Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Mol Microbiol ; 90(4): 869-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24112477

ABSTRACT

The helical cell shape of Helicobacter pylori is highly conserved and contributes to its ability to swim through and colonize the viscous gastric mucus layer. A multi-faceted peptidoglycan (PG) modification programme involving four recently characterized peptidases and two accessory proteins is essential for maintaining H. pylori's helicity. To expedite identification of additional shape-determining genes, we employed flow cytometry with fluorescence-activated cell sorting (FACS) to enrich a transposon library for bacterial cells with altered light scattering profiles that correlate with perturbed cell morphology. After a single round of sorting, 15% of our clones exhibited a stable cell shape defect, reflecting 37-fold enrichment. Sorted clones with straight rod morphology contained insertions in known PG peptidases, as well as an insertion in csd6, which we demonstrated has ld-carboxypeptidase activity and cleaves monomeric tetrapeptides in the PG sacculus, yielding tripeptides. Other mutants had only slight changes in helicity due to insertions in genes encoding MviN/MurJ, a protein possibly involved in initiating PG synthesis, and the hypothetical protein HPG27_782. Our findings demonstrate FACS robustly detects perturbations of bacterial cell shape and identify additional PG peptide modifications associated with helical cell shape in H. pylori.


Subject(s)
Bacterial Proteins/metabolism , Genes, Bacterial , Helicobacter pylori/cytology , Helicobacter pylori/genetics , Bacterial Proteins/genetics , Biological Evolution , Carboxypeptidases/genetics , Carboxypeptidases/metabolism , Cell Movement , Cell Wall/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Flow Cytometry , Helicobacter pylori/enzymology , Mutation , Peptidoglycan/metabolism
3.
Trends Microbiol ; 20(11): 540-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22944244

ABSTRACT

The peptidoglycan cell wall maintains turgor pressure and cell shape of most bacteria. Cell wall hydrolases are essential, together with synthases, for growth and daughter cell separation. Recent work in diverse organisms has uncovered new cell wall hydrolases that act autonomously or on neighboring cells to modulate invasion of prey cells, cell shape, innate immune detection, intercellular communication, and competitor lysis. The hydrolases involved in these processes catalyze the cleavage of bonds throughout the sugar and peptide moities of peptidoglycan. Phenotypes associated with these diverse hydrolases reveal new functions of the bacterial cell wall beyond growth and division.


Subject(s)
Bacteria/enzymology , Bacteria/metabolism , Cell Wall/enzymology , Hydrolases/metabolism , Glycopeptides/metabolism , Hydrolysis
4.
PLoS Pathog ; 8(3): e1002603, 2012.
Article in English | MEDLINE | ID: mdl-22457625

ABSTRACT

Helical cell shape of the gastric pathogen Helicobacter pylori has been suggested to promote virulence through viscosity-dependent enhancement of swimming velocity. However, H. pylori csd1 mutants, which are curved but lack helical twist, show normal velocity in viscous polymer solutions and the reason for their deficiency in stomach colonization has remained unclear. Characterization of new rod shaped mutants identified Csd4, a DL-carboxypeptidase of peptidoglycan (PG) tripeptide monomers and Csd5, a putative scaffolding protein. Morphological and biochemical studies indicated Csd4 tripeptide cleavage and Csd1 crosslinking relaxation modify the PG sacculus through independent networks that coordinately generate helical shape. csd4 mutants show attenuation of stomach colonization, but no change in proinflammatory cytokine induction, despite four-fold higher levels of Nod1-agonist tripeptides in the PG sacculus. Motility analysis of similarly shaped mutants bearing distinct alterations in PG modifications revealed deficits associated with shape, but only in gel-like media and not viscous solutions. As gastric mucus displays viscoelastic gel-like properties, our results suggest enhanced penetration of the mucus barrier underlies the fitness advantage conferred by H. pylori's characteristic shape.


Subject(s)
Bacterial Physiological Phenomena , Bacterial Proteins/metabolism , Cell Wall/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori , Peptidoglycan/metabolism , Animals , Bacterial Physiological Phenomena/genetics , Cell Wall/genetics , Disease Models, Animal , Female , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Helicobacter Infections/metabolism , Helicobacter pylori/cytology , Helicobacter pylori/pathogenicity , Helicobacter pylori/physiology , Mice , Mice, Inbred C57BL , Movement , Mucus/metabolism , Mucus/microbiology , Mutation
5.
J Mol Biol ; 343(5): 1379-89, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15491619

ABSTRACT

Characterization of protein-protein interactions that are critical to the specific function of many biological systems has become a primary goal of structural biology research. Analysis of these interactions by structural techniques is, however, challenging due to inherent limitations of the techniques and because many of the interactions are transient, and suitable complexes are difficult to isolate. In particular, structural studies of large protein complexes by traditional solution NMR methods are difficult due to a priori requirement of extensive assignments and a large number of intermolecular restraints for the complex. An approach overcoming some of these challenges by utilizing orientational restraints from residual dipolar couplings collected on solution NMR samples is presented. The approach exploits existing structures of individual components, including the symmetry properties of some of these structures, to assemble rapidly models for relatively large protein-protein complexes. An application is illustrated with a 95 kDa homotrimeric complex of the acyltransferase protein, LpxA (UDP-N-acetylglucosamine acyltransferase), and acyl carrier protein. LpxA catalyzes the first step in the biosynthesis of the lipid A component of lipopolysaccharide in Gram-negative bacteria. The structural model generated for this complex can be useful in the design of new anti-bacterial agents that inhibit the biosynthesis of lipid A.


Subject(s)
Acyl Carrier Protein/chemistry , Acyltransferases/chemistry , Acyl Carrier Protein/metabolism , Acyltransferases/metabolism , Computational Biology , Crystallography, X-Ray , Escherichia coli/enzymology , Escherichia coli/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Structure, Tertiary
6.
Proc Natl Acad Sci U S A ; 100(13): 7907-12, 2003 Jun 24.
Article in English | MEDLINE | ID: mdl-12810959

ABSTRACT

The bacterium Pseudomonas aeruginosa causes chronic respiratory infections in cystic fibrosis (CF) patients. Such infections are extremely difficult to control because the bacteria exhibit a biofilm-mode of growth, rendering P. aeruginosa resistant to antibiotics and phagocytic cells. During the course of infection, P. aeruginosa usually undergoes a phenotypic switch to a mucoid colony, which is characterized by the overproduction of the exopolysaccharide alginate. Alginate overproduction has been implicated in protecting P. aeruginosa from the harsh environment present in the CF lung, as well as facilitating its persistence as a biofilm by providing an extracellular matrix that promotes adherence. Because of its association with biofilms in CF patients, it has been assumed that alginate is also the primary exopolysaccharide expressed in biofilms of environmental nonmucoid P. aeruginosa. In this study, we examined the chemical nature of the biofilm matrix produced by wild-type and isogenic alginate biosynthetic mutants of P. aeruginosa. The results clearly indicate that alginate biosynthetic genes are not expressed and that alginate is not required during the formation of nonmucoid biofilms in two P. aeruginosa strains, PAO1 and PA14, that have traditionally been used to study biofilms. Because nonmucoid P. aeruginosa strains are the predominant environmental phenotype and are also involved in the initial colonization in CF patients, these studies have implications in understanding the early events of the infectious process in the CF airway.


Subject(s)
Alginates/chemistry , Alginates/metabolism , Dioxygenases , Extracellular Matrix/chemistry , Polysaccharides/chemistry , Pseudomonas aeruginosa/metabolism , Carbohydrate Dehydrogenases/metabolism , Carbohydrate Metabolism , Catechol 2,3-Dioxygenase , Cell Division , Drug Resistance, Microbial , Glucuronic Acid , Hexuronic Acids , Operon , Oxygenases/genetics , Plasmids/metabolism , Species Specificity , Time Factors , Uronic Acids/metabolism
7.
Microbiology (Reading) ; 148(Pt 11): 3423-3430, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12427934

ABSTRACT

When mucoid (alginate-producing) Pseudomonas aeruginosa FRD1 is grown under low oxygen conditions in liquid culture (static), non-mucoid variants appear and eventually predominate. This conversion is not readily observed in aerobic, shaken cultures or static cultures containing the alternative electron acceptor nitrate. In this study, it is shown that the non-mucoid variants that arise under static growth conditions are almost exclusively algT mutants. It has been shown that AlgT not only positively regulates alginate biosynthesis, but also directly or indirectly negatively regulates flagellum synthesis. Indeed, during static growth, conversion to the non-mucoid phenotype is accompanied by the acquisition of flagellum-mediated motility. Surprisingly, by using a reporter gene fusion with the fliC promoter (pfliC::xylE), it was found that fliC expression begins within hours of static growth and is reversible after returning the culture to shaking conditions. The ability of the strain to produce alginate seems to be irrelevant to this phenomenon, as an AlgT(+) deltaalgD strain showed identical results. Thus, it is suggested that the first effect of static growth is to induce motility as an adaptive measure in the presence of wild-type algT. This may afford P. aeruginosa the ability to swim towards areas of higher oxygen concentrations. Subsequent to this, algT mutations are likely to secure the motile phenotype.


Subject(s)
Flagella/physiology , Movement/physiology , Pseudomonas aeruginosa/growth & development , Alginates/metabolism , Flagellin/genetics , Flagellin/metabolism , Glucuronic Acid , Hexuronic Acids , Oxygen/metabolism , Phenotype , Pseudomonas aeruginosa/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...