Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Intervalo de año de publicación
1.
PLoS Biol ; 22(3): e3002558, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38478588

RESUMEN

Polyphosphates (polyP) are chains of inorganic phosphates that can reach over 1,000 residues in length. In Escherichia coli, polyP is produced by the polyP kinase (PPK) and is thought to play a protective role during the response to cellular stress. However, the molecular pathways impacted by PPK activity and polyP accumulation remain poorly characterized. In this work, we used label-free mass spectrometry to study the response of bacteria that cannot produce polyP (Δppk) during starvation to identify novel pathways regulated by PPK. In response to starvation, we found 92 proteins significantly differentially expressed between wild-type and Δppk mutant cells. Wild-type cells were enriched for proteins related to amino acid biosynthesis and transport, while Δppk mutants were enriched for proteins related to translation and ribosome biogenesis, suggesting that without PPK, cells remain inappropriately primed for growth even in the absence of the required building blocks. From our data set, we were particularly interested in Arn and EptA proteins, which were down-regulated in Δppk mutants compared to wild-type controls, because they play a role in lipid A modifications linked to polymyxin resistance. Using western blotting, we confirm differential expression of these and related proteins in K-12 strains and a uropathogenic isolate, and provide evidence that this mis-regulation in Δppk cells stems from a failure to induce the BasRS two-component system during starvation. We also show that Δppk mutants unable to up-regulate Arn and EptA expression lack the respective L-Ara4N and pEtN modifications on lipid A. In line with this observation, loss of ppk restores polymyxin sensitivity in resistant strains carrying a constitutively active basR allele. Overall, we show a new role for PPK in lipid A modification during starvation and provide a rationale for targeting PPK to sensitize bacteria towards polymyxin treatment. We further anticipate that our proteomics work will provide an important resource for researchers interested in the diverse pathways impacted by PPK.


Asunto(s)
Escherichia coli , Lipopolisacáridos , Fosfotransferasas (Aceptor del Grupo Fosfato) , Escherichia coli/metabolismo , Lipopolisacáridos/metabolismo , Lípido A/metabolismo , Polifosfatos/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(37): 22984-22991, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32868431

RESUMEN

Immune evasion through membrane remodeling is a hallmark of Yersinia pestis pathogenesis. Yersinia remodels its membrane during its life cycle as it alternates between mammalian hosts (37 °C) and ambient (21 °C to 26 °C) temperatures of the arthropod transmission vector or external environment. This shift in growth temperature induces changes in number and length of acyl groups on the lipid A portion of lipopolysaccharide (LPS) for the enteric pathogens Yersinia pseudotuberculosis (Ypt) and Yersinia enterocolitica (Ye), as well as the causative agent of plague, Yersinia pestis (Yp). Addition of a C16 fatty acid (palmitate) to lipid A by the outer membrane acyltransferase enzyme PagP occurs in immunostimulatory Ypt and Ye strains, but not in immune-evasive Yp Analysis of Yp pagP gene sequences identified a single-nucleotide polymorphism that results in a premature stop in translation, yielding a truncated, nonfunctional enzyme. Upon repair of this polymorphism to the sequence present in Ypt and Ye, lipid A isolated from a Yp pagP+ strain synthesized two structures with the C16 fatty acids located in acyloxyacyl linkage at the 2' and 3' positions of the diglucosamine backbone. Structural modifications were confirmed by mass spectrometry and gas chromatography. With the genotypic restoration of PagP enzymatic activity in Yp, a significant increase in lipid A endotoxicity mediated through the MyD88 and TRIF/TRAM arms of the TLR4-signaling pathway was observed. Discovery and repair of an evolutionarily lost lipid A modifying enzyme provides evidence of lipid A as a crucial determinant in Yp infectivity, pathogenesis, and host innate immune evasion.


Asunto(s)
Aciltransferasas/inmunología , Evasión Inmune/inmunología , Inmunidad Innata/inmunología , Lípido A/inmunología , Yersinia pestis/inmunología , Animales , Evolución Biológica , Línea Celular , Línea Celular Tumoral , Células HEK293 , Humanos , Leucocitos Mononucleares/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple/inmunología , Células THP-1/inmunología , Células U937 , Yersinia pseudotuberculosis/inmunología
3.
PLoS Pathog ; 15(9): e1007972, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31487328

RESUMEN

The biogenesis of bacterial cell-envelope polysaccharides requires the translocation, across the plasma membrane, of sugar sub-units that are produced inside the cytoplasm. To this end, the hydrophilic sugars are anchored to a lipid phosphate carrier (undecaprenyl phosphate (C55-P)), yielding membrane intermediates which are translocated to the outer face of the membrane. Finally, the glycan moiety is transferred to a nascent acceptor polymer, releasing the carrier in the "inactive" undecaprenyl pyrophosphate (C55-PP) form. Thus, C55-P is generated through the dephosphorylation of C55-PP, itself arising from either de novo synthesis or recycling. Two types of integral membrane C55-PP phosphatases were described: BacA enzymes and a sub-group of PAP2 enzymes (type 2 phosphatidic acid phosphatases). The human pathogen Helicobacter pylori does not contain BacA homologue but has four membrane PAP2 proteins: LpxE, LpxF, HP0350 and HP0851. Here, we report the physiological role of HP0851, renamed HupA, via multiple and complementary approaches ranging from a detailed biochemical characterization to the assessment of its effect on cell envelope metabolism and microbe-host interactions. HupA displays a dual function as being the main C55-PP pyrophosphatase (UppP) and phosphatidylglycerol phosphate phosphatase (PGPase). Although not essential in vitro, HupA was essential in vivo for stomach colonization. In vitro, the remaining UppP activity was carried out by LpxE in addition to its lipid A 1-phosphate phosphatase activity. Both HupA and LpxE have crucial roles in the biosynthesis of several cell wall polysaccharides and thus constitute potential targets for new therapeutic strategies.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Helicobacter pylori/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Pared Celular/metabolismo , Proteínas de Unión al ADN , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Femenino , Helicobacter pylori/patogenicidad , Ratones , Ratones Endogámicos , Pruebas de Sensibilidad Microbiana , Fosfatidato Fosfatasa , Monoéster Fosfórico Hidrolasas/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Polimixina B/farmacología , Pirofosfatasas/metabolismo , Estómago
4.
J Bacteriol ; 201(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31427391

RESUMEN

Helicobacter pylori uses a cluster of polar, sheathed flagella for motility, which it requires for colonization of the gastric epithelium in humans. As part of a study to identify factors that contribute to localization of the flagella to the cell pole, we disrupted a gene encoding a cardiolipin synthase (clsC) in H. pylori strains G27 and B128. Flagellum biosynthesis was abolished in the H. pylori G27 clsC mutant but not in the B128 clsC mutant. Transcriptome sequencing analysis showed that flagellar genes encoding proteins needed early in flagellum assembly were expressed at wild-type levels in the G27 clsC mutant. Examination of the G27 clsC mutant by cryo-electron tomography indicated the mutant assembled nascent flagella that contained the MS ring, C ring, flagellar protein export apparatus, and proximal rod. Motile variants of the G27 clsC mutant were isolated after allelic exchange mutagenesis using genomic DNA from the B128 clsC mutant as the donor. Genome resequencing of seven motile G27 clsC recipients revealed that each isolate contained the flgI (encodes the P-ring protein) allele from B128. Replacing the flgI allele in the G27 clsC mutant with the B128 flgI allele rescued flagellum biosynthesis. We postulate that H. pylori G27 FlgI fails to form the P ring when cardiolipin levels in the cell envelope are low, which blocks flagellum assembly at this point. In contrast, H. pylori B128 FlgI can form the P ring when cardiolipin levels are low and allows for the biosynthesis of mature flagella.IMPORTANCEH. pylori colonizes the epithelial layer of the human stomach, where it can cause a variety of diseases, including chronic gastritis, peptic ulcer disease, and gastric cancer. To colonize the stomach, H. pylori must penetrate the viscous mucous layer lining the stomach, which it accomplishes using its flagella. The significance of our research is identifying factors that affect the biosynthesis and assembly of the H. pylori flagellum, which will contribute to our understanding of motility in H. pylori, as well as other bacterial pathogens that use their flagella for host colonization.


Asunto(s)
Flagelos/genética , Helicobacter pylori/genética , Proteínas de la Membrana/genética , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética , Alelos , Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica/genética , Humanos , Mutagénesis/genética , Mutación/genética , Transcriptoma/genética
5.
Proc Natl Acad Sci U S A ; 115(38): E8968-E8976, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-30126994

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) is a global diarrheal pathogen that utilizes adhesins and secreted enterotoxins to cause disease in mammalian hosts. Decades of research on virulence factor regulation in ETEC has revealed a variety of environmental factors that influence gene expression, including bile, pH, bicarbonate, osmolarity, and glucose. However, other hallmarks of the intestinal tract, such as low oxygen availability, have not been examined. Further, determining how ETEC integrates these signals in the complex host environment is challenging. To address this, we characterized ETEC's response to the human host using samples from a controlled human infection model. We found ETEC senses environmental oxygen to globally influence virulence factor expression via the oxygen-sensitive transcriptional regulator fumarate and nitrate reduction (FNR) regulator. In vitro anaerobic growth replicates the in vivo virulence factor expression profile, and deletion of fnr in ETEC strain H10407 results in a significant increase in expression of all classical virulence factors, including the colonization factor antigen I (CFA/I) adhesin operon and both heat-stable and heat-labile enterotoxins. These data depict a model of ETEC infection where FNR activity can globally influence virulence gene expression, and therefore proximity to the oxygenated zone bordering intestinal epithelial cells likely influences ETEC virulence gene expression in vivo. Outside of the host, ETEC biofilms are associated with seasonal ETEC epidemics, and we find FNR is a regulator of biofilm production. Together these data suggest FNR-dependent oxygen sensing in ETEC has implications for human infection inside and outside of the host.


Asunto(s)
Escherichia coli Enterotoxigénica/patogenicidad , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Proteínas Hierro-Azufre/genética , Adulto , Biopelículas , Diarrea/epidemiología , Diarrea/microbiología , Diarrea/prevención & control , Células Epiteliales/microbiología , Infecciones por Escherichia coli/epidemiología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Proteínas de Escherichia coli/metabolismo , Vacunas contra Escherichia coli/administración & dosificación , Femenino , Voluntarios Sanos , Humanos , Intestinos/citología , Intestinos/microbiología , Proteínas Hierro-Azufre/metabolismo , Masculino , Persona de Mediana Edad , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Adulto Joven
6.
Infect Immun ; 86(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29866904

RESUMEN

Salmonella enterica serovars are associated with diarrhea and gastroenteritis and are a helpful model for understanding host-pathogen mechanisms. Salmonella enterica serovar Typhimurium regulates the distribution of O antigen (OAg) and presents a trimodal distribution based on Wzy polymerase and the WzzST (long-chain-length OAg [L-OAg]) and WzzfepE (very-long-chain-length OAg [VL-OAg]) copolymerases; however, several mechanisms regulating this process remain unclear. Here, we report that LPS modifications modulate the infectious process and that OAg chain length determination plays an essential role during infection. An increase in VL-OAg is dependent on Wzy polymerase, which is promoted by a growth condition resembling the environment of Salmonella-containing vacuoles (SCVs). The virulence- and stress-related periplasmic protein (VisP) participates in OAg synthesis, as a ΔvisP mutant presents a semirough OAg phenotype. The ΔvisP mutant has greatly decreased motility and J774 macrophage survival in a colitis model of infection. Interestingly, the phenotype is restored after mutation of the wzzST or wzzfepE gene in a ΔvisP background. Loss of both the visP and wzzST genes promotes an imbalance in flagellin secretion. L-OAg may function as a shield against host immune systems in the beginning of an infectious process, and VL-OAg protects bacteria during SCV maturation and facilitates intramacrophage replication. Taken together, these data highlight the roles of OAg length in generating phenotypes during S Typhimurium pathogenesis and show the periplasmic protein VisP as a novel protein in the OAg biosynthesis pathway.


Asunto(s)
Proteínas Bacterianas/metabolismo , Antígenos O/metabolismo , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/patología , Salmonella typhimurium/metabolismo , Animales , Carga Bacteriana , Línea Celular , Colitis/microbiología , Colitis/patología , Modelos Animales de Enfermedad , Heces/microbiología , Femenino , Macrófagos/inmunología , Macrófagos/microbiología , Ratones Endogámicos C57BL , Viabilidad Microbiana , Fagocitosis
7.
Infect Immun ; 82(10): 4390-404, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25114109

RESUMEN

Here, we constructed a Yersinia pseudotuberculosis mutant strain with arabinose-dependent regulated and delayed shutoff of crp expression (araC P(BAD) crp) and replacement of the msbB gene with the Escherichia coli msbB gene to attenuate it. Then, we inserted the asd mutation into this construction to form χ10057 [Δasd-206 ΔmsbB868::P(msbB) msbB(EC) ΔP(crp21)::TT araC P(BAD) crp] for use with a balanced-lethal Asd-positive (Asd(+)) plasmid to facilitate antigen synthesis. A hybrid protein composed of YopE (amino acids [aa]1 to 138) fused with full-length LcrV (YopE(Nt138)-LcrV) was synthesized in χ10057 harboring an Asd(+) plasmid (pYA5199, yopE(Nt138)-lcrV) and could be secreted through a type III secretion system (T3SS) in vitro and in vivo. Animal studies indicated that mice orally immunized with χ10057(pYA5199) developed titers of IgG response to whole-cell lysates of Y. pestis (YpL) and subunit LcrV similar to those seen with χ10057(pYA3332) (χ10057 plus an empty plasmid). However, only immunization of mice with χ10057(pYA5199) resulted in a significant secretory IgA response to LcrV. χ10057(pYA5199) induced a higher level of protection (80% survival) against intranasal (i.n.) challenge with ~240 median lethal doses (LD50) (2.4 × 10(4) CFU) of Y. pestis KIM6+(pCD1Ap) than χ10057(pYA3332) (40% survival). Splenocytes from mice vaccinated with χ10057(pYA5199) produced significant levels of gamma interferon (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin-17 (IL-17) after restimulation with LcrV and YpL antigens. Our results suggest that it is possible to use an attenuated Y. pseudotuberculosis strain delivering the LcrV antigen via the T3SS as a potential vaccine candidate against pneumonic plague.


Asunto(s)
Antígenos Bacterianos/inmunología , Vacuna contra la Peste/inmunología , Peste/prevención & control , Proteínas Citotóxicas Formadoras de Poros/inmunología , Yersinia pseudotuberculosis/inmunología , Administración Oral , Animales , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina A Secretora/sangre , Inmunoglobulina G/sangre , Leucocitos Mononucleares/inmunología , Ratones , Peste/inmunología , Vacuna contra la Peste/administración & dosificación , Vacuna contra la Peste/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Bazo/inmunología , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Yersinia pseudotuberculosis/genética
8.
Annu Rev Biochem ; 83: 99-128, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24580642

RESUMEN

Lipopolysaccharide molecules represent a unique family of glycolipids based on a highly conserved lipid moiety known as lipid A. These molecules are produced by most gram-negative bacteria, in which they play important roles in the integrity of the outer-membrane permeability barrier and participate extensively in host-pathogen interplay. Few bacteria contain lipopolysaccharide molecules composed only of lipid A. In most forms, lipid A is glycosylated by addition of the core oligosaccharide that, in some bacteria, provides an attachment site for a long-chain O-antigenic polysaccharide. The complexity of lipopolysaccharide structures is reflected in the processes used for their biosynthesis and export. Rapid growth and cell division depend on the bacterial cell's capacity to synthesize and export lipopolysaccharide efficiently and in large amounts. We review recent advances in those processes, emphasizing the reactions that are essential for viability.


Asunto(s)
Lipopolisacáridos/biosíntesis , Lipopolisacáridos/metabolismo , Adenosina Trifosfato/metabolismo , Bacterias , Fenómenos Fisiológicos Bacterianos , Proteínas Bacterianas/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Glucolípidos/metabolismo , Glicosilación , Bacterias Gramnegativas/metabolismo , Antígenos O/metabolismo , Permeabilidad , Polisacáridos/metabolismo
9.
Infect Immun ; 82(2): 660-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24478081

RESUMEN

The Vps/VacJ ABC transporter system is proposed to function in maintaining the lipid asymmetry of the outer membrane. Mutations in vps or vacJ in Shigella flexneri resulted in increased sensitivity to lysis by the detergent sodium dodecyl sulfate (SDS), and the vpsC mutant showed minor differences in its phospholipid profile compared to the wild type. vpsC mutants were unable to form plaques in cultured epithelial cells, but this was not due to a failure to invade, to replicate intracellularly, or to polymerize actin via IcsA for movement within epithelial cells. The addition of the outer membrane phospholipase gene pldA on a multicopy plasmid in a vpsC or vacJ mutant restored its resistance to SDS, suggesting a restoration of lipid asymmetry to the outer membrane. However, the pldA plasmid did not restore the mutant's ability to form plaques in tissue culture cells. Increased PldA levels also failed to restore the mutant's phospholipid profile to that of the wild type. We propose a dual function of the Vps/VacJ ABC transporter system in S. flexneri in both the maintenance of lipid asymmetry in the outer membrane and the intercellular spread of the bacteria between adjacent epithelial cells.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Shigella flexneri/fisiología , Transportadoras de Casetes de Unión a ATP/genética , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Línea Celular , Células Epiteliales/microbiología , Humanos , Proteínas de la Membrana/genética , Mutación , Fosfolípidos/análisis , Shigella flexneri/química
10.
Gut Microbes ; 4(6): 439-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23859890

RESUMEN

Helicobacter pylori is an adapted gastric pathogen that colonizes the human stomach, causing severe gastritis and gastric cancer. A hallmark of infection is the ability of this organism to evade detection by the human immune system. H. pylori has evolved a number of features to achieve this, many of which involve glyco-conjugates including the lipopolysaccharide, peptidoglycan layer, glycoproteins, and glucosylated cholesterol. These major bacterial components possess unique features from those of other gram-negative organisms, including differences in structure, assembly, and modification. These defining characteristics of H. pylori glycobiology help the pathogen establish a long-lived infection by providing camouflage, modulating the host immune response, and promoting virulence mechanisms. In this way, glyco-conjugates are essential for H. pylori pathogenicity and survival, allowing it to carve out a niche in the formidable environment of the human stomach.


Asunto(s)
Colesterol/química , Glicoproteínas/química , Helicobacter pylori/patogenicidad , Lipopolisacáridos/química , Peptidoglicano/química , Mucosa Gástrica/microbiología , Gastritis/microbiología , Infecciones por Helicobacter/patología , Helicobacter pylori/citología , Humanos , Neoplasias Gástricas/microbiología , Virulencia
11.
Mol Cell Proteomics ; 12(9): 2604-14, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23695934

RESUMEN

The use of ultraviolet photodissociation (UVPD) for the activation and dissociation of peptide anions is evaluated for broader coverage of the proteome. To facilitate interpretation and assignment of the resulting UVPD mass spectra of peptide anions, the MassMatrix database search algorithm was modified to allow automated analysis of negative polarity MS/MS spectra. The new UVPD algorithms were developed based on the MassMatrix database search engine by adding specific fragmentation pathways for UVPD. The new UVPD fragmentation pathways in MassMatrix were rigorously and statistically optimized using two large data sets with high mass accuracy and high mass resolution for both MS(1) and MS(2) data acquired on an Orbitrap mass spectrometer for complex Halobacterium and HeLa proteome samples. Negative mode UVPD led to the identification of 3663 and 2350 peptides for the Halo and HeLa tryptic digests, respectively, corresponding to 655 and 645 peptides that were unique when compared with electron transfer dissociation (ETD), higher energy collision-induced dissociation, and collision-induced dissociation results for the same digests analyzed in the positive mode. In sum, 805 and 619 proteins were identified via UVPD for the Halobacterium and HeLa samples, respectively, with 49 and 50 unique proteins identified in contrast to the more conventional MS/MS methods. The algorithm also features automated charge determination for low mass accuracy data, precursor filtering (including intact charge-reduced peaks), and the ability to combine both positive and negative MS/MS spectra into a single search, and it is freely open to the public. The accuracy and specificity of the MassMatrix UVPD search algorithm was also assessed for low resolution, low mass accuracy data on a linear ion trap. Analysis of a known mixture of three mitogen-activated kinases yielded similar sequence coverage percentages for UVPD of peptide anions versus conventional collision-induced dissociation of peptide cations, and when these methods were combined into a single search, an increase of up to 13% sequence coverage was observed for the kinases. The ability to sequence peptide anions and cations in alternating scans in the same chromatographic run was also demonstrated. Because ETD has a significant bias toward identifying highly basic peptides, negative UVPD was used to improve the identification of the more acidic peptides in conjunction with positive ETD for the more basic species. In this case, tryptic peptides from the cytosolic section of HeLa cells were analyzed by polarity switching nanoLC-MS/MS utilizing ETD for cation sequencing and UVPD for anion sequencing. Relative to searching using ETD alone, positive/negative polarity switching significantly improved sequence coverages across identified proteins, resulting in a 33% increase in unique peptide identifications and more than twice the number of peptide spectral matches.


Asunto(s)
Cromatografía Liquida/métodos , Bases de Datos de Proteínas , Ensayos Analíticos de Alto Rendimiento , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Rayos Ultravioleta , Algoritmos , Aniones , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Halobacterium/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Peso Molecular , Péptidos/metabolismo , Proteoma/química , Curva ROC , Reproducibilidad de los Resultados , Análisis de Secuencia de Proteína
12.
Proc Natl Acad Sci U S A ; 110(4): 1470-5, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23302685

RESUMEN

Gram-negative bacteria have an outer membrane containing LPS. LPS is constituted of an oligosaccharide portion and a lipid-A moiety that embeds this molecule within the outer membrane. LPS is a pathogen-associated molecular pattern, and several pathogens modify their lipid-A as a stealth strategy to avoid recognition by the innate immune system and gain resistance to host factors that disrupt the bacterial cell envelope. An essential feature of Salmonella enterica Typhimurium pathogenesis is its ability to replicate within vacuoles in professional macrophages. S. Typhimurium modifies its lipid-A by hydroxylation by the Fe2+/α-ketoglutarate-dependent dioxygenase enzyme (LpxO). Here, we show that a periplasmic protein of the bacterial oligonucleotide/oligosaccharide-binding fold family, herein named virulence and stress-related periplasmic protein (VisP), on binding to the sugar moiety of peptidoglycan interacts with LpxO. This interaction inhibits LpxO function, leading to decreased LpxO-dependent lipid-A modifications and increasing resistance to stressors within the vacuole environment during intramacrophage replication promoting systemic disease. Consequently, ΔvisP is avirulent in systemic murine infections, where VisP acts through LpxO. Several Gram-negative pathogens harbor both VisP and LpxO, suggesting that this VisP-LpxO mechanism of lipid-A modifications has broader implications in bacterial pathogenesis. Bacterial species devoid of LpxO (e.g., Escherichia coli) have no lipid-A phenotypes associated with the lack of VisP; however, VisP also controls LpxO-independent phenotypes. VisP and LpxO act independently in the S. Typhimurium murine colitis model, with both mutants being attenuated for diverging reasons; ΔvisP is less resistant to cationic antimicrobial peptides, whereas ΔlpxO is deficient for epithelial cell invasion. VisP converges bacterial cell wall homeostasis, stress responses, and pathogenicity.


Asunto(s)
Proteínas Bacterianas/fisiología , Interacciones Huésped-Patógeno/fisiología , Proteínas Periplasmáticas/fisiología , Salmonella typhimurium/patogenicidad , Factores de Virulencia/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Línea Celular , Femenino , Genes Bacterianos , Células HeLa , Interacciones Huésped-Patógeno/genética , Humanos , Lípido A/química , Lípido A/metabolismo , Macrófagos/microbiología , Macrófagos/fisiología , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas Periplasmáticas/química , Proteínas Periplasmáticas/genética , Regulón , Salmonelosis Animal/microbiología , Salmonelosis Animal/fisiopatología , Salmonella typhimurium/genética , Salmonella typhimurium/fisiología , Homología de Secuencia de Aminoácido , Virulencia/genética , Virulencia/fisiología , Factores de Virulencia/química , Factores de Virulencia/genética
13.
Infect Immun ; 80(9): 3122-31, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22710876

RESUMEN

The adaptation of Pseudomonas aeruginosa to its environment, including the host, is tightly controlled by its network of regulatory systems. The two-component regulatory system PhoPQ has been shown to play a role in the virulence and polymyxin resistance of P. aeruginosa as well as several other Gram-negative species. Dysregulation of this system has been demonstrated in clinical isolates, yet how it affects virulence of P. aeruginosa is unknown. To investigate this, an assay was used whereby bacteria were cocultured with human bronchial epithelial cells. The interaction of wild-type (WT) bacteria that had adhered to epithelial cells led to a large upregulation of the expression of the oprH-phoP-phoQ operon and its target, the arn lipopolysaccharide (LPS) modification operon, in a PhoQ-dependent manner, compared to cells in the supernatant that had failed to adhere. Relative to the wild type, a phoQ mutant cocultured on epithelial cells produced less secreted protease and lipase and, like the phoQ mutant, piv, lipH, and lasB mutants demonstrated reduced cytotoxicity toward epithelial cells. Mutation in phoQ also resulted in alterations to lipid A and to increased inflammatory LPS. These data indicate that mutation of phoQ results in a phenotype that is similar to the less virulent but more inflammatory phenotype of clinical strains isolated from chronic-stage cystic fibrosis lung infections.


Asunto(s)
Proteínas Bacterianas/metabolismo , Células Epiteliales/microbiología , Regulación Bacteriana de la Expresión Génica , Interacciones Huésped-Patógeno , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Adhesión Bacteriana , Proteínas Bacterianas/genética , Línea Celular , Humanos , Inflamación , Mutación
14.
Mol Microbiol ; 78(4): 837-52, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20659292

RESUMEN

Helicobacter pylori produces a unique surface lipopolysaccharide (LPS) characterized by strikingly low endotoxicity that is thought to aid the organism in evading the host immune response. This reduction in endotoxicity is predicted to arise from the modification of the Kdo-lipid A domain of Helicobacter LPS by a series of membrane bound enzymes including a Kdo (3-deoxy-d-manno-octulosonic acid) hydrolase responsible for the modification of the core oligosaccharide. Here, we report that Kdo hydrolase activity is dependent upon a putative two-protein complex composed of proteins Hp0579 and Hp0580. Inactivation of Kdo hydrolase activity produced two phenotypes associated with cationic antimicrobial peptide resistance and O-antigen expression. Kdo hydrolase mutants were highly sensitive to polymyxin B, which could be attributed to a defect in downstream modifications to the lipid A 4'-phosphate group. Production of a fully extended O-antigen was also diminished in a Kdo hydrolase mutant, with a consequent increase in core-lipid A. Finally, expression of O-antigen Lewis X and Y epitopes, known to mimic glycoconjugates found on human tissues, was also affected. Taken together, we have demonstrated that loss of Kdo hydrolase activity affects all three domains of H. pylori LPS, thus highlighting its role in the maintenance of the bacterial surface.


Asunto(s)
Helicobacter pylori/química , Helicobacter pylori/metabolismo , Hidrolasas/metabolismo , Lipopolisacáridos/metabolismo , Azúcares Ácidos/metabolismo , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Hidrolasas/genética , Antígenos del Grupo Sanguíneo de Lewis/metabolismo , Antígeno Lewis X/metabolismo , Antígenos O/biosíntesis , Polimixina B/farmacología
15.
Mol Microbiol ; 67(2): 264-77, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18047581

RESUMEN

One-third of the lipid A found in the Escherichia coli outer membrane contains an unsubstituted diphosphate unit at position 1 (lipid A 1-diphosphate). We now report an inner membrane enzyme, LpxT (YeiU), which specifically transfers a phosphate group to lipid A, forming the 1-diphosphate species. (32)P-labelled lipid A obtained from lpxT mutants do not produce lipid A 1-diphosphate. In vitro assays with Kdo(2)-[4'-(32)P]lipid A as the acceptor shows that LpxT uses undecaprenyl pyrophosphate as the substrate donor. Inhibition of lipid A 1-diphosphate formation in wild-type bacteria was demonstrated by sequestering undecaprenyl pyrophosphate with the cyclic polypeptide antibiotic bacitracin, providing evidence that undecaprenyl pyrophosphate serves as the donor substrate within whole bacteria. LpxT-catalysed phosphorylation is dependent upon transport of lipid A across the inner membrane by MsbA, a lipid A flippase, indicating a periplasmic active site. In conclusion, we demonstrate a novel pathway in the periplasmic modification of lipid A that is directly linked to the synthesis of undecaprenyl phosphate, an essential carrier lipid required for the synthesis of various bacterial polymers, such as peptidoglycan.


Asunto(s)
Lípido A/metabolismo , Periplasma/enzimología , Fosfatos de Poliisoprenilo/biosíntesis , Transportadoras de Casetes de Unión a ATP/metabolismo , Antibacterianos/farmacología , Bacitracina/farmacología , Proteínas Bacterianas/metabolismo , Escherichia coli K12/enzimología , Escherichia coli K12/genética , Lípido A/antagonistas & inhibidores , Lípidos de la Membrana/metabolismo , Mutación , Peptidil Transferasas/metabolismo , Fosfatos/metabolismo , Fosforilación/efectos de los fármacos , Fosfatos de Poliisoprenilo/antagonistas & inhibidores , Fosfatos de Poliisoprenilo/metabolismo , Pirofosfatasas/genética , Pirofosfatasas/metabolismo
16.
J Bacteriol ; 188(1): 191-201, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16352835

RESUMEN

Lipopolysaccharide (LPS) is the major surface component of gram-negative bacteria, and a component of LPS, lipid A, is recognized by the innate immune system through the Toll-like receptor 4/MD-2 complex. Pseudomonas aeruginosa, an environmental gram-negative bacterium that opportunistically infects the respiratory tracts of patients with cystic fibrosis (CF), can synthesize various structures of lipid A. Lipid A from P. aeruginosa strains isolated from infants with CF has a specific structure that includes the removal of the 3 position 3-OH C10 fatty acid. Here we demonstrate increased expression of the P. aeruginosa lipid A 3-O-deacylase (PagL) in isolates from CF infants compared to that in environmental isolates. PagL activity was increased in environmental isolates by growth in medium limited for magnesium and decreased by growth at low temperature in laboratory-adapted strains of P. aeruginosa. P. aeruginosa PagL was shown to be an outer membrane protein by isopycnic density gradient centrifugation. Heterologous expression of P. aeruginosa pagL in Salmonella enterica serovar Typhimurium and Escherichia coli resulted in removal of the 3-OH C14 fatty acid from lipid A, indicating that P. aeruginosa PagL recognizes either 3-OH C10 or 3-OH C14. Finally, deacylated lipid A species were not observed in some clinical P. aeruginosa isolates from patients with severe pulmonary disease, suggesting that loss of PagL function can occur during long-term adaptation to the CF airway.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Fibrosis Quística/microbiología , Regulación Bacteriana de la Expresión Génica , Lípido A/metabolismo , Pseudomonas aeruginosa/enzimología , Sistema Respiratorio/microbiología , Acilación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Hidrolasas de Éster Carboxílico/genética , Niño , Fibrosis Quística/fisiopatología , Histidina , Humanos , Magnesio , Pseudomonas aeruginosa/genética , Sistema Respiratorio/fisiopatología , Temperatura
17.
J Biol Chem ; 280(31): 28186-94, 2005 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15951433

RESUMEN

Attachment of positively charged, amine-containing residues such as 4-amino-4-deoxy-l-arabinose (l-Ara4N) and phosphoethanolamine (pEtN) to Escherichia coli and Salmonella typhimurium lipid A is required for resistance to the cationic antimicrobial peptide, polymyxin. In an attempt to discover additional lipid A modifications important for polymyxin resistance, we generated polymyxin-sensitive mutants of an E. coli pmrA(C) strain, WD101. A subset of polymyxin-sensitive mutants produced a lipid A that lacked both the 3'-acyloxyacyl-linked myristate (C(14)) and l-Ara4N, even though the necessary enzymatic machinery required to synthesize l-Ara4N-modified lipid A was present. Inactivation of lpxM in both E. coli and S. typhimurium resulted in the loss of l-Ara4N addition, as well as, increased sensitivity to polymyxin. However, decoration of the lipid A phosphate groups with pEtN residues was not effected in lpxM mutants. In summary, we demonstrate that attachment of l-Ara4N to the phosphate groups of lipid A and the subsequent resistance to polymyxin is dependent upon the presence of the secondary linked myristoyl group.


Asunto(s)
Farmacorresistencia Bacteriana , Escherichia coli/efectos de los fármacos , Lípido A/metabolismo , Ácido Mirístico/metabolismo , Polimixinas/farmacología , Salmonella typhimurium/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Conformación de Carbohidratos , Secuencia de Carbohidratos , Hexosiltransferasas/metabolismo , Lípido A/química , Datos de Secuencia Molecular , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA