbiology , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Immune Evasion , Models, Molecular , Neisseria meningitidis/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Bacterial Adhesion , Cell Line , Cells, Cultured , Conserved Sequence , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Fimbriae Proteins/chemistry , Fimbriae Proteins/genetics , Fimbriae, Bacterial/immunology , Fimbriae, Bacterial/ultrastructure , Gene Deletion , Glycosylation , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/microbiology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Meningococcal Infections/immunology , Meningococcal Infections/metabolism , Meningococcal Infections/microbiology , Meningococcal Infections/pathology , Microscopy, Electron, Transmission , Neisseria meningitidis/immunology , Neisseria meningitidis/ultrastructure , Sequence Homology, Amino Acid , Species Specificity , Surface Properties
20.
Microbiol Immunol ; 61(9): 359-370, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28736993

ABSTRACT

Streptococcus pneumoniae, a common human pathogen, colonizes the nasopharynx and causes diseases including acute otitis media (AOM). Herein, pneumococcal serotype distributions in children before and after PCV7 vaccination and in patients with pneumococcal disease in Siberian Russia (Krasnoyarsk) are reported. Analyses included antimicrobial susceptibility testing, sequence typing (ST), pulsed field gel electrophoresis, virulence-related surface protein gene (VSG) typing with novel primers and structural analysis by scanning electron microscopy. In healthy children (HC) prior to administration of PCV7, drug-susceptible serotype23F/ST1500 was a major pneumococcal genotype. In the PCV7 trial, multidrug-resistant serotype19A/ST320 emerged in vaccinees after PCV7, exhibiting a PCV7-induced serotype replacement. Multidrug-resistant serotype19A/ST320 was evident in patients with AOM. Community-acquired pneumonia (CAP) isolates showed genetic similarities to the AOM (ST320) genotype, constituting a common non-invasive AOM-CAP group. In contrast, meningitis isolates were more divergent. Overall, 25 ST types were identified; five (20%) of which were Krasnoyarsk-native. Regarding VSGs, PI-1 (rlrA/rrgB), PI-2 (pitA/B), psrP and cbpA were present at 54.3%, 38.6%, 48.6%, and 95.7%, respectively, with two major VSG content types, PI-1- /PI-2- /psrP+ /cbpA+ and PI-1+ /PI-2+ /psrP- /cbpA+ , being found for HC and non-invasive diseases, respectively. A major clone of serotype19A/ST320 (PI-1+ /PI-2+ ) produced the longest pneumococcal wire (pilus) structures in colonies. ST1016 (PI-1- /PI-2- ) in HC had HEp-2 cell-adherent pili. These results suggest that serotype19A/ST320 and related genotypes, with the VSG content type PI-1+ /PI-2+ /psrP- /cbpA+ , emerged in vaccinees after PCV7 in Siberia, accompanying diseases in non-vaccinated children, and that some genotypes (serotypes19A/ST320 and 18/ST1016) produced novel pneumococcal structures, predicting their roles in colony formation and adherence.


Subject(s)
Fimbriae, Bacterial/ultrastructure , Heptavalent Pneumococcal Conjugate Vaccine/immunology , Otitis Media/epidemiology , Pneumococcal Infections/epidemiology , Streptococcus pneumoniae/classification , Bacterial Adhesion/physiology , Cell Line , Child, Preschool , Humans , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Multilocus Sequence Typing , Otitis Media/microbiology , Otitis Media/prevention & control , Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Russia/epidemiology , Siberia/epidemiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Vaccination , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL