Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
PLoS Pathog ; 16(12): e1008686, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33370399

RESUMEN

Helicobacter pylori chronically infects the stomach of approximately half of the world's population. Manifestation of clinical diseases associated with H. pylori infection, including cancer, is driven by strain properties and host responses; and as chronic infection persists, both are subject to change. Previous studies have documented frequent and extensive within-host bacterial genetic variation. To define how within-host diversity contributes to phenotypes related to H. pylori pathogenesis, this project leverages a collection of 39 clinical isolates acquired prospectively from a single subject at two time points and from multiple gastric sites. During the six years separating collection of these isolates, this individual, initially harboring a duodenal ulcer, progressed to gastric atrophy and concomitant loss of acid secretion. Whole genome sequence analysis identified 1,767 unique single nucleotide polymorphisms (SNPs) across isolates and a nucleotide substitution rate of 1.3x10-4 substitutions/site/year. Gene ontology analysis identified cell envelope genes among the genes with excess accumulation of nonsynonymous SNPs (nSNPs). A maximum likelihood tree based on genetic similarity clusters isolates from each time point separately. Within time points, there is segregation of subgroups with phenotypic differences in bacterial morphology, ability to induce inflammatory cytokines, and mouse colonization. Higher inflammatory cytokine induction in recent isolates maps to shared polymorphisms in the Cag PAI protein, CagY, while rod morphology in a subgroup of recent isolates mapped to eight mutations in three distinct helical cell shape determining (csd) genes. The presence of subgroups with unique genetic and phenotypic properties suggest complex selective forces and multiple niches within the stomach during chronic infection.


Asunto(s)
Úlcera Duodenal/microbiología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/genética , Helicobacter pylori/genética , Gastropatías/microbiología , Animales , Atrofia/microbiología , Enfermedad Crónica , Ácido Gástrico , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Gastropatías/patología
2.
J Bacteriol ; 201(14)2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31036730

RESUMEN

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.


Asunto(s)
Membrana Externa Bacteriana/química , Proteínas Bacterianas/química , Endopeptidasas/química , Genoma Bacteriano , Helicobacter pylori/citología , Helicobacter pylori/genética , Proteínas Bacterianas/genética , Pared Celular , Citoesqueleto/química , Endopeptidasas/genética , Mutación
3.
Mol Microbiol ; 110(1): 114-127, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30039535

RESUMEN

Chronic infection with Helicobacter pylori can lead to the development of gastric ulcers and stomach cancers. The helical cell shape of H. pylori promotes stomach colonization. Screens for loss of helical shape have identified several periplasmic peptidoglycan (PG) hydrolases and non-enzymatic putative scaffolding proteins, including Csd5. Both over and under expression of the PG hydrolases perturb helical shape, but the mechanism used to coordinate and localize their enzymatic activities is not known. Using immunoprecipitation and mass spectrometry we identified Csd5 interactions with cytosolic proteins CcmA, a bactofilin required for helical shape, and MurF, a PG precursor synthase, as well as the inner membrane spanning ATP synthase. A combination of Csd5 domain deletions, point mutations, and transmembrane domain chimeras revealed that the N-terminal transmembrane domain promotes MurF, CcmA, and ATP synthase interactions, while the C-terminal SH3 domain mediates PG binding. We conclude that Csd5 promotes helical shape as part of a membrane associated, multi-protein shape complex that includes interactions with the periplasmic cell wall, a PG precursor synthesis enzyme, the bacterial cytoskeleton, and ATP synthase.


Asunto(s)
Pared Celular/metabolismo , Citoesqueleto/metabolismo , Helicobacter pylori/citología , Helicobacter pylori/enzimología , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Péptido Sintasas/metabolismo , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Eliminación de Gen , Helicobacter pylori/genética , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , N-Acetil Muramoil-L-Alanina Amidasa/química , N-Acetil Muramoil-L-Alanina Amidasa/genética , Péptido Sintasas/química , Péptido Sintasas/genética , Periplasma/metabolismo , Análisis de Secuencia de Proteína
4.
J Biol Chem ; 290(6): 3622-38, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25505267

RESUMEN

Peptidoglycan modifying carboxypeptidases (CPs) are important determinants of bacterial cell shape. Here, we report crystal structures of Csd4, a three-domain protein from the human gastric pathogen Helicobacter pylori. The catalytic zinc in Csd4 is coordinated by a rare His-Glu-Gln configuration that is conserved among most Csd4 homologs, which form a distinct subfamily of CPs. Substitution of the glutamine to histidine, the residue found in prototypical zinc carboxypeptidases, resulted in decreased enzyme activity and inhibition by phosphate. Expression of the histidine variant at the native locus in a H. pylori csd4 deletion strain did not restore the wild-type helical morphology. Biochemical assays show that Csd4 can cleave a tripeptide peptidoglycan substrate analog to release m-DAP. Structures of Csd4 with this substrate analog or product bound at the active site reveal determinants of peptidoglycan specificity and the mechanism to cleave an isopeptide bond to release m-DAP. Our data suggest that Csd4 is the archetype of a new CP subfamily with a domain scheme that differs from this large family of peptide-cleaving enzymes.


Asunto(s)
Proteínas Bacterianas/química , Carboxipeptidasas/química , Glutamina/metabolismo , Helicobacter pylori/enzimología , Zinc/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Carboxipeptidasas/genética , Carboxipeptidasas/metabolismo , Glutamina/química , Glutamina/genética , Helicobacter pylori/citología , Ligandos , Datos de Secuencia Molecular , Mutación , Péptidos/metabolismo , Peptidoglicano/metabolismo , Unión Proteica
5.
Infect Immun ; 83(4): 1705-14, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25667269

RESUMEN

Dermatophytes cause superficial and cutaneous fungal infections in immunocompetent hosts and invasive disease in immunocompromised hosts. However, the host mechanisms that regulate innate immune responses against these fungi are largely unknown. Here, we utilized commercially available epidermal tissues and primary keratinocytes to assess (i) damage induction by anthropophilic, geophilic, and zoophilic dermatophyte strains and (ii) the keratinocyte signaling pathways, transcription factors, and proinflammatory responses induced by a representative dermatophyte, Trichophyton equinum. Initially, five dermatophyte species were tested for their ability to invade, cause tissue damage, and induce cytokines, with Microsporum gypseum inducing the greatest level of damage and cytokine release. Using T. equinum as a representative dermatophyte, we found that the mitogen-activated protein kinase (MAPK) pathways were predominantly affected, with increased levels of phospho-p38 and phospho-Jun N-terminal protein kinase (JNK) but decreased levels of phospho-extracellular signal-regulated kinases 1 and 2 (ERK1/2). Notably, the NF-κB and PI3K pathways were largely unaffected. T. equinum also significantly increased expression of the AP-1-associated transcription factor, c-Fos, and the MAPK regulatory phosphatase, MKP1. Importantly, the ability of T. equinum to invade, cause tissue damage, activate signaling and transcription factors, and induce proinflammatory responses correlated with germination, indicating that germination may be important for dermatophyte virulence and host immune activation.


Asunto(s)
Arthrodermataceae/inmunología , Dermatomicosis/inmunología , Queratinocitos/microbiología , Sistema de Señalización de MAP Quinasas/inmunología , Trichophyton/inmunología , Arthrodermataceae/patogenicidad , Células Cultivadas , Fosfatasa 1 de Especificidad Dual/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inmunidad Innata , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Factor de Transcripción AP-1/biosíntesis , Trichophyton/patogenicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
J Bacteriol ; 195(18): 4085-93, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23836866

RESUMEN

Natural competence is a process by which bacteria construct a membrane-associated machine for the uptake and integration of exogenous DNA. Many bacteria harbor genes for the DNA uptake machinery and yet are recalcitrant to DNA uptake for unknown reasons. For example, domesticated laboratory strains of Bacillus subtilis are renowned for high-frequency natural transformation, but the ancestral B. subtilis strain NCIB3610 is poorly competent. Here we find that endogenous plasmid pBS32 encodes a small protein, ComI, that inhibits transformation in the 3610 strain. ComI is a single-pass trans-membrane protein that appears to functionally inhibit the competence DNA uptake machinery. Functional inhibition of transformation may be common, and abolishing such inhibitors could be the key to permitting convenient genetic manipulation of a variety of industrially and medically relevant bacteria.


Asunto(s)
Bacillus subtilis/genética , Proteínas Bacterianas/farmacología , Competencia de la Transformación por ADN/efectos de los fármacos , Proteínas de la Membrana/farmacología , Plásmidos/genética , Transformación Bacteriana , Bacillus subtilis/crecimiento & desarrollo , Bacillus subtilis/metabolismo , Bacillus subtilis/fisiología , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , ADN Bacteriano/genética , Regulación Bacteriana de la Expresión Génica , Proteínas de la Membrana/genética , Reacción en Cadena de la Polimerasa , Transformación Bacteriana/efectos de los fármacos
8.
PLoS Genet ; 6(12): e1001243, 2010 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-21170308

RESUMEN

Many bacteria inhibit motility concomitant with the synthesis of an extracellular polysaccharide matrix and the formation of biofilm aggregates. In Bacillus subtilis biofilms, motility is inhibited by EpsE, which acts as a clutch on the flagella rotor to inhibit motility, and which is encoded within the 15 gene eps operon required for EPS production. EpsE shows sequence similarity to the glycosyltransferase family of enzymes, and we demonstrate that the conserved active site motif is required for EPS biosynthesis. We also screen for residues specifically required for either clutch or enzymatic activity and demonstrate that the two functions are genetically separable. Finally, we show that, whereas EPS synthesis activity is dominant for biofilm formation, both functions of EpsE synergize to stabilize cell aggregates and relieve selective pressure to abolish motility by genetic mutation. Thus, the transition from motility to biofilm formation may be governed by a single bifunctional enzyme.


Asunto(s)
Bacillus subtilis/fisiología , Proteínas Bacterianas/metabolismo , Biopelículas , Espacio Extracelular/metabolismo , Flagelos/enzimología , Glicosiltransferasas/metabolismo , Polisacáridos Bacterianos/metabolismo , Bacillus subtilis/enzimología , Bacillus subtilis/genética , Proteínas Bacterianas/genética , Espacio Extracelular/genética , Flagelos/genética , Regulación Bacteriana de la Expresión Génica , Glicosiltransferasas/genética , Operón
9.
Appl Environ Microbiol ; 78(3): 778-85, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22113911

RESUMEN

Transposons are mobile genetic elements bounded by insertion sequences that are recognized by a specific mobilizing transposase enzyme. The transposase may mobilize not only the insertion sequences but also intervening DNA. mariner is a particularly efficient transposon for the random chromosomal integration of genes and insertional mutagenesis. Here, we modify an existing mariner transposon, TnYLB, such that it can easily be genetically manipulated and introduced into Bacillus subtilis. We generate a series of three new mariner derivatives that mobilize spectinomycin, chloramphenicol, and kanamycin antibiotic resistance cassettes. Furthermore, we generate a series of transposons with a strong, outward-oriented, optionally isopropyl-ß-D-thiogalactopyranoside (IPTG)-inducible promoter for the random overexpression of neighboring genes and a series of transposons with a promoterless lacZ gene for the random generation of transcriptional reporter fusions. We note that the modification of the base transposon is not restricted to B. subtilis and should be applicable to any mariner-compatible host organism, provided that in vitro mutagenesis or an in vivo species-specific delivery vector is employed.


Asunto(s)
Bacillus subtilis/genética , Elementos Transponibles de ADN , Genética Microbiana/métodos , Mutagénesis Insercional/métodos , Fusión Artificial Génica , Farmacorresistencia Bacteriana , Genes Reporteros , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
10.
Elife ; 92020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31916938

RESUMEN

Helical cell shape is necessary for efficient stomach colonization by Helicobacter pylori, but the molecular mechanisms for generating helical shape remain unclear. The helical centerline pitch and radius of wild-type H. pylori cells dictate surface curvatures of considerably higher positive and negative Gaussian curvatures than those present in straight- or curved-rod H. pylori. Quantitative 3D microscopy analysis of short pulses with either N-acetylmuramic acid or D-alanine metabolic probes showed that cell wall growth is enhanced at both sidewall curvature extremes. Immunofluorescence revealed MreB is most abundant at negative Gaussian curvature, while the bactofilin CcmA is most abundant at positive Gaussian curvature. Strains expressing CcmA variants with altered polymerization properties lose helical shape and associated positive Gaussian curvatures. We thus propose a model where CcmA and MreB promote PG synthesis at positive and negative Gaussian curvatures, respectively, and that this patterning is one mechanism necessary for maintaining helical shape.


Round spheres, straight rods, and twisting corkscrews, bacteria come in many different shapes. The shape of bacteria is dictated by their cell wall, the strong outer barrier of the cell. As bacteria grow and multiply, they must add to their cell wall while keeping the same basic shape. The cells walls are made from long chain-like molecules via processes that are guided by protein scaffolds within the cell. Many common antibiotics, including penicillin, stop bacterial infections by interrupting the growth of cell walls. Helicobacter pylori is a common bacterium that lives in the gut and, after many years, can cause stomach ulcers and stomach cancer. H. pylori are shaped in a twisting helix, much like a corkscrew. This shape helps H. pylori to take hold and colonize the stomach. It remains unclear how H. pylori creates and maintains its helical shape. The helix is much more curved than other bacteria, and H. pylori does not have the same helpful proteins that other curved bacteria do. If H. pylori grows asymmetrically, adding more material to the cell wall on its long outer side to create a twisting helix, what controls the process? To find out, Taylor et al. grew H. pylori cells and watched how the cell walls took shape. First, a fluorescent dye was attached to the building blocks of the cell wall or to underlying proteins that were thought to help direct its growth. The cells were then imaged in 3D, and images from hundreds of cells were reconstructed to analyze the growth patterns of the bacteria's cell wall. A protein called CcmA was found most often on the long side of the twisting H. pylori. When the CcmA protein was isolated in a dish, it spontaneously formed sheets and helical bundles, confirming its role as a structural scaffold for the cell wall. When CcmA was absent from the cell of H. pylori, Taylor et al. observed that the pattern of cell growth changed substantially. This work identifies a key component directing the growth of the cell wall of H. pylori and therefore, a new target for antibiotics. Its helical shape is essential for H. pylori to infect the gut, so blocking the action of the CcmA protein may interrupt cell wall growth and prevent stomach infections.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Proteínas del Citoesqueleto/metabolismo , Helicobacter pylori/metabolismo , Alanina/metabolismo , Helicobacter pylori/citología , Ácidos Murámicos/metabolismo , Peptidoglicano/biosíntesis
11.
J Bacteriol ; 191(12): 3981-91, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19363116

RESUMEN

Biofilms are multicellular aggregates stabilized by an extracellular matrix. In Bacillus subtilis, the biofilm matrix is composed of an extracellular polysaccharide and the secreted protein TasA. Expression of both of the matrix components is repressed by the DNA-binding master regulator, SinR. Here we identify two small protein regulators of the extracellular matrix: RemA (formerly YlzA) and RemB (formerly YaaB). Mutation of RemA or RemB impairs pellicle formation, complex colony architecture, and motility inhibition in a sinR mutant background. Both proteins are required for the activation of the matrix biosynthesis operons and appear to act in parallel to SinR and two other known biofilm regulators, AbrB and DegU.


Asunto(s)
Bacillus subtilis/fisiología , Proteínas Bacterianas/metabolismo , Biopelículas , Matriz Extracelular/metabolismo , Regulación Bacteriana de la Expresión Génica , Operón , Bacillus subtilis/genética , Proteínas Bacterianas/genética , Matriz Extracelular/genética
12.
J Bacteriol ; 191(18): 5775-84, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19542270

RESUMEN

Exponentially growing populations of Bacillus subtilis contain two morphologically and functionally distinct cell types: motile individuals and nonmotile multicellular chains. Motility differentiation arises because RNA polymerase and the alternative sigma factor sigma(D) activate expression of flagellin in a subpopulation of cells. Here we demonstrate that the peptidoglycan-remodeling autolysins under sigma(D) control, LytC, LytD, and LytF, are expressed in the same subpopulation of cells that complete flagellar synthesis. Morphological heterogeneity is explained by the expression of LytF that is necessary and sufficient for cell separation. Moreover, LytC is required for motility but not at the level of cell separation or flagellum biosynthesis. Rather, LytC appears to be important for flagellar function, and motility was restored to a LytC mutant by mutation of either lonA, encoding the LonA protease, or a gene encoding a previously unannotated swarming motility inhibitor, SmiA. We conclude that heterogeneous activation of sigma(D)-dependent gene expression is sufficient to explain both the morphological heterogeneity and functional heterogeneity present in vegetative B. subtilis populations.


Asunto(s)
Bacillus subtilis/crecimiento & desarrollo , Bacillus subtilis/fisiología , Regulación Bacteriana de la Expresión Génica , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Factor sigma/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Medios de Cultivo , Elementos Transponibles de ADN , Flagelos/fisiología , Microscopía Fluorescente , Mutagénesis Insercional , N-Acetil Muramoil-L-Alanina Amidasa/genética , Factor sigma/genética
13.
Microbiol Mol Biol Rev ; 80(1): 187-203, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26864431

RESUMEN

Bacteria display an abundance of cellular forms and can change shape during their life cycle. Many plausible models regarding the functional significance of cell morphology have emerged. A greater understanding of the genetic programs underpinning morphological variation in diverse bacterial groups, combined with assays of bacteria under conditions that mimic their varied natural environments, from flowing freshwater streams to diverse human body sites, provides new opportunities to probe the functional significance of cell shape. Here we explore shape diversity among bacteria, at the levels of cell geometry, size, and surface appendages (both placement and number), as it relates to survival in diverse environments. Cell shape in most bacteria is determined by the cell wall. A major challenge in this field has been deconvoluting the effects of differences in the chemical properties of the cell wall and the resulting cell shape perturbations on observed fitness changes. Still, such studies have begun to reveal the selective pressures that drive the diverse forms (or cell wall compositions) observed in mammalian pathogens and bacteria more generally, including efficient adherence to biotic and abiotic surfaces, survival under low-nutrient or stressful conditions, evasion of mammalian complement deposition, efficient dispersal through mucous barriers and tissues, and efficient nutrient acquisition.


Asunto(s)
Bacillus subtilis/ultraestructura , Pared Celular/ultraestructura , Escherichia coli/ultraestructura , Fimbrias Bacterianas/ultraestructura , Animales , Bacillus subtilis/fisiología , Pared Celular/fisiología , Ambiente , Escherichia coli/fisiología , Fimbrias Bacterianas/fisiología , Interacciones Huésped-Patógeno/fisiología , Humanos , Consorcios Microbianos/fisiología , Viabilidad Microbiana , Movimiento/fisiología
14.
ACS Chem Biol ; 11(4): 981-91, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26735022

RESUMEN

Helicobacter pylori and Campylobacter jejuni are human pathogens and causative agents of gastric ulcers/cancer and gastroenteritis, respectively. Recent studies have uncovered a series of proteases that are responsible for maintaining the helical shape of these organisms. The H. pylori metalloprotease Csd4 and its C. jejuni homologue Pgp1 cleave the amide bond between meso-diaminopimelate and iso-d-glutamic acid in truncated peptidoglycan side chains. Deletion of either csd4 or pgp1 results in bacteria with a straight rod phenotype, a reduced ability to move in viscous media, and reduced pathogenicity. In this work, a phosphinic acid-based pseudodipeptide inhibitor was designed to act as a tetrahedral intermediate analog against the Csd4 enzyme. The phosphinic acid was shown to inhibit the cleavage of the alternate substrate, Ac-l-Ala-iso-d-Glu-meso-Dap, with a Ki value of 1.5 µM. Structural analysis of the Csd4-inhibitor complex shows that the phosphinic acid displaces the zinc-bound water and chelates the metal in a bidentate fashion. The phosphinate oxygens also interact with the key acid/base residue, Glu222, and the oxyanion-stabilizing residue, Arg86. The results are consistent with the "promoted-water pathway" mechanism for carboxypeptidase A catalysis. Studies on cultured bacteria showed that the inhibitor causes significant cell straightening when incubated with H. pylori at millimolar concentrations. A diminished, yet observable, effect on the morphology of C. jejuni was also apparent. Cell straightening was more pronounced with an acapsular C. jejuni mutant strain compared to the wild type, suggesting that the capsule impaired inhibitor accessibility. These studies demonstrate that a highly polar compound is capable of crossing the outer membrane and altering cell shape, presumably by inhibiting cell shape determinant proteases. Peptidoglycan proteases acting as cell shape determinants represent novel targets for the development of antimicrobials against these human pathogens.


Asunto(s)
Campylobacter jejuni/metabolismo , Helicobacter pylori/metabolismo
15.
Science ; 320(5883): 1636-8, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18566286

RESUMEN

Biofilms are multicellular aggregates of sessile bacteria encased by an extracellular matrix and are important medically as a source of drug-resistant microbes. In Bacillus subtilis, we found that an operon required for biofilm matrix biosynthesis also encoded an inhibitor of motility, EpsE. EpsE arrested flagellar rotation in a manner similar to that of a clutch, by disengaging motor force-generating elements in cells embedded in the biofilm matrix. The clutch is a simple, rapid, and potentially reversible form of motility control.


Asunto(s)
Bacillus subtilis/fisiología , Proteínas Bacterianas/fisiología , Biopelículas/crecimiento & desarrollo , Flagelos/fisiología , Proteínas Motoras Moleculares/fisiología , Secuencia de Aminoácidos , Bacillus subtilis/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Genes Bacterianos , Proteínas Motoras Moleculares/genética , Datos de Secuencia Molecular , Movimiento , Mutación , Operón , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA