Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cell ; 183(6): 1562-1571.e12, 2020 12 10.
Article in English | MEDLINE | ID: mdl-33306955

ABSTRACT

Ticks transmit a diverse array of microbes to vertebrate hosts, including human pathogens, which has led to a human-centric focus in this vector system. Far less is known about pathogens of ticks themselves. Here, we discover that a toxin in blacklegged ticks (Ixodes scapularis) horizontally acquired from bacteria-called domesticated amidase effector 2 (dae2)-has evolved to kill mammalian skin microbes with remarkable efficiency. Secreted into the saliva and gut of ticks, Dae2 limits skin-associated staphylococci in ticks while feeding. In contrast, Dae2 has no intrinsic ability to kill Borrelia burgdorferi, the tick-borne Lyme disease bacterial pathogen. These findings suggest ticks resist their own pathogens while tolerating symbionts. Thus, just as tick symbionts can be pathogenic to humans, mammalian commensals can be harmful to ticks. Our study underscores how virulence is context-dependent and bolsters the idea that "pathogen" is a status and not an identity.


Subject(s)
Bacteria/metabolism , Immunologic Factors/metabolism , Ixodes/physiology , Skin/microbiology , Symbiosis , Animals , Anti-Bacterial Agents/pharmacology , Biocatalysis , Cell Wall/metabolism , Feeding Behavior , Female , Gastrointestinal Tract/metabolism , Host-Pathogen Interactions , Mice , Models, Molecular , Peptidoglycan/metabolism , Phylogeny , Saliva/metabolism , Salivary Glands/metabolism , Staphylococcus epidermidis/physiology , Structural Homology, Protein , Substrate Specificity , Up-Regulation
2.
PLoS Pathog ; 19(8): e1011563, 2023 08.
Article in English | MEDLINE | ID: mdl-37585473

ABSTRACT

Trichomonas vaginalis is a human protozoan parasite that causes trichomoniasis, a prevalent sexually transmitted infection. Trichomoniasis is accompanied by a shift to a dysbiotic vaginal microbiome that is depleted of lactobacilli. Studies on co-cultures have shown that vaginal bacteria in eubiosis (e.g. Lactobacillus gasseri) have antagonistic effects on T. vaginalis pathogenesis, suggesting that the parasite might benefit from shaping the microbiome to dysbiosis (e.g. Gardnerella vaginalis among other anaerobes). We have recently shown that T. vaginalis has acquired NlpC/P60 genes from bacteria, expanding them to a repertoire of nine TvNlpC genes in two distinct clans, and that TvNlpCs of clan A are active against bacterial peptidoglycan. Here, we expand this characterization to TvNlpCs of clan B. In this study, we show that the clan organisation of NlpC/P60 genes is a feature of other species of Trichomonas, and that Histomonas meleagridis has sequences related to one clan. We characterized the 3D structure of TvNlpC_B3 alone and with the inhibitor E64 bound, probing the active site of these enzymes for the first time. Lastly, we demonstrated that TvNlpC_B3 and TvNlpC_B5 have complementary activities with the previously described TvNlpCs of clan A and that exogenous expression of these enzymes empower this mucosal parasite to take over populations of vaginal lactobacilli in mixed cultures. TvNlpC_B3 helps control populations of L. gasseri, but not of G. vaginalis, which action is partially inhibited by E64. This study is one of the first to show how enzymes produced by a mucosal protozoan parasite may contribute to a shift on the status of a microbiome, helping explain the link between trichomoniasis and vaginal dysbiosis. Further understanding of this process might have significant implications for treatments in the future.


Subject(s)
Trichomonas Infections , Trichomonas Vaginitis , Trichomonas vaginalis , Female , Humans , Trichomonas vaginalis/genetics , Lactobacillus/genetics , Peptidoglycan , N-Acetylmuramoyl-L-alanine Amidase , Dysbiosis , Bacteria
3.
Cell ; 141(5): 822-33, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20510929

ABSTRACT

The mechanisms by which bacterial cells generate helical cell shape and its functional role are poorly understood. Helical shape of the human pathogen Helicobacter pylori may facilitate penetration of the thick gastric mucus where it replicates. We identified four genes required for helical shape: three LytM peptidoglycan endopeptidase homologs (csd1-3) and a ccmA homolog. Surrounding the cytoplasmic membrane of most bacteria, the peptidoglycan (murein) sacculus is a meshwork of glycan strands joined by peptide crosslinks. Intact cells and isolated sacculi from mutants lacking any single csd gene or ccmA formed curved rods and showed increased peptidoglycan crosslinking. Quantitative morphological analyses of multiple-gene deletion mutants revealed each protein uniquely contributes to a shape-generating pathway. This pathway is required for robust colonization of the stomach in spite of normal directional motility. Our findings suggest that the coordinated action of multiple proteins relaxes peptidoglycan crosslinking, enabling helical cell curvature and twist.


Subject(s)
Helicobacter Infections/microbiology , Helicobacter pylori/cytology , Helicobacter pylori/pathogenicity , Peptidoglycan/metabolism , Stomach/microbiology , Animals , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Endopeptidases/metabolism , Female , Helicobacter pylori/enzymology , Helicobacter pylori/genetics , Metalloexopeptidases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms
4.
Cell ; 143(7): 1097-109, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21183073

ABSTRACT

Growth of the mesh-like peptidoglycan (PG) sacculus located between the bacterial inner and outer membranes (OM) is tightly regulated to ensure cellular integrity, maintain cell shape, and orchestrate division. Cytoskeletal elements direct placement and activity of PG synthases from inside the cell, but precise spatiotemporal control over this process is poorly understood. We demonstrate that PG synthases are also controlled from outside of the sacculus. Two OM lipoproteins, LpoA and LpoB, are essential for the function, respectively, of PBP1A and PBP1B, the major E. coli bifunctional PG synthases. Each Lpo protein binds specifically to its cognate PBP and stimulates its transpeptidase activity, thereby facilitating attachment of new PG to the sacculus. LpoB shows partial septal localization, and our data suggest that the LpoB-PBP1B complex contributes to OM constriction during cell division. LpoA/LpoB and their PBP-docking regions are restricted to γ-proteobacteria, providing models for niche-specific regulation of sacculus growth.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/cytology , Escherichia coli/metabolism , Peptidoglycan/biosynthesis , Bacterial Outer Membrane Proteins/chemistry , Cell Division , Cell Wall/metabolism , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Lipoproteins/chemistry , Lipoproteins/metabolism , Penicillin-Binding Proteins/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Protein Interaction Domains and Motifs
5.
PLoS Genet ; 18(3): e1010143, 2022 03.
Article in English | MEDLINE | ID: mdl-35344548

ABSTRACT

Despite extensive studies on the curve-shaped bacterium Vibrio cholerae, the causative agent of the diarrheal disease cholera, its virulence-associated regulatory two-component signal transduction system VarS/VarA is not well understood. This pathway, which mainly signals through the downstream protein CsrA, is highly conserved among gamma-proteobacteria, indicating there is likely a broader function of this system beyond virulence regulation. In this study, we investigated the VarA-CsrA signaling pathway and discovered a previously unrecognized link to the shape of the bacterium. We observed that varA-deficient V. cholerae cells showed an abnormal spherical morphology during late-stage growth. Through peptidoglycan (PG) composition analyses, we discovered that these mutant bacteria contained an increased content of disaccharide dipeptides and reduced peptide crosslinks, consistent with the atypical cellular shape. The spherical shape correlated with the CsrA-dependent overproduction of aspartate ammonia lyase (AspA) in varA mutant cells, which likely depleted the cellular aspartate pool; therefore, the synthesis of the PG precursor amino acid meso-diaminopimelic acid was impaired. Importantly, this phenotype, and the overall cell rounding, could be prevented by means of cell wall recycling. Collectively, our data provide new insights into how V. cholerae use the VarA-CsrA signaling system to adjust its morphology upon unidentified external cues in its environment.


Subject(s)
Cholera , Vibrio cholerae , Bacterial Proteins/metabolism , Cell Shape , Cholera/genetics , Cholera/microbiology , Gene Expression Regulation, Bacterial , Humans , Peptidoglycan/genetics , Peptidoglycan/metabolism , Vibrio cholerae/metabolism
6.
J Biol Chem ; 298(11): 102556, 2022 11.
Article in English | MEDLINE | ID: mdl-36183829

ABSTRACT

The type VI secretion system (T6SS) is a molecular machine that Gram-negative bacteria have adapted for multiple functions, including interbacterial competition. Bacteria use the T6SS to deliver protein effectors into adjacent cells to kill rivals and establish niche dominance. Central to T6SS-mediated bacterial competition is an arms race to acquire diverse effectors to attack and neutralize target cells. The peptidoglycan has a central role in bacterial cell physiology, and effectors that biochemically modify peptidoglycan structure effectively induce cell death. One such T6SS effector is Tlde1a from Salmonella Typhimurium. Tlde1a functions as an LD-carboxypeptidase to cleave tetrapeptide stems and as an LD-transpeptidase to exchange the terminal D-alanine of a tetrapeptide stem with a noncanonical D-amino acid. To understand how Tlde1a exhibits toxicity at the molecular level, we determined the X-ray crystal structure of Tlde1a alone and in complex with D-amino acids. Our structural data revealed that Tlde1a possesses a unique LD-transpeptidase fold consisting of a dual pocket active site with a capping subdomain. This includes an exchange pocket to bind a D-amino acid for exchange and a catalytic pocket to position the D-alanine of a tetrapeptide stem for cleavage. Our toxicity assays in Escherichia coli and in vitro peptidoglycan biochemical assays with Tlde1a variants correlate Tlde1a molecular features directly to its biochemical functions. We observe that the LD-carboxypeptidase and LD-transpeptidase activities of Tlde1a are both structurally and functionally linked. Overall, our data highlight how an LD-transpeptidase fold has been structurally altered to create a toxic effector in the T6SS arms race.


Subject(s)
Peptidyl Transferases , Type VI Secretion Systems , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism , Peptidoglycan/metabolism , Peptidyl Transferases/metabolism , Escherichia coli/metabolism , Amino Acids/metabolism , Alanine/metabolism , Bacterial Proteins/metabolism
7.
PLoS Pathog ; 17(3): e1009468, 2021 03.
Article in English | MEDLINE | ID: mdl-33788901

ABSTRACT

Peptidoglycan is the major structural component of the Staphylococcus aureus cell wall, in which it maintains cellular integrity, is the interface with the host, and its synthesis is targeted by some of the most crucial antibiotics developed. Despite this importance, and the wealth of data from in vitro studies, we do not understand the structure and dynamics of peptidoglycan during infection. In this study we have developed methods to harvest bacteria from an active infection in order to purify cell walls for biochemical analysis ex vivo. Isolated ex vivo bacterial cells are smaller than those actively growing in vitro, with thickened cell walls and reduced peptidoglycan crosslinking, similar to that of stationary phase cells. These features suggested a role for specific peptidoglycan homeostatic mechanisms in disease. As S. aureus missing penicillin binding protein 4 (PBP4) has reduced peptidoglycan crosslinking in vitro its role during infection was established. Loss of PBP4 resulted in an increased recovery of S. aureus from the livers of infected mice, which coincided with enhanced fitness within murine and human macrophages. Thicker cell walls correlate with reduced activity of peptidoglycan hydrolases. S. aureus has a family of 4 putative glucosaminidases, that are collectively crucial for growth. Loss of the major enzyme SagB, led to attenuation during murine infection and reduced survival in human macrophages. However, loss of the other three enzymes Atl, SagA and ScaH resulted in clustering dependent attenuation, in a zebrafish embryo, but not a murine, model of infection. A combination of pbp4 and sagB deficiencies resulted in a restoration of parental virulence. Our results, demonstrate the importance of appropriate cell wall structure and dynamics during pathogenesis, providing new insight to the mechanisms of disease.


Subject(s)
Cell Wall/physiology , Host-Pathogen Interactions/physiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Virulence/physiology , Animals , Mice , Peptidoglycan/metabolism , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/metabolism , Zebrafish
8.
PLoS Genet ; 16(12): e1009276, 2020 12.
Article in English | MEDLINE | ID: mdl-33370261

ABSTRACT

Rod-shape of most bacteria is maintained by the elongasome, which mediates the synthesis and insertion of peptidoglycan into the cylindrical part of the cell wall. The elongasome contains several essential proteins, such as RodA, PBP2, and the MreBCD proteins, but how its activities are regulated remains poorly understood. Using E. coli as a model system, we investigated the interactions between core elongasome proteins in vivo. Our results show that PBP2 and RodA form a complex mediated by their transmembrane and periplasmic parts and independent of their catalytic activity. MreC and MreD also interact directly with PBP2. MreC elicits a change in the interaction between PBP2 and RodA, which is suppressed by MreD. The cytoplasmic domain of PBP2 is required for this suppression. We hypothesize that the in vivo measured PBP2-RodA interaction change induced by MreC corresponds to the conformational change in PBP2 as observed in the MreC-PBP2 crystal structure, which was suggested to be the "on state" of PBP2. Our results indicate that the balance between MreC and MreD determines the activity of PBP2, which could open new strategies for antibiotic drug development.


Subject(s)
Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Membrane Proteins/metabolism , Penicillin-Binding Proteins/metabolism , Bacterial Proteins/chemistry , Binding Sites , Escherichia coli , Escherichia coli Proteins/chemistry , Membrane Proteins/chemistry , Penicillin-Binding Proteins/chemistry , Protein Binding
9.
Glycobiology ; 32(8): 712-719, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35452117

ABSTRACT

Akkermansia muciniphila is an intestinal symbiont known to improve the gut barrier function in mice and humans. Various cell envelope components have been identified to play a critical role in the immune signaling of A. muciniphila, but the chemical composition and role of peptidoglycan (PG) remained elusive. Here, we isolated PG fragments from A. muciniphila MucT (ATCC BAA-835), analyzed their composition and evaluated their immune signaling capacity. Structurally, the PG of A. muciniphila was found to be noteworthy due of the presence of some nonacetylated glucosamine residues, which presumably stems from deacetylation of N-acetylglucosamine. Some of the N-acetylmuramic acid (MurNAc) subunits were O-acetylated. The immunological assays revealed that muropeptides released from the A. muciniphila PG could both activate the intracellular NOD1 and NOD2 receptors to a comparable extent as muropeptides from Escherichia coli BW25113. These data challenge the hypothesis that non-N-acetylattion of PG can be used as a NOD-1 evasion mechanism. Our results provide new insights into the diversity of cell envelope structures of key gut microbiota members and their role in steering host-microbiome interactions.


Subject(s)
Gastrointestinal Microbiome , Peptidoglycan , Akkermansia , Animals , Humans , Mice , Verrucomicrobia/physiology
10.
Mol Microbiol ; 116(1): 329-342, 2021 07.
Article in English | MEDLINE | ID: mdl-33660879

ABSTRACT

The integrity of the cell envelope of E. coli relies on the concerted activity of multi-protein machineries that synthesize the peptidoglycan (PG) and the outer membrane (OM). Our previous work found that the depletion of lipopolysaccharide (LPS) export to the OM induces an essential PG remodeling process involving LD-transpeptidases (LDTs), the glycosyltransferase function of PBP1B and the carboxypeptidase PBP6a. Consequently, cells with defective OM biogenesis lyse if they lack any of these PG enzymes. Here we report that the morphological defects, and lysis associated with a ldtF mutant with impaired LPS transport, are alleviated by the loss of the predicted OM-anchored lipoprotein ActS (formerly YgeR). We show that ActS is an inactive member of LytM-type peptidoglycan endopeptidases due to a degenerated catalytic domain. ActS is capable of activating all three main periplasmic peptidoglycan amidases, AmiA, AmiB, and AmiC, which were previously reported to be activated only by EnvC and/or NlpD. Our data also suggest that in vivo ActS preferentially activates AmiC and that its function is linked to cell envelope stress.


Subject(s)
Bacterial Outer Membrane/physiology , Carboxypeptidases/metabolism , Endopeptidases/metabolism , Escherichia coli/metabolism , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Carboxypeptidases/genetics , Cell Membrane/physiology , Cell Wall/metabolism , Endopeptidases/genetics , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Gene Deletion , Lipopolysaccharides/metabolism , N-Acetylmuramoyl-L-alanine Amidase/genetics , Penicillin-Binding Proteins/metabolism , Peptidoglycan/metabolism , Peptidoglycan Glycosyltransferase/metabolism , Plasmids/genetics , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Stress, Physiological/physiology
11.
PLoS Genet ; 15(2): e1007897, 2019 02.
Article in English | MEDLINE | ID: mdl-30707707

ABSTRACT

Many bacteria have complex cell shapes, but the mechanisms producing their distinctive morphologies are still poorly understood. Caulobacter crescentus, for instance, exhibits a stalk-like extension that carries an adhesive holdfast mediating surface attachment. This structure forms through zonal peptidoglycan biosynthesis at the old cell pole and elongates extensively under phosphate-limiting conditions. We analyzed the composition of cell body and stalk peptidoglycan and identified significant differences in the nature and proportion of peptide crosslinks, indicating that the stalk represents a distinct subcellular domain with specific mechanical properties. To identify factors that participate in stalk formation, we systematically inactivated and localized predicted components of the cell wall biosynthetic machinery of C. crescentus. Our results show that the biosynthesis of stalk peptidoglycan involves a dedicated peptidoglycan biosynthetic complex that combines specific components of the divisome and elongasome, suggesting that the repurposing of preexisting machinery provides a straightforward means to evolve new morphological traits.


Subject(s)
Bacterial Proteins/metabolism , Caulobacter crescentus/metabolism , Cell Wall/metabolism , Peptidoglycan/biosynthesis , Phosphates/metabolism
12.
Proc Natl Acad Sci U S A ; 116(27): 13498-13507, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209025

ABSTRACT

Lyme disease is a multisystem disorder caused by the spirochete Borrelia burgdorferi A common late-stage complication of this disease is oligoarticular arthritis, often involving the knee. In ∼10% of cases, arthritis persists after appropriate antibiotic treatment, leading to a proliferative synovitis typical of chronic inflammatory arthritides. Here, we provide evidence that peptidoglycan (PG), a major component of the B. burgdorferi cell envelope, may contribute to the development and persistence of Lyme arthritis (LA). We show that B. burgdorferi has a chemically atypical PG (PGBb) that is not recycled during cell-wall turnover. Instead, this pathogen sheds PGBb fragments into its environment during growth. Patients with LA mount a specific immunoglobulin G response against PGBb, which is significantly higher in the synovial fluid than in the serum of the same patient. We also detect PGBb in 94% of synovial fluid samples (32 of 34) from patients with LA, many of whom had undergone oral and intravenous antibiotic treatment. These same synovial fluid samples contain proinflammatory cytokines, similar to those produced by human peripheral blood mononuclear cells stimulated with PGBb In addition, systemic administration of PGBb in BALB/c mice elicits acute arthritis. Altogether, our study identifies PGBb as a likely contributor to inflammatory responses in LA. Persistence of this antigen in the joint may contribute to synovitis after antibiotics eradicate the pathogen. Furthermore, our finding that B. burgdorferi sheds immunogenic PGBb fragments during growth suggests a potential role for PGBb in the immunopathogenesis of other Lyme disease manifestations.


Subject(s)
Antigens, Bacterial/immunology , Borrelia burgdorferi/immunology , Lyme Disease/immunology , Peptidoglycan/immunology , Adaptive Immunity/immunology , Animals , Cytokines/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Peptidoglycan/analysis , Peptidoglycan/chemistry , Synovial Fluid/chemistry , Synovial Fluid/immunology
13.
Nature ; 518(7537): 98-101, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25470067

ABSTRACT

Horizontal gene transfer allows organisms to rapidly acquire adaptive traits. Although documented instances of horizontal gene transfer from bacteria to eukaryotes remain rare, bacteria represent a rich source of new functions potentially available for co-option. One benefit that genes of bacterial origin could provide to eukaryotes is the capacity to produce antibacterials, which have evolved in prokaryotes as the result of eons of interbacterial competition. The type VI secretion amidase effector (Tae) proteins are potent bacteriocidal enzymes that degrade the cell wall when delivered into competing bacterial cells by the type VI secretion system. Here we show that tae genes have been transferred to eukaryotes on at least six occasions, and that the resulting domesticated amidase effector (dae) genes have been preserved for hundreds of millions of years through purifying selection. We show that the dae genes acquired eukaryotic secretion signals, are expressed within recipient organisms, and encode active antibacterial toxins that possess substrate specificity matching extant Tae proteins of the same lineage. Finally, we show that a dae gene in the deer tick Ixodes scapularis limits proliferation of Borrelia burgdorferi, the aetiologic agent of Lyme disease. Our work demonstrates that a family of horizontally acquired toxins honed to mediate interbacterial antagonism confers previously undescribed antibacterial capacity to eukaryotes. We speculate that the selective pressure imposed by competition between bacteria has produced a reservoir of genes encoding diverse antimicrobial functions that are tailored for co-option by eukaryotic innate immune systems.


Subject(s)
Bacteria/enzymology , Bacteria/genetics , Bacterial Toxins/genetics , Eukaryota/genetics , Eukaryota/immunology , Gene Transfer, Horizontal/genetics , Genes, Bacterial/genetics , Immunity, Innate , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Bacteria/cytology , Bacteria/immunology , Bacterial Secretion Systems , Bacterial Toxins/metabolism , Borrelia burgdorferi/cytology , Borrelia burgdorferi/growth & development , Borrelia burgdorferi/immunology , Cell Wall/metabolism , Conserved Sequence/genetics , Eukaryota/metabolism , Immunity, Innate/genetics , Ixodes/genetics , Ixodes/immunology , Ixodes/metabolism , Ixodes/microbiology , Phylogeny , Substrate Specificity
14.
Mol Microbiol ; 112(1): 280-301, 2019 07.
Article in English | MEDLINE | ID: mdl-31070821

ABSTRACT

Campylobacter jejuni is a prevalent enteric pathogen that changes morphology from helical to coccoid under unfavorable conditions. Bacterial peptidoglycan maintains cell shape. As C. jejuni transformed from helical to coccoid, peptidoglycan dipeptides increased and tri- and tetrapeptides decreased. The DL-carboxypeptidase Pgp1 important for C. jejuni helical morphology and putative N-acetylmuramoyl-L-alanyl amidase AmiA were both involved in the coccoid transition. Mutants in pgp1 and amiA showed reduced coccoid formation, with ∆pgp1∆amiA producing minimal coccoids. Both ∆amiA and ∆amiA∆pgp1 lacked flagella and formed unseparated chains of cells consistent with a role for AmiA in cell separation. All strains accumulated peptidoglycan dipeptides over time, but only strains capable of becoming coccoid displayed tripeptide changes. C. jejuni helical shape and corresponding peptidoglycan structure are important for pathogenesis-related attributes. Concomitantly, changing to a coccoid morphology resulted in differences in pathogenic properties; coccoid C. jejuni were non-motile and non-infectious, with minimal adherence and invasion of epithelial cells and an inability to stimulate IL-8. Coccoid peptidoglycan exhibited reduced activation of innate immune receptors Nod1 and Nod2 versus helical peptidoglycan. C. jejuni also transitioned to coccoid within epithelial cells, so the inability of the immune system to detect coccoid C. jejuni may be significant in its pathogenesis.


Subject(s)
Campylobacter jejuni/metabolism , Cell Shape/physiology , Peptidoglycan/metabolism , Bacterial Proteins/metabolism , Campylobacter Infections/microbiology , Campylobacter jejuni/pathogenicity , Campylobacter jejuni/physiology , Carboxypeptidases/metabolism , Cell Wall/metabolism , Peptidoglycan/chemistry , Peptidoglycan/immunology
15.
J Bacteriol ; 201(14)2019 07 15.
Article in English | MEDLINE | ID: mdl-31036730

ABSTRACT

Evident in its name, the gastric pathogen Helicobacter pylori has a helical cell morphology which facilitates efficient colonization of the human stomach. An improved light-focusing strategy allowed us to robustly distinguish even subtle perturbations of H. pylori cell morphology by deviations in light-scattering properties measured by flow cytometry. Profiling of an arrayed genome-wide deletion library identified 28 genes that influence different aspects of cell shape, including properties of the helix, cell length or width, cell filament formation, cell shape heterogeneity, and cell branching. Included in this mutant collection were two that failed to form any helical cells, a soluble lytic transglycosylase and a previously uncharacterized putative multipass inner membrane protein HPG27_0728, renamed Csd7. A combination of cell fractionation, mutational, and immunoprecipitation experiments show that Csd7 and Csd2 collaborate to stabilize the Csd1 peptidoglycan (PG) endopeptidase. Thus, both csd2 and csd7 mutants show the same enhancement of PG tetra-pentapeptide cross-linking as csd1 mutants. Csd7 also links Csd1 with the bactofilin CcmA via protein-protein interactions. Although Csd1 is stable in ccmA mutants, these mutants show altered PG tetra-pentapeptide cross-linking, suggesting that Csd7 may directly or indirectly activate as well as stabilize Csd1. These data begin to illuminate a highly orchestrated program to regulate PG modifications that promote helical shape, which includes nine nonessential nonredundant genes required for helical shape and 26 additional genes that further modify H. pylori's cell morphology.IMPORTANCE The stomach ulcer and cancer-causing pathogen Helicobacter pylori has a helical cell shape which facilitates stomach infection. Using light scattering to measure perturbations of cell morphology, we identified 28 genes that influence different aspects of cell shape. A mutant in a previously uncharacterized protein renamed Csd7 failed to form any helical cells. Biochemical analyses showed that Csd7 collaborates with other proteins to stabilize the cell wall-degrading enzyme Csd1. Csd7 also links Csd1 with a putative filament-forming protein via protein-protein interactions. These data suggest that helical cell shape arises from a highly orchestrated program to regulate cell wall modifications. Targeting of this helical cell shape-promoting program could offer new ways to block infectivity of this important human pathogen.


Subject(s)
Bacterial Outer Membrane/chemistry , Bacterial Proteins/chemistry , Endopeptidases/chemistry , Genome, Bacterial , Helicobacter pylori/cytology , Helicobacter pylori/genetics , Bacterial Proteins/genetics , Cell Wall , Cytoskeleton/chemistry , Endopeptidases/genetics , Mutation
16.
Mol Microbiol ; 110(3): 390-410, 2018 11.
Article in English | MEDLINE | ID: mdl-30066424

ABSTRACT

Clostridium difficile remains the leading cause of antibiotic-associated diarrhoea in hospitals worldwide, linked to significant morbidity and mortality. As a strict anaerobe, it produces dormant cell forms - spores - which allow it to survive in the aerobic environment. Importantly, spores are the transmission agent of C. difficile infections. A key aspect of sporulation is the engulfment of the future spore by the mother cell and several proteins have been proposed to be involved. Here, we investigated the role of the SpoIID, SpoIIM and SpoIIP (DMP) machinery and its interplay with the SpoIIQ:SpoIIIAH (Q:AH) complex in C. difficile. We show that, surprisingly, SpoIIM, the proposed machinery anchor, is not required for efficient engulfment and sporulation. We demonstrate the requirement of DP for engulfment due to their sequential peptidoglycan degradation activity, both in vitro and in vivo. Finally, new interactions within DMP and between DMP and Q:AH suggest that both systems form a single engulfment machinery to keep the mother cell and forespore membranes together throughout engulfment. This work sheds new light upon the engulfment process and on how different sporeformers might use the same components in different ways to drive spore formation.


Subject(s)
Clostridioides difficile/enzymology , Clostridioides difficile/growth & development , Endopeptidases/metabolism , Peptidoglycan/metabolism , Phosphoric Monoester Hydrolases/metabolism , Spores, Bacterial/enzymology , Spores, Bacterial/growth & development , Endopeptidases/genetics , Hydrolysis , Phosphoric Monoester Hydrolases/genetics , Protein Interaction Maps
17.
Biochem J ; 475(2): 415-428, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29229757

ABSTRACT

The Gram-negative bacterium Serratia marcescens secretes many proteins that are involved in extracellular chitin degradation. This so-called chitinolytic machinery includes three types of chitinase enzymes and a lytic polysaccharide monooxygenase. An operon has been identified in S. marcescens, chiWXYZ, that is thought to be involved in the secretion of the chitinolytic machinery. Genetic evidence points to the ChiX protein being a key player in the secretion mechanism, since deletion of the chiX gene in S. marcescens led to a mutant strain blocked for secretion of all members of the chitinolytic machinery. In this work, a detailed structural and biochemical characterisation of ChiX is presented. The high-resolution crystal structure of ChiX reveals the protein to be a member of the LAS family of peptidases. ChiX is shown to be a zinc-containing metalloenzyme, and in vitro assays demonstrate that ChiX is an l-Ala d-Glu endopeptidase that cleaves the cross-links in bacterial peptidoglycan. This catalytic activity is shown to be intimately linked with the secretion of the chitinolytic machinery, since substitution of the ChiX Asp-120 residue results in a variant protein that is both unable to digest peptidoglycan and cannot rescue the phenoytype of a chiX mutant strain.


Subject(s)
Bacterial Proteins/chemistry , Chitinases/metabolism , Gene Expression Regulation, Bacterial , N-Acetylmuramoyl-L-alanine Amidase/chemistry , Peptidoglycan/chemistry , Serratia marcescens/enzymology , Zinc/chemistry , Amino Acid Motifs , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Chitin/metabolism , Chitinases/genetics , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hydrolysis , Models, Molecular , N-Acetylmuramoyl-L-alanine Amidase/genetics , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Operon , Peptidoglycan/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serratia marcescens/genetics , Substrate Specificity , Zinc/metabolism
18.
Proc Natl Acad Sci U S A ; 113(33): 9162-70, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27506799

ABSTRACT

Agents that cause Lyme disease, relapsing fever, leptospirosis, and syphilis belong to the phylum Spirochaetae-a unique lineage of bacteria most known for their long, spiral morphology. Despite the relevance to human health, little is known about the most fundamental aspects of spirochete growth. Here, using quantitative microscopy to track peptidoglycan cell-wall synthesis, we found that the Lyme disease spirochete Borrelia burgdorferi displays a complex pattern of growth. B. burgdorferi elongates from discrete zones that are both spatially and temporally regulated. In addition, some peptidoglycan incorporation occurs along the cell body, with the notable exception of a large region at the poles. Newborn cells inherit a highly active zone of peptidoglycan synthesis at midcell that contributes to elongation for most of the cell cycle. Concomitant with the initiation of nucleoid separation and cell constriction, second and third zones of elongation are established at the 1/4 and 3/4 cellular positions, marking future sites of division for the subsequent generation. Positioning of elongation zones along the cell is robust to cell length variations and is relatively precise over long distances (>30 µm), suggesting that cells ?sense" relative, as opposed to absolute, cell length to establish zones of peptidoglycan synthesis. The transition from one to three zones of peptidoglycan growth during the cell cycle is also observed in relapsing fever Borrelia. However, this mode of growth does not extend to representative species from other spirochetal genera, suggesting that this distinctive growth mode represents an evolutionary divide in the spirochete phylum.


Subject(s)
Borrelia/growth & development , Lyme Disease/microbiology , Peptidoglycan/biosynthesis , Relapsing Fever/microbiology , Borrelia burgdorferi/growth & development , Cell Cycle , Humans
19.
Mol Microbiol ; 104(6): 948-971, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28316093

ABSTRACT

Campylobacter jejuni helical shape is important for colonization and host interactions with straight mutants having altered biological properties. Passage on calcofluor white (CFW) resulted in C. jejuni 81-176 isolates with morphology changes: either a straight morphology from frameshift mutations and single nucleotide polymorphisms in peptidoglycan hydrolase genes pgp1 or pgp2 or a reduction in curvature due a frameshift mutation in cjj81176_1105, a putative peptidoglycan endopeptidase. Shape defects were restored by complementation. Whole genome sequencing of CFW-passaged strains showed no specific changes correlating to CFW exposure. The cjj81176_1279 (recR; recombinational DNA repair) and cjj81176_1449 (unknown function) genes were highly variable in all 81-176 strains sequenced. A frameshift mutation in pgp1 of our laboratory isolate of the straight genome sequenced variant of 11168 (11168-GS) was also identified. The PG muropeptide profile of 11168-GS was identical to that of Δpgp1 in the original minimally passaged 11168 strain (11168-O). Introduction of wild type pgp1 into 11168-GS did not restore helical morphology. The recR gene was also highly variable in 11168 strains. Microbial cell-to-cell heterogeneity is proposed as a mechanism of ensuring bacterial survival in sub-optimal conditions. In certain environments, changes in C. jejuni morphology due to genetic heterogeneity may promote C. jejuni survival.


Subject(s)
Campylobacter jejuni/cytology , Campylobacter jejuni/genetics , Bacterial Proteins/metabolism , Benzenesulfonates , Campylobacter Infections/microbiology , Clone Cells , Gene Expression Regulation, Bacterial/genetics , Peptidoglycan/metabolism
20.
Int J Mol Sci ; 19(6)2018 May 30.
Article in English | MEDLINE | ID: mdl-29848957

ABSTRACT

Burkholderia cepacia complex (BCC) bacteria are a group of opportunistic pathogens that cause severe lung infections in cystic fibrosis (CF). Treatment of BCC infections is difficult, due to the inherent and acquired multidrug resistance of BCC. There is a pressing need to find new bacterial targets for antimicrobials. Here, we demonstrate that the novel compound Q22, which is related to the bacterial cytoskeleton destabilising compound A22, can reduce the growth rate and inhibit growth of BCC bacteria. We further analysed the phenotypic effects of Q22 treatment on BCC virulence traits, to assess its feasibility as an antimicrobial. BCC bacteria were grown in the presence of Q22 with a broad phenotypic analysis, including resistance to H2O2-induced oxidative stress, changes in the inflammatory potential of cell surface components, and in-vivo drug toxicity studies. The influence of the Q22 treatment on inflammatory potential was measured by monitoring the cytokine responses of BCC whole cell lysates, purified lipopolysaccharide, and purified peptidoglycan extracted from bacterial cultures grown in the presence or absence of Q22 in differentiated THP-1 cells. BCC bacteria grown in the presence of Q22 displayed varying levels of resistance to H2O2-induced oxidative stress, with some strains showing increased resistance after treatment. There was strain-to-strain variation in the pro-inflammatory ability of bacterial lysates to elicit TNFα and IL-1ß from human myeloid cells. Despite minimal toxicity previously shown in vitro with primary CF cell lines, in-vivo studies demonstrated Q22 toxicity in both zebrafish and mouse infection models. In summary, destabilisation of the bacterial cytoskeleton in BCC, using compounds such as Q22, led to increased virulence-related traits in vitro. These changes appear to vary depending on strain and BCC species. Future development of antimicrobials targeting the BCC bacterial cytoskeleton may be hampered if such effects translate into the in-vivo environment of the CF infection.


Subject(s)
Anti-Infective Agents/therapeutic use , Burkholderia cepacia complex/metabolism , Burkholderia cepacia complex/pathogenicity , Cytoskeleton/metabolism , Animals , Burkholderia Infections/drug therapy , Burkholderia Infections/prevention & control , Disease Models, Animal , Humans , Mice , THP-1 Cells , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL