Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 578(7796): 582-587, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051588

RESUMEN

Addressing the ongoing antibiotic crisis requires the discovery of compounds with novel mechanisms of action that are capable of treating drug-resistant infections1. Many antibiotics are sourced from specialized metabolites produced by bacteria, particularly those of the Actinomycetes family2. Although actinomycete extracts have traditionally been screened using activity-based platforms, this approach has become unfavourable owing to the frequent rediscovery of known compounds. Genome sequencing of actinomycetes reveals an untapped reservoir of biosynthetic gene clusters, but prioritization is required to predict which gene clusters may yield promising new chemical matter2. Here we make use of the phylogeny of biosynthetic genes along with the lack of known resistance determinants to predict divergent members of the glycopeptide family of antibiotics that are likely to possess new biological activities. Using these predictions, we uncovered two members of a new functional class of glycopeptide antibiotics-the known glycopeptide antibiotic complestatin and a newly discovered compound we call corbomycin-that have a novel mode of action. We show that by binding to peptidoglycan, complestatin and corbomycin block the action of autolysins-essential peptidoglycan hydrolases that are required for remodelling of the cell wall during growth. Corbomycin and complestatin have low levels of resistance development and are effective in reducing bacterial burden in a mouse model of skin MRSA infection.


Asunto(s)
Antibacterianos , Descubrimiento de Drogas , Péptidos Cíclicos , Peptidoglicano/efectos de los fármacos , Peptidoglicano/metabolismo , Actinobacteria/química , Actinobacteria/genética , Actinobacteria/metabolismo , Animales , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Vías Biosintéticas/genética , Pared Celular/metabolismo , Clorofenoles/química , Clorofenoles/metabolismo , Clorofenoles/farmacología , Modelos Animales de Enfermedad , Farmacorresistencia Microbiana/efectos de los fármacos , Farmacorresistencia Microbiana/genética , Femenino , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Pruebas de Sensibilidad Microbiana , Familia de Multigenes , N-Acetil Muramoil-L-Alanina Amidasa/antagonistas & inhibidores , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Péptidos Cíclicos/farmacología , Filogenia , Piel/microbiología , Infecciones Estafilocócicas/microbiología
2.
Artículo en Inglés | MEDLINE | ID: mdl-30509943

RESUMEN

Bacterial membrane vesicle research has so far focused mainly on Gram-negative bacteria. Only recently have Gram-positive bacteria been demonstrated to produce and release extracellular membrane vesicles (MVs) that contribute to bacterial virulence. Although treatment of bacteria with antibiotics is a well-established trigger of bacterial MV formation, the underlying mechanisms are poorly understood. In this study, we show that antibiotics can induce MVs through different routes in the important human pathogen Staphylococcus aureus DNA-damaging agents and antibiotics inducing the SOS response triggered vesicle formation in lysogenic strains of S. aureus but not in their phage-devoid counterparts. The ß-lactam antibiotics flucloxacillin and ceftaroline increased vesicle formation in a prophage-independent manner by weakening the peptidoglycan layer. We present evidence that the amount of DNA associated with MVs formed by phage lysis is greater than that for MVs formed by ß-lactam antibiotic-induced blebbing. The purified MVs derived from S. aureus protected the bacteria from challenge with daptomycin, a membrane-targeting antibiotic, both in vitro and ex vivo in whole blood. In addition, the MVs protected S. aureus from killing in whole blood, indicating that antibiotic-induced MVs function as a decoy and thereby contribute to the survival of the bacterium.


Asunto(s)
Antibacterianos/farmacología , Vesículas Citoplasmáticas/efectos de los fármacos , Vesículas Citoplasmáticas/virología , Lisogenia/fisiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/virología , Bacteriófagos/fisiología , Cefalosporinas/farmacología , ADN Bacteriano/efectos de los fármacos , ADN Bacteriano/genética , Daptomicina/farmacología , Floxacilina/farmacología , Humanos , Lisogenia/genética , Peptidoglicano/efectos de los fármacos , Ceftarolina
3.
Int J Med Microbiol ; 309(6): 151335, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31378704

RESUMEN

The type VI secretion system (T6SS) injects effector proteins into neighboring bacteria and host cells. Effector translocation is driven by contraction of a tubular sheath in the cytoplasm that expels an inner needle across the cell envelope. The AAA + ATPase ClpV disassembles and recycles the contracted sheath. While ClpV-1-GFP of the Burkholderia T6SS-1, which targets prokaryotic cells, assembles into randomly localized foci, ClpV-5-GFP of the virulence-associated T6SS-5 displays a polar distribution. The mechanisms underlying the localization of T6SSs to a particular site in the bacterial cell are currently unknown. We recently showed that ClpV-5-GFP retains its polar localization in the absence of all T6SS-5 components during infection of host cells. Herein, we set out to identify factors involved in the distribution of ClpV-5 and ClpV-1 in Burkholderia thailandensis. We show that focal assembly and polar localization of ClpV-5-GFP is not dependent on the intracellular host cell environment, known to contain the signal to induce T6SS-5 gene expression. In contrast to ClpV-5-GFP, localization of ClpV-1-GFP was dependent on the cognate T6SS. Foci formation of both ClpV5-GFP and ClpV-1-GFP was decreased by D cycloserine-mediated inhibition of peptidoglycan synthesis while treatment of B. thailandensis with A22 blocking the cytoskeletal protein MreB did not affect assembly of ClpV-5 and ClpV-1 into single discrete foci. Furthermore, we found that surface contact promotes but is not essential for localization of ClpV-5-GFP to the pole whereas expression of clpV-1-gfp appears to be induced by surface contact. In summary, the study provides novel insights into the localization of ClpV ATPases of T6SSs targeting prokaryotic and eukaryotic cells.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Burkholderia/fisiología , Sistemas de Secreción Tipo VI/metabolismo , Factores de Virulencia/metabolismo , Adhesión Bacteriana , Burkholderia/efectos de los fármacos , Burkholderia/genética , Cicloserina/farmacología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células HeLa , Humanos , Peptidoglicano/biosíntesis , Peptidoglicano/efectos de los fármacos , Transporte de Proteínas/fisiología , Eliminación de Secuencia , Sistemas de Secreción Tipo VI/genética
4.
Artículo en Inglés | MEDLINE | ID: mdl-30275084

RESUMEN

The Gram-negative human pathogen Neisseria gonorrhoeae has progressively developed resistance to antibiotic monotherapies, and recent failures of dual-drug therapy have heightened concerns that strains resistant to all available antibiotics will begin circulating globally. Targeting bacterial cell wall assembly has historically been effective at treating infections with N. gonorrhoeae, but as the effectiveness of ß-lactams (including cephalosporins) is challenged by increasing resistance, research has expanded into compounds that target the numerous other enzymes with roles in peptidoglycan metabolism. One example is the dithiazoline compound JNJ-853346 (DTZ), which inhibits the activity of an Escherichia coli serine protease l,d-carboxypeptidase (LdcA). Recently, the characterization of an LdcA homolog in N. gonorrhoeae revealed localization and activity differences from the characterized E. coli LdcA, prompting us to explore the effectiveness of DTZ against N. gonorrhoeae We found that DTZ is effective at inhibiting N. gonorrhoeae in all growth phases, unlike the specific stationary-phase inhibition seen in E. coli Surprisingly, DTZ does not inhibit gonococcal LdcA enzyme activity, and DTZ sensitivity is not significantly decreased in ldcA mutants. While effective against numerous N. gonorrhoeae strains, including recent multidrug-resistant isolates, DTZ is much less effective at inhibiting growth of the commensal species Lactobacillus gasseri DTZ treatment during coinfections of epithelial cells resulted in significant lowering of gonococcal burden and interleukin-8 secretion without significantly impacting recovery of viable L. gasseri This selective toxicity presents a possible pathway for the use of DTZ as an effective antigonococcal agent at concentrations that do not impact vaginal commensals.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/efectos de los fármacos , Lactobacillus gasseri/efectos de los fármacos , Neisseria gonorrhoeae/efectos de los fármacos , Tiazoles/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Carboxipeptidasas A/genética , Carboxipeptidasas A/metabolismo , Supervivencia Celular/efectos de los fármacos , Pared Celular/metabolismo , Expresión Génica , Células HCT116 , Humanos , Interleucina-8/genética , Interleucina-8/inmunología , Lactobacillus gasseri/crecimiento & desarrollo , Lactobacillus gasseri/metabolismo , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Mutación , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/crecimiento & desarrollo , Neisseria gonorrhoeae/metabolismo , Peptidoglicano/biosíntesis , Peptidoglicano/efectos de los fármacos , Probióticos/química , Especificidad de la Especie
5.
Parasitology ; 145(2): 116-133, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-27976597

RESUMEN

Mycobacterium tuberculosis (Mtb), the etiological agent of tuberculosis (TB), is recognized as a global health emergency as promoted by the World Health Organization. Over 1 million deaths per year, along with the emergence of multi- and extensively-drug resistant strains of Mtb, have triggered intensive research into the pathogenicity and biochemistry of this microorganism, guiding the development of anti-TB chemotherapeutic agents. The essential mycobacterial cell wall, sharing some common features with all bacteria, represents an apparent 'Achilles heel' that has been targeted by TB chemotherapy since the advent of TB treatment. This complex structure composed of three distinct layers, peptidoglycan, arabinogalactan and mycolic acids, is vital in supporting cell growth, virulence and providing a barrier to antibiotics. The fundamental nature of cell wall synthesis and assembly has rendered the mycobacterial cell wall as the most widely exploited target of anti-TB drugs. This review provides an overview of the biosynthesis of the prominent cell wall components, highlighting the inhibitory mechanisms of existing clinical drugs and illustrating the potential of other unexploited enzymes as future drug targets.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Sistemas de Liberación de Medicamentos , Mycobacterium tuberculosis/metabolismo , Pared Celular/química , Diseño de Fármacos , Galactanos/biosíntesis , Galactanos/metabolismo , Humanos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Ácidos Micólicos/metabolismo , Peptidoglicano/biosíntesis , Peptidoglicano/efectos de los fármacos , Peptidoglicano/metabolismo , Tuberculosis/tratamiento farmacológico , Tuberculosis/microbiología , Virulencia
6.
Nat Prod Rep ; 34(7): 909-932, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28675405

RESUMEN

Covering: up to 2017History points to the bacterial cell wall biosynthetic network as a very effective target for antibiotic intervention, and numerous natural product inhibitors have been discovered. In addition to the inhibition of enzymes involved in the multistep synthesis of the macromolecular layer, in particular, interference with membrane-bound substrates and intermediates essential for the biosynthetic reactions has proven a valuable antibacterial strategy. A prominent target within the peptidoglycan biosynthetic pathway is lipid II, which represents a particular "Achilles' heel" for antibiotic attack, as it is readily accessible on the outside of the cytoplasmic membrane. Lipid II is a unique non-protein target that is one of the structurally most conserved molecules in bacterial cells. Notably, lipid II is more than just a target molecule, since sequestration of the cell wall precursor may be combined with additional antibiotic activities, such as the disruption of membrane integrity or disintegration of membrane-bound multi-enzyme machineries. Within the membrane bilayer lipid II is likely organized in specific anionic phospholipid patches that form a particular "landing platform" for antibiotics. Nature has invented a variety of different "lipid II binders" of at least 5 chemical classes, and their antibiotic activities can vary substantially depending on the compounds' physicochemical properties, such as amphiphilicity and charge, and thus trigger diverse cellular effects that are decisive for antibiotic activity.


Asunto(s)
Bacterias/citología , Pared Celular/metabolismo , Uridina Difosfato Ácido N-Acetilmurámico/análogos & derivados , Antibacterianos/química , Antibacterianos/farmacología , Productos Biológicos/antagonistas & inhibidores , Vías Biosintéticas , Estructura Molecular , Peptidoglicano/efectos de los fármacos , Unión Proteica , Uridina Difosfato Ácido N-Acetilmurámico/metabolismo
7.
EMBO J ; 30(16): 3442-53, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21792174

RESUMEN

Production of non-canonical D-amino acids (NCDAAs) in stationary phase promotes remodelling of peptidoglycan (PG), the polymer that comprises the bacterial cell wall. Impairment of NCDAAs production leads to excessive accumulation of PG and hypersensitivity to osmotic shock; however, the mechanistic bases for these phenotypes were not previously determined. Here, we show that incorporation of NCDAAs into PG is a critical means by which NCDAAs control PG abundance and strength. We identified and reconstituted in vitro two (of at least three) distinct processes that mediate NCDAA incorporation. Diverse bacterial phyla incorporate NCDAAs into their cell walls, either through periplasmic editing of the mature PG or via incorporation into PG precursor subunits in the cytosol. Production of NCDAAs in Vibrio cholerae requires the stress response sigma factor RpoS, suggesting that NCDAAs may aid bacteria in responding to varied environmental challenges. The widespread capacity of diverse bacteria, including non-producers, to incorporate NCDAAs suggests that these amino acids may serve as both autocrine- and paracrine-like regulators of chemical and physical properties of the cell wall in microbial communities.


Asunto(s)
Aminoácidos/metabolismo , Aminoácidos/farmacología , Pared Celular/metabolismo , Proteínas de la Membrana/metabolismo , Peptidoglicano/efectos de los fármacos , Secuencia de Aminoácidos , Aminoácidos/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Proteínas de la Membrana/genética , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Peptidoglicano/biosíntesis , Peptidil Transferasas/genética , Peptidil Transferasas/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Estereoisomerismo , Relación Estructura-Actividad , Especificidad por Sustrato , Vibrio cholerae/genética , Vibrio cholerae/metabolismo , Vibrio cholerae/ultraestructura
8.
J Antimicrob Chemother ; 70(5): 1453-65, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25630640

RESUMEN

OBJECTIVES: In the light of increasing drug resistance in Staphylococcus aureus, bacteriophage endolysins [peptidoglycan hydrolases (PGHs)] have been suggested as promising antimicrobial agents. The aim of this study was to determine the antimicrobial activity of nine enzymes representing unique homology groups within a diverse class of staphylococcal PGHs. METHODS: PGHs were recombinantly expressed, purified and tested for staphylolytic activity in multiple in vitro assays (zymogram, turbidity reduction assay and plate lysis) and against a comprehensive set of strains (S. aureus and CoNS). PGH cut sites in the staphylococcal peptidoglycan were determined by biochemical assays (Park-Johnson and Ghuysen procedures) and MS analysis. The enzymes were tested for their ability to eradicate static S. aureus biofilms and compared for their efficacy against systemic MRSA infection in a mouse model. RESULTS: Despite similar modular architectures and unexpectedly conserved cleavage sites in the peptidoglycan (conferred by evolutionarily divergent catalytic domains), the enzymes displayed varying degrees of in vitro lytic activity against numerous staphylococcal strains, including cell surface mutants and drug-resistant strains, and proved effective against static biofilms. In a mouse model of systemic MRSA infection, six PGHs provided 100% protection from death, with animals being free of clinical signs at the end of the experiment. CONCLUSIONS: Our results corroborate the high potential of PGHs for treatment of S. aureus infections and reveal unique antimicrobial and biochemical properties of the different enzymes, suggesting a high diversity of potential applications despite highly conserved peptidoglycan target sites.


Asunto(s)
Antibacterianos/uso terapéutico , Bacteriófagos/enzimología , Terapia Biológica/métodos , Endopeptidasas/uso terapéutico , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Animales , Bacteriemia/tratamiento farmacológico , Bacteriemia/microbiología , Pared Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Endopeptidasas/genética , Endopeptidasas/metabolismo , Femenino , Hidrólisis , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Peptidoglicano/efectos de los fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapéutico , Infecciones Estafilocócicas/microbiología , Análisis de Supervivencia , Resultado del Tratamiento
9.
Clin Infect Dis ; 54 Suppl 3: S214-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22431851

RESUMEN

Oritavancin is a semisynthetic lipoglycopeptide analogue of vancomycin that contains the heptapeptide core common to all glycopeptides. It differs from vancomycin by the presence of a hydrophobic N-4-(4-chlorophenyl)benzyl (also referred to as 4'-chlorobiphenylmethyl) substituent on the disaccharide sugar, the addition of a 4-epi-vancosamine monosaccharide to the amino acid residue in ring 6, and the replacement of the vancosamine moiety by 4-epi-vancosamine. One mechanism of action of oritavancin is inhibition of transglycosylation (important in peptidoglycan synthesis) by binding to D-alanyl-D-alanine stem termini in Gram-positive bacteria. The inhibition of peptidoglycan synthesis via inhibition of transglycosylation is common to all glycopeptides (vancomycin) and lipoglycopeptides. Secondary binding of oritavancin to the pentaglycyl (Asp/Asn) bridging segment in peptidoglycan also occurs, which distinguishes it from vancomycin and contributes to oritavancin's activity versus vancomycin-resistant organisms. The presence of the hydrophobic 4'-chlorobiphenylmethyl group allows for interaction and disruption of the cell membrane, resulting in depolarization, permeabilization, and concentration-dependent, rapid cell death. This mechanism is shared with telavancin but not vancomycin and results in activity against daptomycin-nonsusceptible organisms. In conclusion, oritavancin's mechanism of action involves at least 3 known mechanisms: inhibition of transglycosylation, inhibition of transpeptidation, and cell membrane interaction/disruption. Oritavancin's multiple mechanisms of action confer activity against vancomycin-susceptible and -resistant organisms, as well as rapid, concentration-dependent killing versus actively growing, stationary phase, and biofilm-producing Gram-positive bacteria.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Glicopéptidos/química , Glicopéptidos/farmacología , Antibacterianos/metabolismo , Membrana Celular/efectos de los fármacos , Enterococcus faecium/efectos de los fármacos , Glicopéptidos/metabolismo , Glicosilación/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Lipoglucopéptidos , Peptidoglicano/efectos de los fármacos , Peptidoglicano/metabolismo , Staphylococcus aureus/efectos de los fármacos
10.
Microbiol Spectr ; 10(1): e0201921, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35171032

RESUMEN

In the current scenario of antibiotic resistance magnification, new weapons against top nosocomial pathogens like Pseudomonas aeruginosa are urgently needed. The interplay between ß-lactam resistance and virulence is considered a promising source of targets to be attacked by antivirulence therapies, and in this regard, we previously showed that a peptidoglycan recycling blockade dramatically attenuated the pathogenic power of P. aeruginosa strains hyperproducing the chromosomal ß-lactamase AmpC. Here, we sought to ascertain whether this observation could be applicable to other ß-lactamases. To do so, P. aeruginosa wild-type or peptidoglycan recycling-defective strains (ΔampG and ΔnagZ) harboring different cloned ß-lactamases (transferable GES, VIM, and OXA types) were used to assess their virulence in Galleria mellonella larvae by determining 50% lethal doses (LD50s). A mild yet significant LD50 increase was observed after peptidoglycan recycling disruption per se, whereas the expression of class A and B enzymes did not impact virulence. While the production of the narrow-spectrum class D OXA-2 entailed a slight attenuation, its extended-spectrum derivatives OXA-226 (W159R [bearing a change of W to R at position 159]), OXA-161 (N148D), and principally, OXA-539 (D149 duplication) were associated with outstanding virulence impairments, especially in recycling-defective backgrounds (with some LD50s being >1,000-fold that of the wild type). Although their exact molecular bases remain to be deciphered, these results suggest that mutations affecting the catalytic center and, therefore, the hydrolytic spectrum of OXA-2-derived enzymes also drastically impact the pathogenic power of P. aeruginosa. This work provides new and relevant knowledge to the complex topic of the interplay between the production of ß-lactamases and virulence that could be useful to build future therapeutic strategies against P. aeruginosa. IMPORTANCE Pseudomonas aeruginosa is one of the leading nosocomial pathogens whose growing resistance makes the development of therapeutic options extremely urgent. The resistance-virulence interplay has classically aroused researchers' interest as a source of therapeutic targets. In this regard, we describe a wide array of virulence attenuations associated with different transferable ß-lactamases, among which the production of OXA-2-derived extended-spectrum ß-lactamases stood out as a dramatic handicap for pathogenesis, likely as a side effect of mutations causing the expansion of their hydrolytic spectrums. Moreover, our results confirm the validity of disturbing peptidoglycan recycling as a weapon to attenuate P. aeruginosa virulence in class C and D ß-lactamase production backgrounds. In the current scenario of dissemination of horizontally acquired ß-lactamases, this work brings out new data on the complex interplay between the production of specific enzymes and virulence attenuation that, if complemented with the characterization of the underlying mechanisms, will likely be exploitable to develop future virulence-targeting antipseudomonal strategies.


Asunto(s)
Peptidoglicano/efectos de los fármacos , Peptidoglicano/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Pared Celular/metabolismo , Cefalosporinasa , Transferencia de Gen Horizontal , Proteínas de Transporte de Membrana , Pruebas de Sensibilidad Microbiana , Mariposas Nocturnas , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Virulencia/efectos de los fármacos , Resistencia betalactámica/efectos de los fármacos , Resistencia betalactámica/genética
11.
J Antibiot (Tokyo) ; 73(2): 125-127, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31654037

RESUMEN

Peptidoglycan is an indispensable component of bacterial cell walls. We recently discovered an alternative peptidoglycan biosynthetic pathway, which involves two enzymes, MurD2 and MurL, catalyzing the ligation of L-Glu to UDP-MurNAc-L-Ala and epimerization of the terminal L-Glu of the MurD2 product, respectively. Because the pathway operates in Xanthomonas oryze, a pathogen causing bacterial blight of rice, we searched for specific inhibitors from metabolites produced by actinomycetes to obtain a lead compound to function as an agrochemical. Actinomycin D was isolated from Streptomyces parvulus NBRC 13193 as a specific inhibitor of the pathway. In vitro analysis indicated that actinomycin D inhibited the MurD2 reaction.


Asunto(s)
Antibacterianos/farmacología , Dactinomicina/farmacología , Peptidoglicano/efectos de los fármacos , Streptomyces/metabolismo , Xanthomonas/efectos de los fármacos , Antibacterianos/aislamiento & purificación , Dactinomicina/aislamiento & purificación , Peptidoglicano/biosíntesis , Xanthomonas/enzimología
12.
Biophys J ; 97(8): 2250-7, 2009 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-19843457

RESUMEN

The oligo-acyl-lysyl, C(12(omega 7))K-beta(12), is comprised of only three Lys residues. Despite its small size, it exhibits potent bacteriostatic activity against Gram-positive bacteria, but it is approximately 10-fold less potent against Gram-negative bacteria. We followed the interactions of C(12(omega 7))K-beta(12) from its initial contact with the bacterial surface across the cell wall down to the cytoplasmic membrane. Binding to anionic lipids, as well as to negatively charged LPS and LTA, occurs with very high affinity. The C(12(omega 7))K-beta(12) does not cross the outer membrane of Gram-negative bacteria; rather, it achieves its action by depositing on the LPS layer, promoting surface adhesion and blocking passage of solutes. In Gram-positive bacteria, the thick peptidoglycan layer containing LTA allows passage of C(12(omega 7))K-beta(12) and promotes its accumulation in the small periplasm. From that location it is then driven to the membrane by strong electrostatic interactions. Despite its high potency against Gram-positive bacteria, this agent is not capable of efficiently breaking down the permeability barrier of the cytoplasmic membrane or of reaching an intracellular target, as suggested by the fact that it does not interact with DNA.


Asunto(s)
Antibacterianos/farmacología , Pared Celular/efectos de los fármacos , Dipéptidos/farmacología , Escherichia coli/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/química , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Pared Celular/química , Quelantes/química , Quelantes/farmacología , ADN Bacteriano/química , Dipéptidos/química , Relación Dosis-Respuesta a Droga , Ácido Edético/química , Ácido Edético/farmacología , Escherichia coli/química , Lipopolisacáridos/química , Membranas Artificiales , Modelos Biológicos , Peptidoglicano/química , Peptidoglicano/efectos de los fármacos , Periplasma/química , Periplasma/efectos de los fármacos , Permeabilidad , Staphylococcus aureus/química , Electricidad Estática , Ácidos Teicoicos/química
13.
Nat Microbiol ; 4(10): 1716-1726, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31285586

RESUMEN

The peptidoglycan cell wall is an essential structure for the growth of most bacteria. However, many are capable of switching into a wall-deficient L-form state in which they are resistant to antibiotics that target cell wall synthesis under osmoprotective conditions, including host environments. L-form cells may have an important role in chronic or recurrent infections. The cellular pathways involved in switching to and from the L-form state remain poorly understood. This work shows that the lack of a cell wall, or blocking its synthesis with ß-lactam antibiotics, results in an increased flux through glycolysis. This leads to the production of reactive oxygen species from the respiratory chain, which prevents L-form growth. Compensating for the metabolic imbalance by slowing down glycolysis, activating gluconeogenesis or depleting oxygen enables L-form growth in Bacillus subtilis, Listeria monocytogenes and Staphylococcus aureus. These effects do not occur in Enterococcus faecium, which lacks the respiratory chain pathway. Our results collectively show that when cell wall synthesis is blocked under aerobic and glycolytic conditions, perturbation of cellular metabolism causes cell death. We provide a mechanistic framework for many anecdotal descriptions of the optimal conditions for L-form growth and non-lytic killing by ß-lactam antibiotics.


Asunto(s)
Bacillus subtilis/efectos de los fármacos , Bacillus subtilis/metabolismo , Carbono/metabolismo , Formas L/efectos de los fármacos , Formas L/metabolismo , beta-Lactamas/farmacología , Bacillus subtilis/genética , Bacillus subtilis/crecimiento & desarrollo , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Transporte de Electrón/genética , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/metabolismo , Gluconeogénesis , Formas L/genética , Formas L/crecimiento & desarrollo , Muramidasa/farmacología , Mutación , Penicilina G/farmacología , Peptidoglicano/efectos de los fármacos , Peptidoglicano/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/toxicidad
14.
J Bacteriol ; 190(24): 7904-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18835985

RESUMEN

The advent of Staphylococcus aureus strains that are resistant to virtually all antibiotics has increased the need for new antistaphylococcal agents. An example of such a potential therapeutic is lysostaphin, an enzyme that specifically cleaves the S. aureus peptidoglycan, thereby lysing the bacteria. Here we tracked over time the structural and physical dynamics of single S. aureus cells exposed to lysostaphin, using atomic force microscopy. Topographic images of native cells revealed a smooth surface morphology decorated with concentric rings attributed to newly formed peptidoglycan. Time-lapse images collected following addition of lysostaphin revealed major structural changes in the form of cell swelling, splitting of the septum, and creation of nanoscale perforations. Notably, treatment of the cells with lysostaphin was also found to decrease the bacterial spring constant and the cell wall stiffness, demonstrating that structural changes were correlated with major differences in cell wall nanomechanical properties. We interpret these modifications as resulting from the digestion of peptidoglycan by lysostaphin, eventually leading to the formation of osmotically fragile cells. This study provides new insight into the lytic activity of lysostaphin and offers promising prospects for the study of new antistaphylococcal agents.


Asunto(s)
Pared Celular/efectos de los fármacos , Lisostafina/farmacología , Peptidoglicano/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Antibacterianos/farmacología , Módulo de Elasticidad , Microscopía de Fuerza Atómica , Staphylococcus aureus/citología
15.
J Proteomics ; 156: 20-28, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28043878

RESUMEN

Tetracyclines are among the most commonly used antibiotics administrated to farm animals for disease treatment and prevention, contributing to the worldwide increase in antibiotic resistance in animal and human pathogens. Although tetracycline mechanisms of resistance are well known, the role of metabolism in bacterial reaction to antibiotic stress is still an important assignment and could contribute to the understanding of tetracycline related stress response. In this study, spectral counts-based label free quantitative proteomics has been applied to study the response to tetracycline of the environmental-borne Escherichia coli EcAmb278 isolate soluble proteome. A total of 1484 proteins were identified by high resolution mass spectrometry at a false discovery rate threshold of 1%, of which 108 were uniquely identified under absence of tetracycline whereas 126 were uniquely identified in presence of tetracycline. These proteins revealed interesting difference in e.g. proteins involved in peptidoglycan-based cell wall proteins and energy metabolism. Upon treatment, 12 proteins were differentially regulated showing more than 2-fold change and p<0.05 (p value corrected for multiple testing). This integrated study using high resolution mass spectrometry based label-free quantitative proteomics to study tetracycline antibiotic response in the soluble proteome of resistant E. coli provides novel insight into tetracycline related stress. SIGNIFICANCE: The lack of new antibiotics to fight infections caused by multidrug resistant microorganisms has motivated the use of old antibiotics, and the search for new drug targets. The evolution of antibiotic resistance is complex, but it is known that agroecosystems play an important part in the selection of antibiotic resistance bacteria. Tetracyclines are still used as phytopharmaceutical agents in crops, selecting resistant bacteria and changing the ecology of farm soil. Little is known about the metabolic response of genetically resistant populations to antibiotic exposure. Indeed, to date there are no quantitative tetracycline resistance studies performed with the latest generation of high resolution mass spectrometers allowing high mass accuracy in both MS and MS/MS scans. Here, we report the proteome profiling of a soil-borne Escherichia coli upon tetracycline stress, so that this new perspective could provide a broaden understanding of the metabolic responses of E. coli to a widely used antibiotic.


Asunto(s)
Farmacorresistencia Bacteriana , Escherichia coli/fisiología , Metaboloma/efectos de los fármacos , Peptidoglicano/efectos de los fármacos , Proteoma/efectos de los fármacos , Tetraciclina/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Proteoma/análisis , Microbiología del Suelo , Contaminantes del Suelo/farmacología , Espectrometría de Masas en Tándem
16.
J Microbiol ; 55(5): 403-408, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28124780

RESUMEN

Bacteriophage endolysin is one of the most promising antibiotic substitutes, but in Gram-negative bacteria, the outer membrane prevents the lysin from hydrolyzing peptidoglycans and blocks the development of lysin applications. The prime strategy for new antibiotic substitutes is allowing lysin to access the peptidoglycan from outside of the bacteria by reformation of the lysin. In this study, the novel Escherichia coli (E. coli) phage lyase lysep3, which lacks outside-in catalytic ability, was fused with the N-terminal region of the Bacillus amyloliquefaciens lysin including its cell wall binding domain D8 through the best manner of protein fusion based on the predicted tertiary structure of lysep3-D8 to obtain an engineered lysin that can lyse bacteria from the outside. Our results showed that lysep3-D8 could lyse both Gramnegative and Gram-positive bacteria, whereas lysep3 and D8 have no impact on bacterial growth. The MIC of lysep3-D8 on E. coli CVCC1418 is 60 µg/ml; lysep3-D8 can inhibit the growth of bacteria up to 12 h at this concentration. The bactericidal spectrum of lysep3-D8 is broad, as it can lyse of all of 14 E. coli strains, 3 P. aeruginosa strains, 1 Acinetobacter baumannii strain, and 1 Streptococcus strain. Lysep3-D8 has sufficient bactericidal effects on the 14 E. coli strains tested at the concentration of 100 µg/ml. The cell wall binding domain of the engineered lysin can destroy the integrity of the outer membrane of bacteria, thus allowing the catalytic domain to reach its target, peptidoglycan, to lyse the bacteria. Lysep3-D8 can be used as a preservative in fodder to benefit the health of animals. The method we used here proved to be a successful exploration of the reformation of phage lysin.


Asunto(s)
Antibacterianos/farmacología , Fagos de Bacillus/química , Bacillus amyloliquefaciens/genética , Bacteriólisis , Colifagos/química , Endopeptidasas/genética , Endopeptidasas/farmacología , Proteínas Virales/farmacología , Acinetobacter baumannii/efectos de los fármacos , Animales , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Peptidoglicano/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Proteínas Virales/genética
17.
Clin Infect Dis ; 42 Suppl 1: S25-34, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16323116

RESUMEN

The first vancomycin-resistant clinical isolates of Enterococcus species were reported in Europe in 1988. Similar strains were later detected in hospitals on the East Coast of the United States. Since then, vancomycin-resistant enterococci have spread with unexpected rapidity and are now encountered in hospitals in most countries. This article reviews the mode of action and the mechanism of bacterial resistance to glycopeptides, as exemplified by the VanA type, which is mediated by transposon Tn1546 and is widely spread in enterococci. The diversity, regulation, evolution, and recent dissemination of methicillin-resistant Staphylococcus aureus are then discussed.


Asunto(s)
Antibacterianos/farmacología , Enterococcus/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Resistencia a la Vancomicina/fisiología , Vancomicina/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Ligasas de Carbono-Oxígeno/genética , Ligasas de Carbono-Oxígeno/fisiología , Farmacorresistencia Microbiana/genética , Farmacorresistencia Microbiana/fisiología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/fisiología , Glicopéptidos/fisiología , Ligasas/genética , Ligasas/fisiología , Péptido Sintasas/genética , Peptidoglicano/biosíntesis , Peptidoglicano/efectos de los fármacos
18.
Pol J Microbiol ; 55(4): 279-88, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17416064

RESUMEN

The susceptibility of 96 strains of Listeria monocytogenes isolated from food to antibiotics and disinfectants currently used in human therapy, veterinary, medicine and food industry was determined by a standard operating procedure--broth dilution method. Antimicrobial agents included the beta-lactams ampicillin and penicillin, the lantibiotic nisin, and the disinfectants benzalkonium chloride and chlorhexidine gluconate. Among the studied strains we found 13 strains with 8-fold, 7 strains with 16-fold and 2 strains with 32-fold decreased susceptibility to ampicillin, as determined by MIC, compared to wild type reference strain. Interestingly, the mentioned strains were isolated from frozen vegetables and soups, none of the isolates from dairy products showed any elevated resistance to the studied antimicrobial agents. The occurrence in food products of strains with increased resistance to ampicillin is disquieting, especially since 3-lactams are the most frequent antibiotic of choice in the therapy of infections caused by the pathogen.


Asunto(s)
Antiinfecciosos/farmacología , Compuestos de Benzalconio/farmacología , Clorhexidina/análogos & derivados , Listeria monocytogenes/efectos de los fármacos , Nisina/farmacología , Penicilinas/farmacología , Ampicilina/farmacología , Bacteriólisis , Clorhexidina/farmacología , Productos Lácteos/microbiología , Desinfectantes/farmacología , Alimentos Congelados/microbiología , Listeria monocytogenes/aislamiento & purificación , Listeria monocytogenes/metabolismo , Proteínas de la Membrana/análisis , Pruebas de Sensibilidad Microbiana , Peptidoglicano/biosíntesis , Peptidoglicano/efectos de los fármacos , Factores de Tiempo , Verduras/microbiología
19.
FEMS Microbiol Rev ; 26(5): 511-32, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12586393

RESUMEN

Oritavancin (LY333328) is a semisynthetic glycopeptide antibiotic having excellent bactericidal activity against glycopeptide-susceptible and -resistant Gram-positive bacteria. Oritavancin is the N-alkyl-p-chlorophenylbenzyl derivative of chloroeremomycin (LY264826) and is currently in phase III clinical trials for use in Gram-positive infections. Studies show that oritavancin and related alkyl glycopeptides inhibit bacterial cell wall formation by blocking the transglycosylation step in peptidoglycan biosynthesis in a substrate-dependent manner. As with other glycopeptide antibiotics, including vancomycin, the effects of oritavancin on cell wall synthesis are attributable to interactions with dipeptidyl residues of peptidoglycan precursors. Unlike vancomycin, however, oritavancin is strongly dimerized and can anchor to the cytoplasmic membrane, the latter facilitated by its alkyl side chain. Cooperative interactions derived from dimerization and membrane anchoring in situ can be of sufficient strength to enable binding to either dipeptidyl or didepsipeptidyl peptidoglycan residues of vancomycin-susceptible and -resistant enterococci, respectively. This review describes the antibacterial activity of oritavancin, and examines the evidence supporting the proposed mechanism of action for this agent and related analogs.


Asunto(s)
Antibacterianos/farmacología , Bacterias Grampositivas/efectos de los fármacos , Vancomicina/análogos & derivados , Antibacterianos/metabolismo , Farmacorresistencia Microbiana , Glicopéptidos , Bacterias Grampositivas/clasificación , Infecciones por Bacterias Grampositivas/prevención & control , Humanos , Lipoglucopéptidos , Modelos Moleculares , Peptidoglicano/efectos de los fármacos , Vancomicina/química , Vancomicina/metabolismo , Vancomicina/farmacología , Resistencia a la Vancomicina/fisiología
20.
Future Microbiol ; 11: 1315-1338, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27633726

RESUMEN

AIM: To evaluate the antibacterial and antibiofilm mechanisms of usnic acid (USN) against methicillin-resistant Staphylococcus aureus from cystic fibrosis patients. MATERIALS & METHODS: The effects exerted by USN at subinhibitory concentrations on S. aureus Sa3 strain was evaluated by proteomic, real-time PCR and electron microscopy analyses. RESULTS & CONCLUSION: Proteomic analysis showed that USN caused damage in peptidoglycan synthesis, as confirmed by microscopy. Real-time PCR analysis showed that antibiofilm activity of USN is mainly due to impaired adhesion to the host matrix binding proteins, and decreasing lipase and thermonuclease expression. Our data show that USN exerts anti-staphylococcal effects through multitarget inhibitory effects, thus confirming the rationale for considering it 'lead compound' for the treatment of cystic fibrosis infections.


Asunto(s)
Antibacterianos/farmacología , Benzofuranos/farmacología , Biopelículas/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Adhesinas Bacterianas/efectos de los fármacos , Antibacterianos/administración & dosificación , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/efectos de los fármacos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Benzofuranos/administración & dosificación , Proteínas Portadoras/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Recuento de Colonia Microbiana , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/microbiología , ADN Bacteriano , Regulación hacia Abajo , Lipasa/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Staphylococcus aureus Resistente a Meticilina/ultraestructura , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Microscopía Electrónica de Rastreo/métodos , Microscopía Electrónica de Transmisión/métodos , Peptidoglicano/biosíntesis , Peptidoglicano/efectos de los fármacos , Propidio/metabolismo , Mapas de Interacción de Proteínas , Proteómica/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Infecciones Estafilocócicas/microbiología , Factores de Tiempo , Virulencia/efectos de los fármacos , Virulencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA