Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Infect Immun ; 89(4)2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33468580

RESUMEN

Mutation of purR was previously shown to enhance the virulence of Staphylococcus aureus in a murine sepsis model, and this cannot be fully explained by increased expression of genes within the purine biosynthesis pathway. Rather, the increased production of specific S. aureus virulence factors, including alpha toxin and the fibronectin-binding proteins, was shown to play an important role. Mutation of purR was also shown previously to result in increased abundance of SarA. Here, we demonstrate by transposon sequencing that mutation of purR in the USA300 strain LAC increases fitness in a biofilm while mutation of sarA has the opposite effect. Therefore, we assessed the impact of sarA on reported purR-associated phenotypes by characterizing isogenic purR, sarA, and sarA/purR mutants. The results confirmed that mutation of purR results in increased abundance of alpha toxin, protein A, the fibronectin-binding proteins, and SarA, decreased production of extracellular proteases, an increased capacity to form a biofilm, and increased virulence in an osteomyelitis model. Mutation of sarA had the opposite effects on all of these phenotypes and, other than bacterial burdens in the bone, all of the phenotypes of sarA/purR mutants were comparable to those of sarA mutants. Limiting the production of extracellular proteases reversed all of the phenotypes of sarA mutants and most of those of sarA/purR mutants. We conclude that a critical component defining the virulence of a purR mutant is the enhanced production of SarA, which limits protease production to an extent that promotes the accumulation of critical S. aureus virulence factors.


Asunto(s)
Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Endopeptidasas/biosíntesis , Mutación , Proteínas Represoras/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/fisiología , Transactivadores/biosíntesis , Factores de Virulencia/genética , Animales , Biopelículas/crecimiento & desarrollo , Elementos Transponibles de ADN , Susceptibilidad a Enfermedades , Espacio Extracelular , Regulación Bacteriana de la Expresión Génica , Ratones , Osteomielitis/microbiología , Staphylococcus aureus/patogenicidad , Virulencia/genética
2.
Infect Immun ; 88(2)2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31740526

RESUMEN

The staphylococcal accessory regulator (sarA) plays an important role in Staphylococcus aureus infections, including osteomyelitis, and the msaABCR operon has been implicated as an important factor in modulating expression of sarA Thus, we investigated the contribution of msaABCR to sarA-associated phenotypes in the S. aureus clinical isolates LAC and UAMS-1. Mutation of msaABCR resulted in reduced production of SarA and a reduced capacity to form a biofilm in both strains. Biofilm formation was enhanced in a LAC msa mutant by restoring the production of SarA, but this was not true in a UAMS-1 msa mutant. Similarly, extracellular protease production was increased in a LAC msa mutant but not a UAMS-1 msa mutant. This difference was reflected in the accumulation and distribution of secreted virulence factors and in the impact of extracellular proteases on biofilm formation in a LAC msa mutant. Most importantly, it was reflected in the relative impact of mutating msa as assessed in a murine osteomyelitis model, which had a significant impact in LAC but not in UAMS-1. In contrast, mutation of sarA had a greater impact on all of these in vitro and in vivo phenotypes than mutation of msaABCR, and it did so in both LAC and UAMS-1. These results suggest that, at least in osteomyelitis, it would be therapeutically preferable to target sarA rather than msaABCR to achieve the desired clinical result, particularly in the context of divergent clinical isolates of S. aureus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Fenotipo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/aislamiento & purificación , Staphylococcus aureus/fisiología , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Biopelículas/crecimiento & desarrollo , Modelos Animales de Enfermedad , Genotipo , Ratones Endogámicos C57BL , Mutación , Osteomielitis/microbiología , Osteomielitis/patología , Staphylococcus aureus/clasificación , Factores de Virulencia/genética
3.
J Proteome Res ; 17(10): 3384-3395, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30209945

RESUMEN

The staphylococcal accessory regulator A ( sarA) impacts the extracellular accumulation of Staphylococcus aureus virulence factors at the level of intracellular production and extracellular protease-mediated degradation. We previously used a proteomics approach that measures protein abundance of all proteoforms to demonstrate that mutation of sarA results in increased levels of extracellular proteases and assesses the impact of this on the accumulation of S. aureus exoproteins. Our previous approach was limited as it did not take into account that large, stable proteolytic products from a given protein could result in false negatives when quantified by total proteoforms. Here, our goal was to use an expanded proteomics approach utilizing a dual quantitative method for measuring abundance at both the total proteoform and full-length exoprotein levels to alleviate these false negatives and thereby provide for characterization of protease-dependent and -independent effects of sarA mutation on the S. aureus exoproteome. Proteins present in conditioned medium from overnight, stationary phase cultures of the USA300 strain LAC, an isogenic sarA mutant, and a sarA mutant unable to produce any of the known extracellular proteases ( sarA/protease) were resolved using one-dimensional gel electrophoresis. Quantitative proteomic comparisons of sarA versus sarA/protease mutants identified proteins that were cleaved in a protease-dependent manner owing to mutation of sarA, and comparisons of sarA/protease mutant versus the LAC parent strain identified proteins in which abundance was altered in a sarA mutant in a protease-independent manner. Furthermore, the proteins uniquely identified by the full-length data analysis approach eliminated false negatives observed in the total proteoform analysis. This expanded approach provided for a more comprehensive analysis of the impact of mutating sarA on the S. aureus exoproteome.


Asunto(s)
Proteínas Bacterianas/metabolismo , Péptido Hidrolasas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Staphylococcus aureus/metabolismo , Proteínas Bacterianas/genética , Biopelículas , Regulación Bacteriana de la Expresión Génica , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/metabolismo , Mutación/genética , Proteoma/genética , Staphylococcus aureus/genética , Staphylococcus aureus/fisiología , Espectrometría de Masas en Tándem , Virulencia/genética
4.
Int J Hyperthermia ; 34(2): 209-219, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29025325

RESUMEN

BACKGROUND: We previously demonstrated that a photoactivatable therapeutic approach employing antibiotic-loaded, antibody-conjugated, polydopamine (PDA)-coated gold nanocages (AuNCs) could be used for the synergistic killing of bacterial cells within a biofilm. The approach was validated with a focus on Staphylococcus aureus using an antibody specific for staphylococcal protein A (Spa) and an antibiotic (daptomycin) active against Gram-positive cocci including methicillin-resistant S. aureus (MRSA). However, an important aspect of this approach is its potential therapeutic versatility. METHODS: In this report, we evaluated this versatility by examining the efficacy of AuNC formulations generated with alternative antibodies and antibiotics targeting S. aureus and alternative combinations targeting the Gram-negative pathogen Pseudomonas aeruginosa. RESULTS: The results confirmed that daptomycin-loaded AuNCs conjugated to antibodies targeting two different S. aureus lipoproteins (SACOL0486 and SACOL0688) also effectively kill MRSA in the context of a biofilm. However, our results also demonstrate that antibiotic choice is critical. Specifically, ceftaroline and vancomycin-loaded AuNCs conjugated to anti-Spa antibodies were found to exhibit reduced efficacy relative to daptomycin-loaded AuNCs conjugated to the same antibody. In contrast, gentamicin-loaded AuNCs conjugated to an antibody targeting a conserved outer membrane protein were highly effective against P. aeruginosa biofilms. CONCLUSIONS: These results confirm the therapeutic versatility of our approach. However, to the extent that its synergistic efficacy is dependent on the ability to achieve both a lethal photothermal effect and the thermally controlled release of a sufficient amount of antibiotic, they also demonstrate the importance of carefully designing appropriate antibody and antibiotic combinations to achieve the desired therapeutic synergy.


Asunto(s)
Antibacterianos/uso terapéutico , Infecciones Bacterianas/terapia , Oro/metabolismo , Nanopartículas/metabolismo , Antibacterianos/farmacología , Infecciones Bacterianas/patología , Biopelículas , Humanos
5.
Clin Orthop Relat Res ; 475(7): 1857-1870, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28097503

RESUMEN

BACKGROUND: Local drug delivery devices offer a promising method for delivering vancomycin and amikacin for musculoskeletal wounds. However, current local delivery devices such as beads and sponges do not necessarily allow for full coverage of a wound surface with eluted antibiotics and do not address the need for reducing the antibiotic diffusion distance to help prevent contamination by bacteria or other microorganisms. We blended chitosan/polyethylene glycol (PEG) pastes/sponges to increase biocompatibility and improve antibiotic coverage within the wound. QUESTIONS/PURPOSES: (1) Are blended chitosan/PEG pastes biodegradable? (2) Are the blended pastes biocompatible? (3) How much force does paste require for placement by injection? (4) Will the pastes elute active antibiotics to inhibit bacteria in vitro? (5) Can the pastes prevent infection in a preclinical model with hardware? METHODS: Our blended paste/sponge formulations (0.5% acidic, 1% acidic, and acidic/neutral) along with a control neutral 1% chitosan sponge were tested in vitro for degradability, cytocompatibility, injectability tested by determining the amount of force needed to inject the pastes, elution of antibiotics, and activity tested using zone of inhibition studies. Along with these studies, in vivo models for biocompatibility and infection prevention were tested using a rodent model and an infected mouse model with hardware, respectively. By evaluating these characteristics, an improved local drug delivery device can be determined. RESULTS: All three of the paste formulations evaluated were almost fully degraded and with 6 days of degradation, the percent remaining being was less than that of the control sponge (percent remaining: control 99.251% ± 1.0%; 0.5% acidic 1.6% ± 2.1%, p = 0.002; 1% acidic 1.7% ± 1.6%, p = 0.002; acidic/neutral 2.3% ± 1.7%, p = 0.010). There was good biocompatibility because cell viability in vitro was high (control 100.0 ± 14.3; 0.5% acidic formulation at 79.4 ± 12.6, p < 0.001; 1% acidic formulation at 98.6 ± 6.1, p = 0.993; acidic/neutral formulation at 106.7 ± 12.8, p = 0.543), and in vivo inflammation was moderate (control 2.1 ± 1.2; 0.5% acidic 3.3 ± 0.2, p = 0.530; 1% acidic 2.5 ± 0.9, p = 0.657; acidic/neutral 2.9 ± 1.1, p = 0.784). Force required to inject the 0.5% acidic and 1% acidic pastes was less than the acidic/neutral paste used as a control (control 167.7 ± 85.6; 0.5% acidic 41.3 ± 10.7, p = 0.070; 1% acidic 28.0 ± 7.0, p = 0.940). At 72 hours, all paste formulations exhibited in vitro activity against Staphylococcus aureus (control 2.6 ± 0.8; 0.5% acidic 98.1 ± 33.5, p = 0.002; 1% acidic 87.3 ± 17.2, p = 0.006; acidic/neutral 83.5 ± 14.3, p = 0.010) and Pseudomonas aeruginosa (control 163.0 ± 1.7; 0.5% acidic 85.7 ± 83.6, p = 0.373; 1% acidic 38.0 ± 45.1, p = 0.896; acidic/neutral 129.7 ± 78.0, p = 0.896). Also, the paste formulations were able to prevent the infection with 100% clearance on the implanted hardware and surrounding tissue with the control being a 0.5% acidic paste group without antibiotics (control 4 × 104 ± 4.8 × 104; 0.5% acidic 0.0 ± 0.0, p value: 0.050; 1% acidic 0.0 ± 0.0, p = 0.050; acidic/neutral 0.0 ± 0.0, p = 0.050). CONCLUSIONS: The preliminary studies demonstrated promising results for the blended chitosan/PEG pastes with antibiotics provided degradability, biocompatibility, injectability, and infection prevention for musculoskeletal-type wounds. CLINICAL RELEVANCE: The preliminary studies with the chitosan paste delivered antibiotics to a contaminated musculoskeletal wound with hardware and prevented infection. More studies in a complex musculoskeletal wound and dosage studies are needed for continued development.


Asunto(s)
Antibacterianos/administración & dosificación , Materiales Biocompatibles/administración & dosificación , Quitosano/administración & dosificación , Portadores de Fármacos , Polietilenglicoles/administración & dosificación , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Combinación de Medicamentos , Técnicas In Vitro , Ratones , Pseudomonas aeruginosa/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos
6.
Clin Orthop Relat Res ; 475(7): 1847-1853, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28050817

RESUMEN

BACKGROUND: Phosphatidylcholine coatings have been shown to elute antibiotics for several days. A recently developed biofilm inhibitor, cis-2-decenoic acid (C2DA), has been shown to exhibit synergistic activity with several common antibiotics. This study aims to evaluate the effectiveness of C2DA and amikacin dual drug delivery from a phosphatidylcholine coating. QUESTIONS/PURPOSES: (1) What are the in vitro elution profiles of amikacin and C2DA from phosphatidylcholine-coated coupons in incubated phosphate-buffered saline? (2) Does the presence of C2DA in eluate samples lower the amount of amikacin needed for bacterial inhibition in overnight bacterial turbidity assays? (3) Does addition of amikacin and C2DA result in decreased colony-forming units (CFUs) on wire implants and bone when compared with phosphatidylcholine coatings alone in a mouse model of periprosthetic joint infection? METHODS: Effects of loading concentrations were assessed during 7-day in vitro elution studies for coatings containing all mixtures of 0%, 5%, 15%, and 25% wt of amikacin and C2DA (n = 4) through quantitative high-performance liquid chromatography concentration determination and plotting concentration eluted over time. Antimicrobial activity was assessed by overnight turbidity testing of elution study samples against Staphylococcus aureus or Pseudomonas aeruginosa. In vivo efficacy was assessed using phosphatidylcholine-coated wire implants in a murine (mouse) model of infection (n = 3). Wire implants were coated with phosphatidylcholine containing no antimicrobials, amikacin alone, C2DA alone, or amikacin and C2DA and then inserted into the intramedullary femur of each mouse and inoculated with S aureus. The number of viable bacterial colonies on the implant surface and in the surrounding bone was determined after 1 week with the goal of achieving complete bacterial clearance. Total viable CFU count and proportion of samples achieving complete clearance were compared between groups. RESULTS: Elution samples showed a burst response of amikacin and C2DA for 1 to 2 days with C2DA release continuing at low levels through Day 4. All tested eluate samples inhibited P aeruginosa. Samples from coatings containing 25% amikacin or 15% amikacin and any amount of C2DA were able to inhibit S aureus formation, but all coatings with 5% amikacin or 15% amikacin but no C2DA were not inhibitory. All in vivo treatment groups achieved complete bacterial clearance on the wire implant, and the C2DA alone and amikacin alone coatings cleared all CFUs in bone (pin: phosphatidylcholine only one of three; amikacin three of three, C2DA three of three, amikacin + C2DA three of three, p = 0.04 [Fisher's exact test]; bone: coating only: zero of three; amikacin: three of three; C2DA; three of three; C2DA + amikacin: one of three; p = 0.03 [Fisher's exact test]). CONCLUSIONS: Phosphatidylcholine coatings elute antimicrobials in vitro under infinite sink conditions for up to 4 days in phosphate-buffered saline and were able to reduce bacterial colonies in a preliminary in vivo model. Turbidity testing with eluate samples containing varying amounts of C2DA and amikacin agrees with previous studies showing synergy between them. CLINICAL RELEVANCE: Used as an adjunctive to systemic therapy, C2DA-loaded phosphatidylcholine coatings have potential value as a prophylactic infection prevention measure. Future studies may include different antibiotics, animal studies with larger sample sizes and more controls, and advanced coating delivery methods.


Asunto(s)
Amicacina/administración & dosificación , Amicacina/farmacología , Biopelículas/efectos de los fármacos , Materiales Biocompatibles Revestidos , Portadores de Fármacos , Ácidos Grasos Monoinsaturados/farmacología , Fosfatidilcolinas/farmacología , Animales , Cromatografía Líquida de Alta Presión , Técnicas In Vitro , Ratones , Pruebas de Sensibilidad Microbiana , Pseudomonas aeruginosa/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos
7.
Infect Immun ; 84(4): 1214-1225, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26857575

RESUMEN

We demonstrate that mutation of xerC, which reportedly encodes a homologue of an Escherichia coli recombinase, limits biofilm formation in the methicillin-resistant Staphylococcus aureus strain LAC and the methicillin-sensitive strain UAMS-1. This was not due to the decreased production of the polysaccharide intracellular adhesin (PIA) in either strain because the amount of PIA was increased in a UAMS-1xerC mutant and undetectable in both LAC and its isogenic xerC mutant. Mutation of xerC also resulted in the increased production of extracellular proteases and nucleases in both LAC and UAMS-1, and limiting the production of either class of enzymes increased biofilm formation in the isogenic xerC mutants. More importantly, the limited capacity to form a biofilm was correlated with increased antibiotic susceptibility in both strains in the context of an established biofilm in vivo. Mutation of xerC also attenuated virulence in a murine bacteremia model, as assessed on the basis of the bacterial loads in internal organs and overall lethality. It also resulted in the decreased accumulation of alpha toxin and the increased accumulation of protein A. These findings suggest that xerC may impact the functional status of agr. This was confirmed by demonstrating the reduced accumulation of RNAIII and AgrA in LAC and UAMS-1xerC mutants. However, this cannot account for the biofilm-deficient phenotype of xerC mutants because mutation of agr did not limit biofilm formation in either strain. These results demonstrate that xerC contributes to biofilm-associated infections and acute bacteremia and that this is likely due to agr-independent and -dependent pathways, respectively.


Asunto(s)
Proteínas Bacterianas/metabolismo , Biopelículas/crecimiento & desarrollo , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Péptidos Cíclicos/metabolismo , Recombinasas/metabolismo , Staphylococcus aureus/enzimología , Proteínas Bacterianas/genética , Staphylococcus aureus Resistente a Meticilina/enzimología , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/metabolismo , Mutación , Operón , Péptidos Cíclicos/genética , Recombinasas/genética , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
8.
Infect Immun ; 84(9): 2586-94, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27354444

RESUMEN

We used a murine model of acute, posttraumatic osteomyelitis to evaluate the virulence of two divergent Staphylococcus aureus clinical isolates (the USA300 strain LAC and the USA200 strain UAMS-1) and their isogenic sarA mutants. The results confirmed that both strains caused comparable degrees of osteolysis and reactive new bone formation in the acute phase of osteomyelitis. Conditioned medium (CM) from stationary-phase cultures of both strains was cytotoxic to cells of established cell lines (MC3TC-E1 and RAW 264.7 cells), primary murine calvarial osteoblasts, and bone marrow-derived osteoclasts. Both the cytotoxicity of CM and the reactive changes in bone were significantly reduced in the isogenic sarA mutants. These results confirm that sarA is required for the production and/or accumulation of extracellular virulence factors that limit osteoblast and osteoclast viability and that thereby promote bone destruction and reactive bone formation during the acute phase of S. aureus osteomyelitis. Proteomic analysis confirmed the reduced accumulation of multiple extracellular proteins in the LAC and UAMS-1 sarA mutants. Included among these were the alpha class of phenol-soluble modulins (PSMs), which were previously implicated as important determinants of osteoblast cytotoxicity and bone destruction and repair processes in osteomyelitis. Mutation of the corresponding operon reduced the cytotoxicity of CM from both UAMS-1 and LAC cultures for osteoblasts and osteoclasts. It also significantly reduced both reactive bone formation and cortical bone destruction by CM from LAC cultures. However, this was not true for CM from cultures of a UAMS-1 psmα mutant, thereby suggesting the involvement of additional virulence factors in such strains that remain to be identified.


Asunto(s)
Proteínas Bacterianas/genética , Osteomielitis/microbiología , Osteomielitis/patología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Factores de Virulencia/genética , Virulencia/genética , Animales , Regulación Bacteriana de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Operón/genética , Osteoblastos/microbiología , Osteoblastos/patología , Osteoclastos/microbiología , Osteoclastos/patología , Proteómica/métodos , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología
9.
Antimicrob Agents Chemother ; 60(10): 5688-94, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27401574

RESUMEN

We used in vitro and in vivo models of catheter-associated biofilm formation to compare the relative activity of antibiotics effective against methicillin-resistant Staphylococcus aureus (MRSA) in the specific context of an established biofilm. The results demonstrated that, under in vitro conditions, daptomycin and ceftaroline exhibited comparable activity relative to each other and greater activity than vancomycin, telavancin, oritavancin, dalbavancin, or tigecycline. This was true when assessed using established biofilms formed by the USA300 methicillin-resistant strain LAC and the USA200 methicillin-sensitive strain UAMS-1. Oxacillin exhibited greater activity against UAMS-1 than LAC, as would be expected, since LAC is an MRSA strain. However, the activity of oxacillin was less than that of daptomycin and ceftaroline even against UAMS-1. Among the lipoglycopeptides, telavancin exhibited the greatest overall activity. Specifically, telavancin exhibited greater activity than oritavancin or dalbavancin when tested against biofilms formed by LAC and was the only lipoglycopeptide capable of reducing the number of viable bacteria below the limit of detection. With biofilms formed by UAMS-1, telavancin and dalbavancin exhibited comparable activity relative to each other and greater activity than oritavancin. Importantly, ceftaroline was the only antibiotic that exhibited greater activity than vancomycin when tested in vivo in a murine model of catheter-associated biofilm formation. These results emphasize the need to consider antibiotics other than vancomycin, most notably, ceftaroline, for the treatment of biofilm-associated S. aureus infections, including by the matrix-based antibiotic delivery methods often employed for local antibiotic delivery in the treatment of these infections.


Asunto(s)
Antibacterianos/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Aminoglicósidos/farmacología , Animales , Biopelículas/efectos de los fármacos , Infecciones Relacionadas con Catéteres/tratamiento farmacológico , Infecciones Relacionadas con Catéteres/microbiología , Evaluación Preclínica de Medicamentos/métodos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Glicopéptidos/farmacología , Lipoglucopéptidos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Ratones , Pruebas de Sensibilidad Microbiana , Teicoplanina/análogos & derivados , Teicoplanina/farmacología
10.
Antimicrob Agents Chemother ; 60(3): 1826-9, 2016 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-26824954

RESUMEN

We previously determined the extent to which mutations of different Staphylococcus aureus regulatory loci impact biofilm formation as assessed under in vitro conditions. Here we extend these studies to determine the extent to which those regulatory loci that had the greatest effect on biofilm formation also impact antibiotic susceptibility. The experiments were done under in vitro and in vivo conditions using two clinical isolates of S. aureus (LAC and UAMS-1) and two functionally diverse antibiotics (daptomycin and ceftaroline). Mutation of the staphylococcal accessory regulator (sarA) or sigB was found to significantly increase susceptibilities to both antibiotics and in both strains in a manner that could not be explained by changes in the MICs. The impact of a mutation in sarA was comparable to that of a mutation in sigB and greater than the impact observed with any other mutant. These results suggest that therapeutic strategies targeting sarA and/or sigB have the greatest potential to facilitate the ability to overcome the intrinsic antibiotic resistance that defines S. aureus biofilm-associated infections.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/genética , Biopelículas/efectos de los fármacos , Cefalosporinas/farmacología , Daptomicina/farmacología , Factor sigma/genética , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Animales , Biopelículas/crecimiento & desarrollo , Infecciones Relacionadas con Catéteres/tratamiento farmacológico , Infecciones Relacionadas con Catéteres/microbiología , Catéteres/microbiología , Farmacorresistencia Bacteriana/genética , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/aislamiento & purificación , Staphylococcus aureus/patogenicidad , Ceftarolina
11.
Mol Microbiol ; 92(6): 1299-312, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24779437

RESUMEN

We investigated the in vivo relevance of the impact of sarA and saeRS on protease production using derivatives of the USA300 strain LAC. The results confirmed that mutation of saeRS or sarA reduces virulence in a bacteremia model to a comparable degree. However, while eliminating protease production restored virulence in the sarA mutant, it had little impact in the saeRS mutant. Additionally, constitutive activation of saeRS (saeRS(C)) enhanced the virulence of LAC and largely restored virulence in the isogenic sarA mutant. Based on these results, together with our analysis of the representative virulence factors alpha toxin, protein A (Spa), and extracellular nucleases, we propose a model in which the attenuation of saeRS mutants is defined primarily by decreased production of such factors, while constitutive activation of saeRS increases virulence, and reverses the attenuation of sarA mutants, because it results in both increased production and decreased protease-mediated degradation of these same factors. This regulatory balance was also apparent in a murine model of catheter-associated infection, with the results suggesting that the impact of saeRS on nuclease production plays an important role during the early stages of these infections that is partially offset by increased protease production in sarA mutants.


Asunto(s)
Proteínas Bacterianas/metabolismo , Staphylococcus aureus Resistente a Meticilina/enzimología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Péptido Hidrolasas/metabolismo , Proteínas Quinasas/metabolismo , Animales , Bacteriemia/microbiología , Bacteriemia/patología , Proteínas Bacterianas/genética , Infecciones Relacionadas con Catéteres/microbiología , Infecciones Relacionadas con Catéteres/patología , Modelos Animales de Enfermedad , Ratones , Factores de Transcripción , Virulencia
12.
Clin Orthop Relat Res ; 473(7): 2270-82, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25604874

RESUMEN

BACKGROUND: Orthopaedic biomaterials are susceptible to biofilm formation. A novel lipid-based material has been developed that may be loaded with antibiotics and applied as an implant coating at point of care. However, this material has not been evaluated for antibiotic elution, biofilm inhibition, or in vivo efficacy. QUESTIONS/PURPOSES: (1) Do antibiotic-loaded coatings inhibit biofilm formation? (2) Is the coating effective in preventing biofilm in vivo? METHODS: Purified phosphatidylcholine was mixed with 25% amikacin or vancomycin or a combination of 12.5% of both. A 7-day elution study for coated titanium and stainless steel coupons was followed by turbidity and zone of inhibition assays against Staphylococcus aureus and Pseudomonas aeruginosa. Coupons were inoculated with bacteria and incubated 24 hours (N = 4 for each test group). Microscopic images of biofilm were obtained. After washing and vortexing, attached bacteria were counted. A mouse biofilm model was modified to include coated and uncoated stainless steel wires inserted into the lumens of catheters inoculated with a mixture of S aureus or P aeruginosa. Colony-forming unit counts (N = 10) and scanning electron microscopy imaging of implants were used to determine antimicrobial activity. RESULTS: Active antibiotics with colony inhibition effects were eluted for up to 6 days. Antibiotic-loaded coatings inhibited biofilm formation on in vitro coupons (log-fold reductions of 4.3 ± 0.4 in S aureus and 3.1 ± 0 for P aeruginosa in phosphatidylcholine-only coatings, 5.6 ± 0 for S aureus and 3.1 ± 0 for P aeruginosa for combination-loaded coatings, 5.5 ± 0.3 for S aureus in vancomycin-loaded coatings, and 3.1 ± 0 for P aeruginosa for amikacin-loaded coatings (p < 0.001 for all comparisons of antibiotic-loaded coatings against uncoated controls for both bacterial strains, p < 0.001 for comparison of antibiotic-loaded coatings against phosphatidylcholine only for S aureus, p = 0.54 for comparison of vancomycin versus combination coating in S aureus, P = 0.99 for comparison of antibiotic- and unloaded phosphatidylcholine coatings in P aeruginosa). Similarly, antibiotic-loaded coatings reduced attachment of bacteria to wires in vivo (log-fold reduction of 2.54 ± 0; p < 0.001 for S aureus and 0.83 ± 0.3; p = 0.112 for P aeruginosa). CONCLUSIONS: Coatings deliver active antibiotics locally to inhibit biofilm formation and bacterial growth in vivo. Future evaluations will include orthopaedic preclinical models to confirm therapeutic efficacy. CLINICAL RELEVANCE: Clinical applications of local drug delivery coating could reduce the rate of implant-associated infections.


Asunto(s)
Amicacina/administración & dosificación , Antibacterianos/administración & dosificación , Biopelículas/efectos de los fármacos , Materiales Biocompatibles Revestidos , Portadores de Fármacos , Prótesis e Implantes , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Vancomicina/administración & dosificación , Amicacina/farmacología , Animales , Antibacterianos/farmacología , Ratones , Sistemas de Atención de Punto , Vancomicina/farmacología
13.
mBio ; 15(4): e0338323, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38415646

RESUMEN

We previously demonstrated that mutation of sarA in Staphylococcus aureus limits biofilm formation, cytotoxicity for osteoblasts and osteoclasts, and virulence in osteomyelitis, and that all of these phenotypes can be attributed to the increased production of extracellular proteases. Here we extend these studies to assess the individual importance of these proteases alone and in combination with each other using the methicillin-resistant USA300 strain LAC, the methicillin-susceptible USA200 strain UAMS-1, and isogenic sarA mutants that were also unable to produce aureolysin (Aur), staphopain A (ScpA), staphylococcal serine protease A (subsp.), staphopain B (SspB), and the staphylococcal serine protease-like proteins A-F (SplA-F). Biofilm formation was restored in LAC and UAMS-1 sarA mutants by subsequent mutation of aur and scpA, while mutation of aur had the greatest impact on cytotoxicity to mammalian cells, particularly with conditioned medium (CM) from the more cytotoxic strain LAC. However, SDS-PAGE and western blot analysis of CM confirmed that mutation of sspAB was also required to mimic the phenotype of sarA mutants unable to produce any extracellular proteases. Nevertheless, in a murine model of post-traumatic osteomyelitis, mutation of aur and scpA had the greatest impact on restoring the virulence of LAC and UAMS-1 sarA mutants, with concurrent mutation of sspAB and the spl operon having relatively little effect. These results demonstrate that the increased production of Aur and ScpA in combination with each other is a primary determinant of the reduced virulence of S. aureus sarA mutants in diverse clinical isolates including both methicillin-resistant and methicillin-susceptible strains.IMPORTANCEPrevious work established that SarA plays a primary role in limiting the production of extracellular proteases to prevent them from limiting the abundance of S. aureus virulence factors. Eliminating the production of all 10 extracellular proteases in the methicillin-resistant strain LAC has also been shown to enhance virulence in a murine sepsis model, and this has been attributed to the specific proteases Aur and ScpA. The importance of this work lies in our demonstration that the increased production of these same proteases largely accounts for the decreased virulence of sarA mutants in a murine model of post-traumatic osteomyelitis not only in LAC but also in the methicillin-susceptible human osteomyelitis isolate UAMS-1. This confirms that sarA-mediated repression of Aur and ScpA production plays a critical role in the posttranslational regulation of S. aureus virulence factors in diverse clinical isolates and diverse forms of S. aureus infection.


Asunto(s)
Metaloendopeptidasas , Osteomielitis , Infecciones Estafilocócicas , Animales , Ratones , Humanos , Staphylococcus aureus/metabolismo , Virulencia/genética , Modelos Animales de Enfermedad , Meticilina/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Proteínas Bacterianas/metabolismo , Péptido Hidrolasas/metabolismo , Biopelículas , Regulación Bacteriana de la Expresión Génica , Mamíferos/metabolismo
14.
Microbiol Spectr ; 12(1): e0314723, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38059629

RESUMEN

IMPORTANCE: Because biofilm formation is such a problematic feature of Staphylococcus aureus infections, much effort has been put into identifying biofilm inhibitors. However, the results observed with these compounds are often reported in isolation, and the methods used to assess biofilm formation vary between labs, making it impossible to assess relative efficacy and prioritize among these putative inhibitors for further study. The studies we report address this issue by directly comparing putative biofilm inhibitors using a consistent in vitro assay. This assay was previously shown to maximize biofilm formation, and the results observed with this assay have been proven to be relevant in vivo. Of the 19 compounds compared using this method, many had no impact on biofilm formation under these conditions. Indeed, only one proved effective at limiting biofilm formation without also inhibiting growth.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Antibacterianos/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Biopelículas , Proyectos de Investigación , Pruebas de Sensibilidad Microbiana
15.
Bone ; : 117181, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38960295

RESUMEN

Staphylococcus aureus osteomyelitis leads to extensive bone destruction. Osteoclasts are bone resorbing cells that are often increased in bone infected with S. aureus. The cytokine RANKL is essential for osteoclast formation under physiological conditions but in vitro evidence suggests that inflammatory cytokines may by-pass the requirement for RANKL. The goal of this study was to determine whether RANKL-dependent osteoclast formation is essential for the bone loss that occurs in a murine model of S. aureus osteomyelitis. To this end, humanized-RANKL mice were infected by direct inoculation of S. aureus into a unicortical defect in the femur. Mice were treated with vehicle or denosumab, a human monoclonal antibody that inhibits RANKL, both before and during a 14-day infection period. The severe cortical bone destruction caused by infection was completely prevented by denosumab administration even though the bacterial burden in the femur was not affected. Osteoclasts were abundant near the inoculation site in vehicle-treated mice but absent in denosumab-treated mice. In situ hybridization demonstrated that S. aureus infection potently stimulated RANKL expression in bone marrow stromal cells. The extensive reactive bone formation that occurs in this osteomyelitis model was also reduced by denosumab administration. Lastly, there was a notable lack of osteoblasts near the infection site suggesting that the normal coupling of bone formation to bone resorption was disrupted by S. aureus infection. These results demonstrate that RANKL-mediated osteoclast formation is required for the bone loss that occurs in S. aureus infection and suggest that disruption of the coupling of bone formation to bone resorption may also contribute to bone loss in this condition.

16.
Mol Microbiol ; 86(5): 1183-96, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23075270

RESUMEN

Mutation of staphylococcal accessory regulator (sarA) results in increased production of extracellular proteases in Staphylococcus aureus, which has been correlated with decreased biofilm formation and decreased accumulation of extracellular toxins. We used murine models of implant-associated biofilm infection and S. aureus bacteraemia (SAB) to compare virulence of USA300 strain LAC, its isogenic sarA mutant, and derivatives of each of these strains with mutations in all 10 of the genes encoding recognized extracellular proteases. The sarA mutant was attenuated in both models, and this was reversed by eliminating production of extracellular proteases. To examine the mechanistic basis, we identified proteins impacted by sarA in a protease-dependent manner. We identified 253 proteins where accumulation was reduced in the sarA mutant compared with the parent strain, and was restored in the sarA/protease mutant. Additionally, in SAB, the LAC protease mutant exhibited a hypervirulent phenotype by comparison with the isogenic parent strain, demonstrating that sarA also positively regulates production of virulence factors, some of which are subject to protease-mediated degradation. We propose a model in which attenuation of sarA mutants is defined by their inability to produce critical factors and simultaneously repress production of extracellular proteases that would otherwise limit accumulation of virulence factors.


Asunto(s)
Proteínas Bacterianas/farmacología , Regulación Bacteriana de la Expresión Génica , Péptido Hidrolasas/efectos de los fármacos , Staphylococcus aureus/patogenicidad , Factores de Virulencia/metabolismo , Animales , Animales no Consanguíneos , Antibacterianos/farmacología , Bacteriemia/microbiología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Infecciones Relacionadas con Catéteres/microbiología , Daptomicina/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Virulencia , Factores de Virulencia/genética
17.
PLoS Pathog ; 7(2): e1001287, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21347352

RESUMEN

Methicillin-resistant Staphylococcus aureus is estimated to cause more U.S. deaths annually than HIV/AIDS. The emergence of hypervirulent and multidrug-resistant strains has further amplified public health concern and accentuated the need for new classes of antibiotics. RNA degradation is a required cellular process that could be exploited for novel antimicrobial drug development. However, such discovery efforts have been hindered because components of the Gram-positive RNA turnover machinery are incompletely defined. In the current study we found that the essential S. aureus protein, RnpA, catalyzes rRNA and mRNA digestion in vitro. Exploiting this activity, high through-put and secondary screening assays identified a small molecule inhibitor of RnpA-mediated in vitro RNA degradation. This agent was shown to limit cellular mRNA degradation and exhibited antimicrobial activity against predominant methicillin-resistant S. aureus (MRSA) lineages circulating throughout the U.S., vancomycin intermediate susceptible S. aureus (VISA), vancomycin resistant S. aureus (VRSA) and other Gram-positive bacterial pathogens with high RnpA amino acid conservation. We also found that this RnpA-inhibitor ameliorates disease in a systemic mouse infection model and has antimicrobial activity against biofilm-associated S. aureus. Taken together, these findings indicate that RnpA, either alone, as a component of the RNase P holoenzyme, and/or as a member of a more elaborate complex, may play a role in S. aureus RNA degradation and provide proof of principle for RNA catabolism-based antimicrobial therapy.


Asunto(s)
Antiinfecciosos/farmacología , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Ribonucleasa P/antagonistas & inhibidores , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus , Animales , Antiinfecciosos/uso terapéutico , Femenino , Células Hep G2 , Humanos , Ratones , Modelos Biológicos , Ribonucleasa P/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/patología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Vancomicina/farmacología , Vancomicina/uso terapéutico , Virulencia/efectos de los fármacos , Virulencia/genética
18.
Virulence ; 14(1): 2175496, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36748843

RESUMEN

We previously demonstrated that MgrA, SarA, SarR, SarS, SarZ, and Rot bind at least three of the four promoters associated with genes encoding primary extracellular proteases in Staphylococcus aureus (Aur, ScpA, SspA/SspB, SplA-F). We also showed that mutation of sarA results in a greater increase in protease production, and decrease in biofilm formation, than mutation of the loci encoding any of these other proteins. However, these conclusions were based on in vitro studies. Thus, the goal of the experiments reported here was to determine the relative impact of the regulatory loci encoding these proteins in vivo. To this end, we compared the virulence of mgrA, sarA, sarR, sarS, sarZ, and rot mutants in a murine osteomyelitis model. Mutants were generated in the methicillin-resistant USA300 strain LAC and the methicillin-sensitive USA200 strain UAMS-1, which was isolated directly from the bone of an osteomyelitis patient during surgical debridement. Mutation of mgrA and rot limited virulence to a statistically significant extent in UAMS-1, but not in LAC, while the sarA mutant exhibited reduced virulence in both strains. The reduced virulence of the sarA mutant was correlated with reduced cytotoxicity for osteoblasts and osteoclasts, reduced biofilm formation, and reduced sensitivity to the antimicrobial peptide indolicidin, all of which were directly attributable to increased protease production in both LAC and UAMS-1. These results illustrate the importance of considering diverse clinical isolates when evaluating the impact of regulatory mutations on virulence and demonstrate the significance of SarA in limiting protease production in vivo in S. aureus.


Asunto(s)
Osteomielitis , Péptido Hidrolasas , Animales , Humanos , Ratones , Virulencia , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Staphylococcus aureus , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Endopeptidasas/genética , Regulación Bacteriana de la Expresión Génica , Biopelículas , Mutación
19.
Infect Immun ; 80(5): 1634-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22354028

RESUMEN

Recent studies suggest that extracellular DNA promotes biofilm formation in Staphylococcus aureus and, conversely, that extracellular nucleases limit the ability to form a biofilm. S. aureus produces at least two extracellular nucleases, and in the study described in this report, we examined the impact of each of these nucleases on biofilm formation under both in vitro and in vivo conditions. Our results demonstrate that both nucleases impact biofilm formation in the clinical isolate UAMS-1. Under certain in vitro conditions, this impact is negative, with mutation of either or both of the nuclease genes (nuc1 and nuc2) resulting in an enhanced capacity to form a biofilm. However, this effect was not apparent in vivo in a murine model of catheter-associated biofilm formation. Rather, mutation of either or both nuclease genes appeared to limit biofilm formation to a degree that could be correlated with increased susceptibility to daptomycin.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Desoxirribonucleasas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Staphylococcus aureus/enzimología , Staphylococcus aureus/fisiología , Animales , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Infecciones Relacionadas con Catéteres/microbiología , Daptomicina/administración & dosificación , Daptomicina/farmacología , Desoxirribonucleasas/genética , Relación Dosis-Respuesta a Droga , Ratones , Pruebas de Sensibilidad Microbiana , Mutación , Staphylococcus aureus/genética
20.
Antimicrob Agents Chemother ; 56(11): 5839-44, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22948866

RESUMEN

Using a rabbit model of postsurgical osteomyelitis, we demonstrate that incorporation of xylitol into polymethylmethacrylate (PMMA) bone cement enhances the elution of daptomycin under in vivo conditions. We also demonstrate that this can be correlated with an improved therapeutic outcome in the treatment of a chronic bone infection following surgical debridement.


Asunto(s)
Antibacterianos/farmacología , Daptomicina/farmacología , Preparaciones de Acción Retardada/farmacología , Osteomielitis/tratamiento farmacológico , Polimetil Metacrilato/química , Infecciones Estafilocócicas/tratamiento farmacológico , Xilitol/química , Animales , Cementos para Huesos/uso terapéutico , Enfermedad Crónica , Desbridamiento , Preparaciones de Acción Retardada/química , Modelos Animales de Enfermedad , Humanos , Masculino , Osteomielitis/microbiología , Osteomielitis/cirugía , Conejos , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/cirugía , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA