Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.088
Filtrar
1.
Biomaterials ; 313: 122774, 2025 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39208699

RESUMEN

Osteomyelitis (OM) is a progressive, inflammatory infection of bone caused predominately by Staphylococcus aureus. Herein, we engineered an antibiotic-eluting collagen-hydroxyapatite scaffold capable of eliminating infection and facilitating bone healing. An iterative freeze-drying and chemical crosslinking approach was leveraged to modify antibiotic release kinetics, resulting in a layered dual-release system whereby an initial rapid release of antibiotic to clear infection was followed by a sustained controlled release to prevent reoccurrence of infection. We observed that the presence of microbial collagenase accelerated antibiotic release from the crosslinked layer of the scaffold, indicating that the material is responsive to microbial activity. As exemplar drugs, vancomycin and gentamicin-eluting scaffolds were demonstrated to be bactericidal, and supported osteogenesis in vitro. In a pilot murine model of OM, vancomycin-eluting scaffolds were observed to reduce S. aureus infection within the tibia. Finally, in a rabbit model of chronic OM, gentamicin-eluting scaffolds both facilitated radial bone defect healing and eliminated S. aureus infection. These results show that antibiotic-eluting collagen-hydroxyapatite scaffolds are a one-stage therapy for OM, which when implanted into infected bone defects simultaneously eradicate infection and facilitate bone tissue healing.


Asunto(s)
Antibacterianos , Gentamicinas , Osteomielitis , Infecciones Estafilocócicas , Staphylococcus aureus , Andamios del Tejido , Animales , Andamios del Tejido/química , Antibacterianos/farmacología , Antibacterianos/química , Infecciones Estafilocócicas/tratamiento farmacológico , Osteomielitis/tratamiento farmacológico , Conejos , Staphylococcus aureus/efectos de los fármacos , Gentamicinas/farmacología , Gentamicinas/administración & dosificación , Gentamicinas/química , Gentamicinas/uso terapéutico , Ratones , Vancomicina/farmacología , Vancomicina/química , Vancomicina/administración & dosificación , Durapatita/química , Cinética , Cicatrización de Heridas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Colágeno/química , Femenino
2.
World J Microbiol Biotechnol ; 40(10): 322, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39283509

RESUMEN

Staphylococcus aureus can develop antibiotic resistance and evade immune responses, causing infections in different body sites. However, the metabolic changes underlying this process are poorly understood. A variant strain, C1V, was derived from the parental strain C1 by exposing it to increasing concentrations of vancomycin in vitro. C1V exhibited a vancomycin-intermediate phenotype and physiological changes compared to C1. It showed higher survival rates than C1 when phagocytosed by Raw264.7 cells. Metabolomics analysis identified significant metabolic differences pre- and post-induction (C1 + SC1 vs. C1V + SC1V: 201 metabolites) as well as pre- and post-phagocytosis (C1 vs. SC1: 50 metabolites; C1V vs. SC1V: 95 metabolites). The variant strain had distinct morphological characteristics, decreased adhesion ability, impaired virulence, and enhanced resistance to phagocytosis compared to the parental strain. Differential metabolites may contribute to S. aureus ' resistance to antibiotics and phagocytosis, offering insights into potential strategies for altering vancomycin nonsusceptibility and enhancing phagocyte killing by manipulating bacterial metabolism.


Asunto(s)
Antibacterianos , Metabolómica , Fagocitosis , Staphylococcus aureus , Vancomicina , Vancomicina/farmacología , Ratones , Animales , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Fagocitosis/efectos de los fármacos , Células RAW 264.7 , Antibacterianos/farmacología , Virulencia , Infecciones Estafilocócicas/microbiología , Pruebas de Sensibilidad Microbiana , Resistencia a la Vancomicina/genética , Metaboloma/efectos de los fármacos , Adhesión Bacteriana/efectos de los fármacos , Adaptación Fisiológica
3.
NPJ Biofilms Microbiomes ; 10(1): 86, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39284817

RESUMEN

Auranofin (AF), a former rheumatoid polyarthritis treatment, gained renewed interest for its use as an antimicrobial. AF is an inhibitor of thioredoxin reductase (TrxB), a thiol and protein repair enzyme, with an antibacterial activity against several bacteria including C. difficile, an enteropathogen causing post-antibiotic diarrhea. Several studies demonstrated the effect of AF on C. difficile physiology, but the crucial questions of resistance mechanisms and impact on microbiota remain unaddressed. We explored potential resistance mechanisms by studying the impact of TrxB multiplicity and by generating and characterizing adaptive mutations. We showed that if mutants inactivated for trxB genes have a lower MIC of AF, the number of TrxBs naturally present in clinical strains does not impact the MIC. All stable mutations isolated after AF long-term exposure were in the anti-sigma factor of σB and strongly affect physiology. Finally, we showed that AF has less impact on human gut microbiota than vancomycin.


Asunto(s)
Antibacterianos , Auranofina , Clostridioides difficile , Microbioma Gastrointestinal , Pruebas de Sensibilidad Microbiana , Humanos , Auranofina/farmacología , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/genética , Microbioma Gastrointestinal/efectos de los fármacos , Antibacterianos/farmacología , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Reductasa de Tiorredoxina-Disulfuro/genética , Mutación , Farmacorresistencia Bacteriana , Adaptación Fisiológica , Factor sigma/genética , Factor sigma/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Vancomicina/farmacología
4.
Sci Rep ; 14(1): 21269, 2024 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-39261496

RESUMEN

Staphylococcus aureus infections are hard to treat due to the emergence of antibiotic resistant strains, as well as their ability to form biofilms. The MazEF toxin-antitoxin system is thought play a role in bacterial biofilm phenotype as well as antibiotic resistance. In S. aureus, the physiologic function of the mazEF gene in the disease transition from acute to chronic infection is not well understood. In methicillin resistant S. aureus (MRSA), loss of mazF expression results in loss of resistance to first generation cephalosporins. mazF::tn displayed sensitivity while the isogenic wild type (WT) remained resistant. mazF::tn displayed significantly increased growth of biofilms on metal implants over 48 h compared to WT and the complemented transposon mutant. mazF::tn biofilms displayed significantly decreased antibiotic tolerance to vancomycin and cefazolin in comparison to WT and complement biofilms. Mice given mazF::tn in a sepsis model displayed less abscess burden and increased survival (100%) when treated with cefazolin compared to WT bacteremia treated with cefazolin (20%). mazF::tn periprosthetic joint infections displayed increased biofilm burden at acute time points and decreased biofilm burden at chronic time points. Our data suggests MazEF in MRSA is responsible for controlling growth of biofilms, antibiotic tolerance, and influence chronic infections in vivo.


Asunto(s)
Antibacterianos , Biopelículas , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/fisiología , Animales , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/tratamiento farmacológico , Antibacterianos/farmacología , Ratones , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Pruebas de Sensibilidad Microbiana , Modelos Animales de Enfermedad , Vancomicina/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Cefazolina/farmacología , Femenino
5.
Sci Rep ; 14(1): 21006, 2024 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251613

RESUMEN

The emission of glyphosate and antibiotic residues from human activities threatens the diversity and functioning of the microbial community. This study examines the impact of a glyphosate-based herbicide (GBH) and common antibiotics on Gram-negative bacteria within the ESKAPEE group (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp. and Escherichia coli). Ten strains, including type and multidrug-resistant strains for each species were analysed and eight antibiotics (cefotaxime, meropenem, aztreonam, ciprofloxacin, gentamicin, tigecycline, sulfamethoxazole-trimethoprim, and colistin) were combined with the GBH. While most combinations yielded additive or indifferent effects in 70 associations, antagonistic effects were observed with ciprofloxacin and gentamicin in five strains. GBH notably decreased the minimum inhibitory concentration of colistin in eight strains and displayed synergistic activity with meropenem against metallo-ß-lactamase (MBL)-producing strains. Investigation into the effect of GBH properties on outer membrane permeability involved exposing strains to a combination of this GBH and vancomycin. Results indicated that GBH rendered strains sensitive to vancomycin, which is typically ineffective against Gram-negative bacteria. Furthermore, we examined the impact of GBH in combination with three carbapenem agents on 14 strains exhibiting varying carbapenem-resistance mechanisms to assess its effect on carbapenemase activity. The GBH efficiently inhibited MBL activity, demonstrating similar effects to EDTA (ethylenediaminetetraacetic acid). Chelating effect of GBH may have multifaceted impacts on bacterial cells, potentially by increasing outer membrane permeability and inactivating metalloenzyme activity.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Glicina , Glifosato , Bacterias Gramnegativas , Herbicidas , Pruebas de Sensibilidad Microbiana , Glicina/análogos & derivados , Glicina/farmacología , Antibacterianos/farmacología , Herbicidas/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Acinetobacter baumannii/efectos de los fármacos , Klebsiella pneumoniae/efectos de los fármacos , Humanos , Escherichia coli/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Ciprofloxacina/farmacología , Enterococcus faecium/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Colistina/farmacología , Vancomicina/farmacología , Enterobacter/efectos de los fármacos , Sinergismo Farmacológico , Meropenem/farmacología , Fenotipo , Gentamicinas/farmacología
6.
Pol J Microbiol ; 73(3): 403-410, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39268956

RESUMEN

Vancomycin-resistant Enterococcus faecium (VRE) has been detected in Türkiye. Only limited information is available on its dissemination in the central regions of the country. This study describes the first epidemiological characterization of VRE clinical isolates detected in patients in a hospital in the province of Aksaray. In this one-year study conducted between 2021 and 2022, stool samples from intensive care unit patients were screened for VRE using the phenotypic E-test method, and the antibiotic sensitivity test was analyzed by using the VITEK® 2 system. A molecular assay for confirmation of species level was carried out by 16S rRNA gene-based sequencing and testing for antibiotic resistance (vanA or vanB) and virulence factor-encoding genes (esp, asa1, and hyl). Further, genotypic characterization was determined by macro-restriction fragment pattern analysis (MRFPA) of genomic DNA digested with SmaI restriction enzyme. Of the total 350 Enterococcus positive patients from different hospital intensive care units, 22 (6.3%) were positive for VRE using the phenotypic E-test method. All isolates showed resistance to ampicillin, ciprofloxacin, vancomycin, and teicoplanin and positive amplification for the vanA gene. However, none of the isolates was positive for the vanB gene. The most prevalent virulence gene was esp. The results indicate that the isolates are persistent in the hospital environment and subsequently transmitted to hospitalized patients, thus representing challenges to an outbreak and infection control. These study results would also help formulate more effective strategies to reduce the transmission and propagation of VRE contamination in various hospital settings.


Asunto(s)
Antibacterianos , Proteínas Bacterianas , Enterococcus faecium , Genotipo , Infecciones por Bacterias Grampositivas , Unidades de Cuidados Intensivos , Pruebas de Sensibilidad Microbiana , Enterococos Resistentes a la Vancomicina , Humanos , Enterococcus faecium/genética , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/aislamiento & purificación , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/aislamiento & purificación , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/epidemiología , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Factores de Virulencia/genética , Vancomicina/farmacología , Heces/microbiología , ARN Ribosómico 16S/genética , Fenotipo , Masculino , Femenino , Resistencia a la Vancomicina/genética , Persona de Mediana Edad
7.
PLoS Pathog ; 20(8): e1012422, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39207957

RESUMEN

Vancomycin has proven remarkably durable to resistance evolution by Staphylococcus aureus despite widespread treatment with vancomycin in the clinic. Only 16 cases of vancomycin-resistant S. aureus (VRSA) have been documented in the United States. It is thought that the failure of VRSA to spread is partly due to the fitness cost imposed by the vanA operon, which is the only known means of high-level resistance. Here, we show that the fitness cost of vanA-mediated resistance can be overcome through laboratory evolution of VRSA in the presence of vancomycin. Adaptation to vancomycin imposed a tradeoff such that fitness in the presence of vancomycin increased, while fitness in its absence decreased in evolved lineages. Comparing the genomes of vancomycin-exposed and vancomycin-unexposed lineages pinpointed the D-alanine:D-alanine ligase gene (ddl) as the target of loss-of-function mutations, which were associated with the observed fitness tradeoff. Vancomycin-exposed lineages exhibited vancomycin dependence and abnormal colony morphology in the absence of drug, which were associated with mutations in ddl. However, further evolution of vancomycin-exposed lineages in the absence of vancomycin enabled some evolved lineages to escape this fitness tradeoff. Many vancomycin-exposed lineages maintained resistance in the absence of vancomycin, unlike their ancestral VRSA strains. These results indicate that VRSA might be able to compensate for the fitness deficit associated with vanA-mediated resistance, which may pose a threat to the prolonged durability of vancomycin in the clinic. Our results also suggest vancomycin treatment should be immediately discontinued in patients after VRSA is identified to mitigate potential adaptations.


Asunto(s)
Antibacterianos , Infecciones Estafilocócicas , Staphylococcus aureus Resistente a Vancomicina , Vancomicina , Vancomicina/farmacología , Antibacterianos/farmacología , Humanos , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus Resistente a Vancomicina/genética , Resistencia a la Vancomicina/genética , Pruebas de Sensibilidad Microbiana , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Mutación
8.
PLoS One ; 19(8): e0309145, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39208074

RESUMEN

Infections are a major complication of open fractures and fracture fixation. In this study, an innovative bioactive medical device was used to experimentally treat MRSE-induced osteomyelitis in rabbit tibia. This paper investigates the clinical significance of inflammatory biomarkers (NLR, PLR, MLR and PMR), SII and IL-6 and assesses their role in the development of osteomyelitis. The main objective is to identify the utility of hematological reports derived from neutrophils, leukocytes, monocytes and platelets in the evolution of implant-related osteomyelitis and the estimation of treatment efficiency. In particular, this study compares the response of these inflammatory markers to different treatments in the presence or absence of bioactive materials and/or topical antibiotics over time. The analysis of the threads showed that NLR, PLR and SII had high values in the acute phase of the disease, so that after chronicization, they decrease. The animals treated with vancomycin nano-functionalized peptide-enriched silk fibroin-coated implants showed lower levels of inflammatory biomarkers compared to the other groups (empty implants and peptide-enriched silk fibroin-coated implants). NLR, PLR and SII, complemented by IL-6 can be used as fairly accurate biomarkers for the diagnosis of osteomyelitis.


Asunto(s)
Biomarcadores , Modelos Animales de Enfermedad , Interleucina-6 , Osteomielitis , Infecciones Estafilocócicas , Animales , Conejos , Osteomielitis/microbiología , Osteomielitis/tratamiento farmacológico , Osteomielitis/inmunología , Interleucina-6/sangre , Interleucina-6/metabolismo , Biomarcadores/sangre , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus epidermidis , Enfermedad Crónica , Antibacterianos/farmacología , Inflamación , Enfermedad Aguda , Vancomicina/farmacología , Relevancia Clínica
9.
ACS Nano ; 18(35): 24327-24349, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39169538

RESUMEN

A series of progress has been made in the field of antimicrobial use of nanozymes due to their superior stability and decreased susceptibility to drug resistance. However, catalytically generated reactive oxygen species (ROS) are insufficient for coping with multidrug-resistant organisms (MDROs) in complex wound environments due to their low targeting ability and insufficient catalytic activity. To address this problem, chemically stable copper-gallic acid-vancomycin (CuGA-VAN) nanoneedles were successfully constructed by a simple approach for targeting bacteria; these nanoneedles exhibit OXD-like and GSH-px-like dual enzyme activities to produce ROS and induce bacterial cuproptosis-like death, thereby eliminating MDRO infections. The results of in vitro experiments showed that the free carboxylic acid of GA could react with the free ammonia of teichoic acid in the methicillin-resistant Staphylococcus aureus (MRSA) cell wall skeleton. Thus, CuGA-VAN nanoneedles can rapidly "capture" MRSA in liquid environments, releasing ROS, VAN and Cu2+ on bacterial surfaces to break down the MRSA barrier, destroying the biofilm. In addition, CuGA-VAN effectively promoted wound repair cell proliferation and angiogenesis to facilitate wound healing while ensuring biosafety. According to transcriptome sequencing, highly internalized Cu2+ causes copper overload toxicity; downregulates genes related to the bacterial glyoxylate cycle, tricarboxylic acid cycle, and oxidative respiratory chain; and induces lipid peroxidation in the cytoplasm, leading to bacterial cuproptosis-like death. In this study, CuGA-VAN was cleverly designed to trigger a cascade reaction of targeting, drug release, ROS-catalyzed antibacterial activity and cuproptosis-like death. This provides an innovative idea for multidrug-resistant infections.


Asunto(s)
Antibacterianos , Cobre , Staphylococcus aureus Resistente a Meticilina , Especies Reactivas de Oxígeno , Antibacterianos/farmacología , Antibacterianos/química , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Cobre/química , Cobre/farmacología , Especies Reactivas de Oxígeno/metabolismo , Vancomicina/farmacología , Vancomicina/química , Pruebas de Sensibilidad Microbiana , Animales , Humanos , Biopelículas/efectos de los fármacos , Nanoestructuras/química , Ratones
10.
Mol Med ; 30(1): 130, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39182021

RESUMEN

BACKGROUND: Vascular calcification is a common vascular lesion associated with high morbidity and mortality from cardiovascular events. Antibiotics can disrupt the gut microbiota (GM) and have been shown to exacerbate or attenuate several human diseases. However, whether antibiotic-induced GM disruption affects vascular calcification remains unclear. METHODS: Antibiotic cocktail (ABX) treatment was utilized to test the potential effects of antibiotics on vascular calcification. The effects of antibiotics on GM and serum short-chain fatty acids (SCFAs) in vascular calcification mice were analyzed using 16 S rRNA gene sequencing and targeted metabolomics, respectively. Further, the effects of acetate, propionate and butyrate on vascular calcification were evaluated. Finally, the potential mechanism by which acetate inhibits osteogenic transformation of VSMCs was explored by proteomics. RESULTS: ABX and vancomycin exacerbated vascular calcification. 16 S rRNA gene sequencing and targeted metabolomics analyses showed that ABX and vancomycin treatments resulted in decreased abundance of Bacteroidetes in the fecal microbiota of the mice and decreased serum levels of SCFAs. In addition, supplementation with acetate was found to reduce calcium salt deposition in the aorta of mice and inhibit osteogenic transformation in VSMCs. Finally, using proteomics, we found that the inhibition of osteogenic transformation of VSMCs by acetate may be related to glutathione metabolism and ubiquitin-mediated proteolysis. After adding the glutathione inhibitor Buthionine sulfoximine (BSO) and the ubiquitination inhibitor MG132, we found that the inhibitory effect of acetate on VSMC osteogenic differentiation was weakened by the intervention of BSO, but MG132 had no effect. CONCLUSION: ABX exacerbates vascular calcification, possibly by depleting the abundance of Bacteroidetes and SCFAs in the intestine. Supplementation with acetate has the potential to alleviate vascular calcification, which may be an important target for future treatment of vascular calcification.


Asunto(s)
Acetatos , Antibacterianos , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Calcificación Vascular , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Calcificación Vascular/metabolismo , Calcificación Vascular/etiología , Calcificación Vascular/tratamiento farmacológico , Ratones , Ácidos Grasos Volátiles/metabolismo , Acetatos/farmacología , Antibacterianos/efectos adversos , Antibacterianos/farmacología , Masculino , Osteogénesis/efectos de los fármacos , ARN Ribosómico 16S/genética , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Vancomicina/efectos adversos , Vancomicina/farmacología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efectos de los fármacos
11.
PLoS Biol ; 22(8): e3002741, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39146240

RESUMEN

Clostridioides difficile is an important human pathogen, for which there are very limited treatment options, primarily the glycopeptide antibiotic vancomycin. In recent years, vancomycin resistance has emerged as a serious problem in several gram-positive pathogens, but high-level resistance has yet to be reported for C. difficile, although it is not known if this is due to constraints upon resistance evolution in this species. Here, we show that resistance to vancomycin can evolve rapidly under ramping selection but is accompanied by fitness costs and pleiotropic trade-offs, including sporulation defects that would be expected to severely impact transmission. We identified 2 distinct pathways to resistance, both of which are predicted to result in changes to the muropeptide terminal D-Ala-D-Ala that is the primary target of vancomycin. One of these pathways involves a previously uncharacterised D,D-carboxypeptidase, expression of which is controlled by a dedicated two-component signal transduction system. Our findings suggest that while C. difficile is capable of evolving high-level vancomycin resistance, this outcome may be limited clinically due to pleiotropic effects on key pathogenicity traits. Moreover, our data identify potential mutational routes to resistance that should be considered in genomic surveillance.


Asunto(s)
Antibacterianos , Clostridioides difficile , Resistencia a la Vancomicina , Vancomicina , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/genética , Clostridioides difficile/patogenicidad , Resistencia a la Vancomicina/genética , Vancomicina/farmacología , Antibacterianos/farmacología , Aptitud Genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Humanos , Transducción de Señal , Mutación , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/genética
12.
World J Microbiol Biotechnol ; 40(10): 297, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39126539

RESUMEN

Vancomycin is a clinically important glycopeptide antibiotic against Gram-positive pathogenic bacteria, especially methicillin-resistant Staphylococcus aureus. In the mutant strain of Amycolatopsis keratiniphila HCCB10007 Δeco-cds4-27, the production of ECO-0501 was disrupted, but enhanced vancomycin yield by 55% was observed compared with the original strain of A. keratiniphila HCCB10007. To gain insights into the mechanism of the enhanced production of vancomycin in the mutant strain, comparative metabolomics analyses were performed between the mutant strain and the original strain, A. keratiniphila HCCB10007 via GC-TOF-MS and UPLC-HRMS. The results of PCA and OPLS-DA revealed a significant distinction of the intracellular metabolites between the two strains during the fermentation process. 64 intracellular metabolites, which involved in amino acids, fatty acids and central carbon metabolism, were identified as differential metabolites. The high-yield mutant strain maintained high levels of glucose-1-phosphate and glucose-6-phosphate and they declined with the increases of vancomycin production. Particularly, a strong association of fatty acids accumulation as well as 3,5-dihydroxyphenylacetic acid and non-proteinogenic amino acid 3,5-dihydroxyphenylglycine (Dpg) with enhancement of vancomycin production was observed in the high-yield mutant strain, indicating that the consumption of fatty acid pools might be beneficial for giving rise to 3,5-dihydroxyphenylacetic acid and Dpg which further lead to improve vancomycin production. In addition, the lower levels of glyoxylic acid and lactic acid and the higher levels of sulfur amino acids might be beneficial for improving vancomycin production. These findings proposed more advanced elucidation of metabolomic characteristics in the high-yield strain for vancomycin production and could provide potential strategies to enhance the vancomycin production.


Asunto(s)
Amycolatopsis , Antibacterianos , Fermentación , Metabolómica , Vancomicina , Vancomicina/farmacología , Vancomicina/metabolismo , Metabolómica/métodos , Antibacterianos/metabolismo , Antibacterianos/farmacología , Amycolatopsis/metabolismo , Amycolatopsis/genética , Redes y Vías Metabólicas , Metaboloma , Mutación , Ácidos Grasos/metabolismo , Glioxilatos/metabolismo , Aminoácidos/metabolismo , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/metabolismo , Staphylococcus aureus Resistente a Meticilina/genética
13.
Gut Microbes ; 16(1): 2390133, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39132815

RESUMEN

Vancomycin (VAN) treatment in Clostridioides difficile infection (CDI) suffers from a relatively high rate of recurrence, with a variety of reasons behind this, including biofilm-induced recurrent infections. C. difficile can form monophyletic or symbiotic biofilms with other microbes in the gut, and these biofilms protect C. difficile from being killed by antibiotics. In this study, we analyzed the ecological relationship between Bacteroides thetaiotaomicron and C. difficile and their formation of symbiotic biofilm in the VAN environment. The production of symbiotic biofilm formed by C. difficile and B. thetaiotaomicron was higher than that of C. difficile and B. thetaiotaomicron alone in the VAN environment. In symbiotic biofilms, C. difficile was characterized by increased production of the toxin protein TcdA and TcdB, up-regulation of the expression levels of the virulence genes tcdA and tcdB, enhanced bacterial cell swimming motility and c-di-GMP content, and increased adhesion to Caco-2 cells. The scanning electron microscope (SEM) combined with confocal laser scanning microscopy (CLSM) results indicated that the symbiotic biofilm was elevated in thickness, dense, and had an increased amount of mixed bacteria, while the fluorescence in situ hybridization (FISH) probe and plate colony counting results further indicated that the symbiotic biofilm had a significant increase in the amount of C. difficile cells, and was able to better tolerate the killing of the simulated intestinal fluid. Taken together, C. difficile and B. thetaiotaomicron become collaborative in the VAN environment, and targeted deletion or attenuation of host gut B. thetaiotaomicron content may improve the actual efficacy of VAN in CDI treatment.


Asunto(s)
Antibacterianos , Proteínas Bacterianas , Bacteroides thetaiotaomicron , Biopelículas , Clostridioides difficile , Simbiosis , Vancomicina , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Clostridioides difficile/efectos de los fármacos , Clostridioides difficile/fisiología , Clostridioides difficile/genética , Humanos , Vancomicina/farmacología , Antibacterianos/farmacología , Células CACO-2 , Bacteroides thetaiotaomicron/efectos de los fármacos , Bacteroides thetaiotaomicron/metabolismo , Bacteroides thetaiotaomicron/fisiología , Bacteroides thetaiotaomicron/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/genética , Enterotoxinas/metabolismo , Enterotoxinas/genética , Adhesión Bacteriana/efectos de los fármacos
14.
Nat Commun ; 15(1): 6734, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112491

RESUMEN

Staphylococcus aureus is responsible for a substantial number of invasive infections globally each year. These infections are problematic because they are frequently recalcitrant to antibiotic treatment. Antibiotic tolerance, the ability of bacteria to persist despite normally lethal doses of antibiotics, contributes to antibiotic treatment failure in S. aureus infections. To understand how antibiotic tolerance is induced, S. aureus biofilms exposed to multiple anti-staphylococcal antibiotics are examined using both quantitative proteomics and transposon sequencing. These screens indicate that arginine metabolism is involved in antibiotic tolerance within a biofilm and support the hypothesis that depletion of arginine within S. aureus communities can induce antibiotic tolerance. Consistent with this hypothesis, inactivation of argH, the final gene in the arginine synthesis pathway, induces antibiotic tolerance. Arginine restriction induces antibiotic tolerance via inhibition of protein synthesis. In murine skin and bone infection models, an argH mutant has enhanced ability to survive antibiotic treatment with vancomycin, highlighting the relationship between arginine metabolism and antibiotic tolerance during S. aureus infection. Uncovering this link between arginine metabolism and antibiotic tolerance has the potential to open new therapeutic avenues targeting previously recalcitrant S. aureus infections.


Asunto(s)
Antibacterianos , Arginina , Biopelículas , Infecciones Estafilocócicas , Staphylococcus aureus , Arginina/metabolismo , Antibacterianos/farmacología , Animales , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Ratones , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Vancomicina/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Femenino , Farmacorresistencia Bacteriana/genética , Farmacorresistencia Bacteriana/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Hidrolasas/metabolismo , Hidrolasas/genética , Proteómica
15.
Int J Biol Macromol ; 278(Pt 3): 134940, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39173806

RESUMEN

The patterns of formation of chitosan nanoparticles doped with vancomycin and coatings based on them in carbonate solutions have been investigated for the first time in this study. Using a technique of radioactive indicators, it was found that at a CO2 pressure of 30 MPa, the yield of the nanoparticles was ∼85 %, and a maximum antibiotic encapsulation efficiency of ∼30 % was achieved. By spectrophotometric and high-resolution microscopy, it was found that the coating of stabilized xenopericardial tissue of bioprosthetic heart valve, based on chitosan nanoparticles doped with vancomycin with a zeta potential |ζ| ∼20 mV completely covers collagen fibers by depositing about 60 nm nanoparticles onto them under direct deposition from carbonic acid at a pressure of 30 MPa CO2. The coating preserves the mechanical strength characteristics of collagen tissue and completely suppresses the growth of S. aureus pathogenic biofilm. This is consistent with the observed increase in antibiotic release of 15 % when the medium was acidified. Histological study demonstrated that the structure of pericardial tissues was not significantly altered by the deposition nanoparticles from carbonic acid. It was found that the rate of biodegradation of polymers and vancomycin in the coating differs by half (16 weeks for the rat model). A significantly lower degradation rate of antibiotics (∼50 % of vancomycin total remaining mass and ∼25 % of chitosan) was associated with its reliable encapsulation into nanoparticles.


Asunto(s)
Dióxido de Carbono , Quitosano , Nanopartículas , Vancomicina , Quitosano/química , Vancomicina/química , Vancomicina/farmacología , Nanopartículas/química , Animales , Dióxido de Carbono/química , Ratas , Agua/química , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo , Antibacterianos/farmacología , Antibacterianos/química , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo
16.
Microb Pathog ; 195: 106886, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39182855

RESUMEN

Given the ability of Staphylococcus aureus to form biofilms and produce persister cells, making infections difficult to treat with antibiotics alone, there is a pressing need for an effective antibiotic adjuvant to address this public health threat. In this study, a series of quinone derivatives were evaluated for their antimicrobial and antibiofilm activities against methicillin-susceptible and methicillin-resistant S. aureus reference strains. Following analyses using broth microdilution, growth curve analysis, checkerboard assay, time-kill experiments, and confocal laser scanning microscopy, menadione was identified as a hit compound. Menadione exhibited a notable antibacterial profile (minimum inhibitory concentration, MIC = 4-16 µg/ml; minimum bactericidal concentration, MBC = 256 µg/ml) against planktonic S. aureus and its biofilms (minimum biofilm inhibitory concentration, MBIC50 = 0.0625-0.25 µg/ml). When combined with oxacillin, erythromycin, and vancomycin, menadione exhibited a synergistic or additive effect against planktonic cells and biofilms of two S. aureus reference strains and six clinical isolates, highlighting its potential as a suitable adjuvant for further development against S. aureus biofilm-associated infections.


Asunto(s)
Antibacterianos , Biopelículas , Sinergismo Farmacológico , Pruebas de Sensibilidad Microbiana , Naftoquinonas , Staphylococcus aureus , Vitamina K 3 , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Antibacterianos/farmacología , Vitamina K 3/farmacología , Staphylococcus aureus/efectos de los fármacos , Naftoquinonas/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Oxacilina/farmacología , Antraquinonas/farmacología , Eritromicina/farmacología , Vancomicina/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Humanos
17.
ACS Infect Dis ; 10(9): 3430-3439, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39185798

RESUMEN

Vancomycin-resistant enterococcus (VRE) is a major nosocomial pathogen that exhibits enhanced infectivity due to its robust virulence and biofilm-forming capabilities. In this study, 6-methoxyldihydrochelerythrine chloride (6-MDC) inhibited the growth of exponential-phase VRE and restored VRE's sensitivity to vancomycin. 6-MDC predominantly suppressed the de novo biosynthetic pathway of pyrimidine and purine in VRE by the RNA-Seq analysis, resulting in obstructed DNA synthesis, which subsequently weakened bacterial virulence and impeded intracellular survival. Furthermore, 6-MDC inhibited biofilm formation, eradicated established biofilms, reduced virulence, and enhanced the host immune response to prevent intracellular survival and replication of VRE. Finally, 6-MDC reduced the VRE load in peritoneal fluid and cells significantly in a murine peritoneal infection model. This paper provides insight into the potential antimicrobial target of benzophenanthridine alkaloids for the first time.


Asunto(s)
Antibacterianos , Benzofenantridinas , Biopelículas , Pruebas de Sensibilidad Microbiana , Enterococos Resistentes a la Vancomicina , Animales , Ratones , Antibacterianos/farmacología , Antibacterianos/química , Benzofenantridinas/farmacología , Benzofenantridinas/química , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Biopelículas/efectos de los fármacos , Virulencia/efectos de los fármacos , Vancomicina/farmacología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Femenino
18.
Int J Pharm ; 664: 124630, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39216651

RESUMEN

The rise of antibiotic resistant bacteria, such as methicillin-resistant Staphylococcus aureus (MRSA), requires novel approaches to combat infections. Medical devices like implants and wound dressings are frequently used in conjunction with antibiotics, motivating the development of antibacterial biomaterials capable of exhibiting combined antibacterial effects with conventional antibiotics. This study explores the synergistic antibacterial effects of combining antimicrobial peptide (AMP) functionalized hydrogel particles with conventional antibiotics, vancomycin (VCM) and oxacillin (OXA), against Staphylococcus aureus and MRSA. The AMP employed, RRPRPRPRPWWWW-NH2, has previously demonstrated broad-spectrum activity and enhanced stability when attached to hydrogel substrates. Here, checkerboard assays revealed additive and synergistic interactions between the free AMP and both VCM and OXA against Staphylococcus aureus and MRSA. Notably, the AMP-OXA combination displayed a significant synergistic effect against MRSA, with a 512-fold reduction in OXA's minimum inhibitory concentration (MIC) when combined with free AMP. The observed synergism against MRSA was retained upon covalent AMP immobilization onto the hydrogel particles; however, at a lower rate with a 64-fold reduction in OXA MIC. Despite this, the OXA-AMP hydrogel particle combinations retained considerable synergistic potential against MRSA, a strain resistant to OXA, highlighting the potential of AMP-functionalized materials for enhancing antibiotic efficacy. These findings underscore the importance of developing antimicrobial biomaterials for future medical devices to fight biomaterial-associated infections and reverse antimicrobial resistance.


Asunto(s)
Antibacterianos , Sinergismo Farmacológico , Hidrogeles , Staphylococcus aureus Resistente a Meticilina , Pruebas de Sensibilidad Microbiana , Oxacilina , Vancomicina , Vancomicina/farmacología , Vancomicina/administración & dosificación , Vancomicina/química , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/administración & dosificación , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Oxacilina/farmacología , Oxacilina/administración & dosificación , Hidrogeles/química , Péptidos Antimicrobianos/farmacología , Péptidos Antimicrobianos/química , Péptidos Antimicrobianos/administración & dosificación , Staphylococcus aureus/efectos de los fármacos
19.
J Appl Microbiol ; 135(9)2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39210508

RESUMEN

AIMS: In Tunisia, limited research has focused on characterizing clinical vancomycin-resistant Enterococcus faecium (VREfm). This study aimed to bridge this knowledge gap by molecular characterization of antimicrobial resistance, determining the genetic elements mediating vancomycin-resistance, and whole-genome sequencing of one representative VREfm isolate. METHODS AND RESULTS: Over 6 years (2011-2016), a total of eighty VREfm isolates responsible for infection or colonization were identified from hospitalized patients, with the incidence rate increasing from 2% in 2011 to 27% in 2016. All of these strains harbored the vanA gene. The screening for antimicrobial resistance genes revealed the predominance of ermB, tetM, and aac(6')-Ie-aph(2'')-Ia genes and 81.2% of strains harbored the Tn1545. Pulsed-field gel electrophoresis identified seven clusters, with two major clusters (belonging to ST117 and ST80) persisting throughout the study period. Seven Tn1546 types were detected, with type VI (truncated transposon) being the most prevalent (57.5%). Whole-genome sequencing revealed a 3 028 373 bp chromosome and five plasmids. Mobile genetic elements and a type I CRISPR-cas locus were identified. Notably, the vanA gene was carried by the classic Tn1546 transposon with ISL3 insertion on a rep17pRUM plasmid. CONCLUSION: A concerning trend in the prevalence of VREfm essentially attributed to CC17 persistence and to horizontal transfer of multiple genetic variants of truncated vanA-Tn1546.


Asunto(s)
Elementos Transponibles de ADN , Enterococcus faecium , Variación Genética , Infecciones por Bacterias Grampositivas , Neutropenia , Enterococos Resistentes a la Vancomicina , Secuenciación Completa del Genoma , Humanos , Enterococcus faecium/genética , Enterococcus faecium/aislamiento & purificación , Enterococcus faecium/efectos de los fármacos , Túnez , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/aislamiento & purificación , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/epidemiología , Elementos Transponibles de ADN/genética , Neutropenia/microbiología , Neutropenia/complicaciones , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Pruebas de Sensibilidad Microbiana , Electroforesis en Gel de Campo Pulsado , Resistencia a la Vancomicina/genética , Vancomicina/farmacología
20.
NPJ Biofilms Microbiomes ; 10(1): 77, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39209878

RESUMEN

Fracture-related infections (FRIs), particularly those caused by methicillin-resistant Staphylococcus aureus (MRSA), are challenging to treat. This study designed and evaluated a hydrogel loaded with a cocktail of bacteriophages and vancomycin (1.2 mg/mL). The co-delivery hydrogel showed 99.72% reduction in MRSA biofilm in vitro. The hydrogel released 54% of phages and 82% of vancomycin within 72 h and maintained activity for eight days, in vivo the co-delivery hydrogel with systemic antibiotic significantly reduced bacterial load by 0.99 log10 CFU compared to controls, with active phages detected in tissues at euthanasia (2 × 103 PFU/mL). No phage resistance was detected in the phage treatment groups, and serum neutralization resulted in only a 20% reduction in phage count. In this work, we show that a phage-antibiotic co-delivery system via CMC hydrogel is a promising adjunct to systemic antibiotic therapy for MRSA-induced FRI, highlighting its potential for localized, sustained delivery and improved treatment outcomes.


Asunto(s)
Antibacterianos , Biopelículas , Hidrogeles , Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Vancomicina , Vancomicina/administración & dosificación , Vancomicina/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Antibacterianos/administración & dosificación , Antibacterianos/farmacología , Animales , Hidrogeles/química , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/terapia , Biopelículas/efectos de los fármacos , Bacteriófagos/fisiología , Fracturas Óseas/terapia , Terapia de Fagos/métodos , Ratones , Sistemas de Liberación de Medicamentos , Humanos , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA