Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Microbiol Spectr ; 10(5): e0299022, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36121287

RESUMEN

Bacterial efflux pumps in the resistance-nodulation-cell division (RND) family of Gram-negative bacteria contribute significantly to the development of antimicrobial resistance by many pathogens. In this study, we selected the MtrD transporter protein of Neisseria gonorrhoeae as it is the sole RND pump possessed by this strictly human pathogen and can export multiple antimicrobials, including antibiotics, bile salts, detergents, dyes, and antimicrobial peptides. Using knowledge from our previously published structures of MtrD in the presence or absence of bound antibiotics as a model and the known ability of MtrCDE to export cationic antimicrobial peptides, we hypothesized that cationic peptides could be accommodated within MtrD binding sites. Furthermore, we thought that MtrD-bound peptides lacking antibacterial action could sensitize bacteria to an antibiotic normally exported by the MtrCDE efflux pump or other similar RND-type pumps possessed by different Gram-negative bacteria. We now report the identification of a novel nonantimicrobial cyclic cationic antimicrobial peptide, which we termed CASP (cationic antibiotic-sensitizing peptide). By single-particle cryo-electron microscopy, we found that CASP binds within the periplasmic cleft region of MtrD using overlapping and distinct amino acid contact sites that interact with another cyclic peptide (colistin) or a linear human cationic antimicrobial peptide derived from human LL-37. While CASP could not sensitize Neisseria gonorrhoeae to an antibiotic (novobiocin) that is a substrate for RND pumps, it could do so against multiple Gram-negative, rod-shaped bacteria. We propose that CASP (or future derivatives) could serve as an adjuvant for the antibiotic treatment of certain Gram-negative infections previously thwarted by RND transporters. IMPORTANCE RND efflux pumps can export numerous antimicrobials that enter Gram-negative bacteria, and their action can reduce the efficacy of antibiotics and provide decreased susceptibility to various host antimicrobials. Here, we identified a cationic antibiotic-sensitizing peptide (CASP) that binds within the periplasmic cleft of an RND transporter protein (MtrD) produced by Neisseria gonorrhoeae. Surprisingly, CASP was able to render rod-shaped Gram-negative bacteria, but not gonococci, susceptible to an antibiotic that is a substrate for the gonococcal MtrCDE efflux pump. CASP (or its future derivatives) could be used as an adjuvant to treat infections for which RND efflux contributes to multidrug resistance.


Asunto(s)
Antiinfecciosos , Colistina , Humanos , Colistina/metabolismo , Novobiocina/metabolismo , Microscopía por Crioelectrón , Detergentes/metabolismo , Detergentes/farmacología , Proteínas Bacterianas/genética , Neisseria gonorrhoeae , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Antiinfecciosos/farmacología , División Celular , Aminoácidos/metabolismo , Ácidos y Sales Biliares/metabolismo , Colorantes/metabolismo , Colorantes/farmacología , Farmacorresistencia Bacteriana Múltiple
2.
Sci Rep ; 11(1): 21676, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34737361

RESUMEN

Development of an effective therapy to overcome colistin resistance in Klebsiella pneumoniae, a common pathogen causing catheter-related biofilm infections in vascular catheters, has become a serious therapeutic challenge that must be addressed urgently. Although colistin and EDTA have successful roles for eradicating biofilms, no in vitro and in vivo studies have investigated their efficacy in catheter-related biofilm infections of colistin-resistant K. pneumoniae. In this study, colistin resistance was significantly reversed in both planktonic and mature biofilms of colistin-resistant K. pneumoniae by a combination of colistin (0.25-1 µg/ml) with EDTA (12 mg/ml). This novel colistin-EDTA combination was also demonstrated to have potent efficacy in eradicating colistin-resistant K. pneumoniae catheter-related biofilm infections, and eliminating the risk of recurrence in vivo. Furthermore, this study revealed significant therapeutic efficacy of colistin-EDTA combination in reducing bacterial load in internal organs, lowering serum creatinine, and protecting treated mice from mortality. Altered in vivo expression of different virulence genes indicate bacterial adaptive responses to survive in hostile environments under different treatments. According to these data discovered in this study, a novel colistin-EDTA combination provides favorable efficacy and safety for successful eradication of colistin-resistant K. pneumonia catheter-related biofilm infections.


Asunto(s)
Colistina/uso terapéutico , Ácido Edético/uso terapéutico , Klebsiella pneumoniae/efectos de los fármacos , Animales , Antibacterianos/uso terapéutico , Biopelículas/efectos de los fármacos , Infecciones Relacionadas con Catéteres/tratamiento farmacológico , Catéteres/microbiología , Colistina/metabolismo , Combinación de Medicamentos , Farmacorresistencia Bacteriana/efectos de los fármacos , Femenino , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/patogenicidad , Ratones , Ratones Endogámicos C57BL , Pruebas de Sensibilidad Microbiana , Virulencia
3.
J Anim Sci ; 98(2)2020 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-31943046

RESUMEN

Organic acids (OA) and phytogenic compounds have been used in pig feeding as alternatives to antibiotic growth promoters. However, few studies have evaluated the systemic effect of the combination of these additives. The aim of this study was to assess the impact of an organic acid-based feed additive (OAFA), containing a blend of OA and cinnamaldehyde, on the tissue integrity of bacterially challenged piglets. Thirty weaned piglets 21 d old were used in a 19-d trial. Pigs received a standard diet during the first 7 d and afterward were allotted to five treatments. Dietary treatments were: Control (basal diet), Escherichia coli (basal diet and challenge with E. coli), colistin (basal diet + 200 mg colistin/kg feed + challenge with E. coli), OAFA1 (basal diet + 1 kg OAFA/ton feed + challenge with E. coli), and OAFA2 (basal diet + 2 kg OAFA/ton feed + challenge with E. coli). Seven days after the beginning of the treatment, the animals were challenged with an enterotoxic strain of E. coli (K88) for pigs. Five days after the challenge, all animals were euthanized for tissue sampling for histological and oxidative stress (intestine and liver) analysis. The reduced glutathione (GSH), ferric-reducing ability potential (FRAP), and free-radical scavenging ability (ABTS) assays were used to evaluate the intestinal antioxidant defense. Lipid peroxidation and superoxide anion production were evaluated through the levels of thiobarbituric acid-reactive substances (TBARS) and nitroblue tetrazolium (NBT) reduction assay, respectively. Animals fed the OAFA (1 and 2) diets had a decrease (P < 0.05) on histological changes in the intestine, liver, mesenteric lymph nodes, and spleen. Greater villus height (VH) and a higher ratio of VH to crypt depth (CD) were observed in animals of the OAFA2 group compared with the control and E. coli groups. The colistin and OAFA groups decreased (P < 0.05) the number of inflammatory cells in intestinal lamina propria. OAFA2 group increased (P < 0.05) intestinal cell proliferation. Colistin and OAFA2 supplementation induced a decrease (P < 0.05) in the levels of TBARS in both the intestine and liver compared with the E. coli group. In addition, an increase (P < 0.05) in GSH and FRAP ileal levels was observed in the OAFA2 group compared with E. coli group. These results show that the supplementation with OAFA in the diet of weaned piglets, especially at a dose of 2 kg/ton (OAFA2) protected tissues against enterotoxigenic Escherichia coli (ETEC) damage.


Asunto(s)
Acroleína/análogos & derivados , Ácidos Carboxílicos/administración & dosificación , Escherichia coli Enterotoxigénica/fisiología , Homeostasis/efectos de los fármacos , Porcinos/fisiología , Acroleína/administración & dosificación , Animales , Antibacterianos/farmacología , Colistina/metabolismo , Dieta/veterinaria , Ingestión de Alimentos , Infecciones por Escherichia coli/tratamiento farmacológico , Femenino , Mucosa Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Hígado/efectos de los fármacos , Masculino , Estrés Oxidativo , Porcinos/crecimiento & desarrollo , Destete
4.
Mol Pharm ; 16(7): 3199-3207, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31125239

RESUMEN

Polymer masked-unmasked protein therapy (PUMPT) uses conjugation of a biodegradable polymer, such as dextrin, hyaluronic acid, or poly(l-glutamic acid), to mask a protein or peptide's activity; subsequent locally triggered degradation of the polymer at the target site regenerates bioactivity in a controllable fashion. Although the concept of PUMPT is well established, the relationship between protein unmasking and reinstatement of bioactivity is unclear. Here, we used dextrin-colistin conjugates to study the relationship between the molecular structure (degree of unmasking) and biological activity. Size exclusion chromatography was employed to collect fractions of differentially degraded conjugates and ultraperformance liquid chromatography-mass spectrometry (UPLC-MS) employed to characterize the corresponding structures. Antimicrobial activity was studied using a minimum inhibitory concentration (MIC) assay and confocal laser scanning microscopy of LIVE/DEAD-stained biofilms with COMSTAT analysis. In vitro toxicity of the degraded conjugate was assessed using an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. UPLC-MS revealed that the fully "unmasked" dextrin-colistin conjugate composed of colistin bound to at least one linker, whereas larger species were composed of colistin with varying lengths of glucose units attached. Increasing the degree of dextrin modification by succinoylation typically led to a greater number of linkers bound to colistin. Greater antimicrobial and antibiofilm activity were observed for the fully "unmasked" conjugate compared to the partially degraded species (MIC = 0.25 and 2-8 µg/mL, respectively), whereas dextrin conjugation reduced colistin's in vitro toxicity toward kidney cells, even after complete unmasking. This study highlights the importance of defining the structure-antimicrobial activity relationship for novel antibiotic derivatives and demonstrates the suitability of LC-MS to aid the design of biodegradable polymer-antibiotic conjugates.


Asunto(s)
Amilasas/metabolismo , Colistina/química , Colistina/metabolismo , Dextrinas/química , Dextrinas/metabolismo , Composición de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/métodos , Antibacterianos/química , Antibacterianos/metabolismo , Biopelículas/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromatografía en Gel , Escherichia coli/efectos de los fármacos , Humanos , Túbulos Renales Proximales/citología , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Microscopía Confocal , Estructura Molecular
5.
Nat Microbiol ; 2: 17028, 2017 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-28263303

RESUMEN

The increasing use of polymyxins1 in addition to the dissemination of plasmid-borne colistin resistance threatens to cause a serious breach in our last line of defence against multidrug-resistant Gram-negative pathogens, and heralds the emergence of truly pan-resistant infections. Colistin resistance often arises through covalent modification of lipid A with cationic residues such as phosphoethanolamine-as is mediated by Mcr-1 (ref. 2)-which reduce the affinity of polymyxins for lipopolysaccharide3. Thus, new strategies are needed to address the rapidly diminishing number of treatment options for Gram-negative infections4. The difficulty in eradicating Gram-negative bacteria is largely due to their highly impermeable outer membrane, which serves as a barrier to many otherwise effective antibiotics5. Here, we describe an unconventional screening platform designed to enrich for non-lethal, outer-membrane-active compounds with potential as adjuvants for conventional antibiotics. This approach identified the antiprotozoal drug pentamidine6 as an effective perturbant of the Gram-negative outer membrane through its interaction with lipopolysaccharide. Pentamidine displayed synergy with antibiotics typically restricted to Gram-positive bacteria, yielding effective drug combinations with activity against a wide range of Gram-negative pathogens in vitro, and against systemic Acinetobacter baumannii infections in mice. Notably, the adjuvant activity of pentamidine persisted in polymyxin-resistant bacteria in vitro and in vivo. Overall, pentamidine and its structural analogues represent unexploited molecules for the treatment of Gram-negative infections, particularly those having acquired polymyxin resistance determinants.


Asunto(s)
Antibacterianos/metabolismo , Colistina/metabolismo , Farmacorresistencia Bacteriana , Sinergismo Farmacológico , Bacterias Gramnegativas/efectos de los fármacos , Pentamidina/metabolismo , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos
6.
Int J Antimicrob Agents ; 48(6): 680-689, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27842757

RESUMEN

The host stress hormone norepinephrine (NE), also called noradrenaline, is reported to augment bacterial growth and pathogenicity, but few studies have focused on the effect of NE on the activity of antimicrobials. The aim of this study was to clarify whether NE affects antimicrobial activity against multidrug-resistant Acinetobacter baumannii (MDR-AB). Time-kill studies of tigecycline (TIG) and colistin (COL) against MDR-AB as well as assays for factors contributing to antibiotic resistance were performed using MDR-AB clinical strains both in the presence and absence of 10 µM NE. In addition, expression of three efflux pump genes (adeB, adeJ and adeG) in the presence and absence of NE was analysed by quantitative reverse transcription PCR. Viable bacterial cell counts in TIG-supplemented medium containing NE were significantly increased compared with those in medium without NE. In contrast, NE had little influence on viable bacterial cell counts in the presence of COL. NE-supplemented medium resulted in an ca. 2 log increase in growth and in bacterial cell numbers adhering on polyurethane, silicone and polyvinylchloride surfaces. Amounts of biofilm in the presence of NE were ca. 3-fold higher than without NE. Expression of the adeG gene was upregulated 4-6-fold in the presence of NE. In conclusion, NE augmented factors contributing to antibiotic resistance and markedly reduced the in vitro antibacterial activity of TIG against MDR-AB. These findings suggest that NE treatment may contribute to the failure of TIG therapy in patients with MDR-AB infections.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Agonistas alfa-Adrenérgicos/metabolismo , Antibacterianos/farmacología , Antagonismo de Drogas , Minociclina/análogos & derivados , Norepinefrina/metabolismo , Acinetobacter baumannii/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Adhesión Bacteriana , Carga Bacteriana , Biopelículas/crecimiento & desarrollo , Colistina/metabolismo , Medios de Cultivo/química , Femenino , Perfilación de la Expresión Génica , Genes Bacterianos , Humanos , Masculino , Viabilidad Microbiana/efectos de los fármacos , Persona de Mediana Edad , Minociclina/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Tigeciclina , Factores de Tiempo
7.
Antimicrob Agents Chemother ; 58(8): 4399-403, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24841267

RESUMEN

Colistin is a key drug for the treatment of infections caused by extensively drug-resistant strains of Enterobacteriaceae producing carbapenemases. However, the emergence of colistin resistance is being increasingly reported, especially among Klebsiella pneumoniae strains producing KPC-type carbapenemases (KPC-KP). In this work, we investigated colistin-susceptible (KPB-1) and colistin-resistant (KPB-2) sequential isolates obtained from a patient with a KPC-KP infection before and after low-dosage colistin treatment, respectively. By using a next-generation sequencing approach and comparative genomic analysis of the two isolates, we detected in KPB-2 a nonsynonymous nucleotide substitution in the gene encoding the PmrB sensor kinase, resulting in a leucine-to-arginine substitution at amino acid position 82. Compared with KPB-1, KPB-2 exhibited upregulated transcription of pmrA and of pmrK, which is part of the pmrHFIJKLM operon responsible for modification of the colistin lipopolysaccharide target. Complementation with wild-type pmrB in KPB-2 restored colistin susceptibility and reduced the transcription of pmrA and pmrK to basal levels, while expression of PmrB(L82R) in KPB-1 did not alter colistin susceptibility or upregulate pmrA and pmrK expression, confirming the dominance of wild-type PmrB versus the PmrB(L82R) mutant. The present results indicated that PmrB mutations mediating colistin resistance may be selected during low-dosage colistin treatment. The colistin-resistant phenotype of KPB-2 was stable for up to 50 generations in the absence of selective pressure and was not associated with a significant fitness cost in a competition experiment.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/genética , Colistina/farmacología , Regulación Bacteriana de la Expresión Génica , Klebsiella pneumoniae/genética , Factores de Transcripción/genética , Resistencia betalactámica/genética , beta-Lactamasas/genética , Sustitución de Aminoácidos , Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Células Clonales , Colistina/metabolismo , Cálculo de Dosificación de Drogas , Humanos , Infecciones por Klebsiella/tratamiento farmacológico , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/enzimología , Klebsiella pneumoniae/aislamiento & purificación , Lipopolisacáridos/metabolismo , Pruebas de Sensibilidad Microbiana , Mutación , Operón , Factores de Transcripción/metabolismo , beta-Lactamasas/metabolismo
8.
Expert Rev Anti Infect Ther ; 12(5): 531-3, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24666285

RESUMEN

The initial use of polymyxins, polymyxin B and colistin (administered as a pro-drug colistin methanesulfonate sodium [CMS]), mostly relied on old pharmacokinetic (PK) studies that lacked appropriate methodology. In recent years, many PK studies in both animals and humans have provided more consistent evidence supporting better use of these invaluable antibiotics. However, translating preclinical data to clinical practice is not always an easy task and some may experience difficulties on how recent knowledge on polymyxins can be applied into the patients' care. Although many questions are still unresolved, there are consistent data able to improve clinical practice when prescribing initial and maintenance doses of both polymyxin B and CMS. Considering the importance of optimal use of polymyxins, this editorial discusses recent PK findings and how to take advantage of them at the bedside to improve the treatment of patient with extensively-drug-resistant Gram-negative bacterial infections.


Asunto(s)
Antibacterianos/farmacocinética , Colistina/análogos & derivados , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Polimixina B/farmacocinética , Antibacterianos/farmacología , Área Bajo la Curva , Colistina/metabolismo , Colistina/farmacocinética , Colistina/farmacología , Esquema de Medicación , Cálculo de Dosificación de Drogas , Farmacorresistencia Bacteriana Múltiple , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/crecimiento & desarrollo , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Polimixina B/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA