Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nat Commun ; 14(1): 8198, 2023 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-38081813

RESUMEN

Antibiotic resistance of bacteria is considered one of the most alarming developments in modern medicine. While varied pathways for bacteria acquiring antibiotic resistance have been identified, there still are open questions concerning the mechanisms underlying resistance. Here, we show that alpha phenol-soluble modulins (PSMαs), functional bacterial amyloids secreted by Staphylococcus aureus, catalyze hydrolysis of ß-lactams, a prominent class of antibiotic compounds. Specifically, we show that PSMα2 and, particularly, PSMα3 catalyze hydrolysis of the amide-like bond of the four membered ß-lactam ring of nitrocefin, an antibiotic ß-lactam surrogate. Examination of the catalytic activities of several PSMα3 variants allowed mapping of the active sites on the amyloid fibrils' surface, specifically underscoring the key roles of the cross-α fibril organization, and the combined electrostatic and nucleophilic functions of the lysine arrays. Molecular dynamics simulations further illuminate the structural features of ß-lactam association upon the fibril surface. Complementary experimental data underscore the generality of the functional amyloid-mediated catalytic phenomenon, demonstrating hydrolysis of clinically employed ß-lactams by PSMα3 fibrils, and illustrating antibiotic degradation in actual S. aureus biofilms and live bacteria environments. Overall, this study unveils functional amyloids as catalytic agents inducing degradation of ß-lactam antibiotics, underlying possible antibiotic resistance mechanisms associated with bacterial biofilms.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Antibióticos Betalactámicos , Antibacterianos/farmacología , Antibacterianos/metabolismo , Monobactamas/metabolismo , beta-Lactamas/farmacología , beta-Lactamas/metabolismo , Infecciones Estafilocócicas/microbiología , Bacterias
2.
Molecules ; 28(20)2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37894491

RESUMEN

Staphylococcus aureus is a common human pathogen. Methicillin-resistant Staphylococcus aureus (MRSA) infections pose significant and challenging therapeutic difficulties. MRSA often acquires the non-native gene PBP2a, which results in reduced susceptibility to ß-lactam antibiotics, thus conferring resistance. PBP2a has a lower affinity for methicillin, allowing bacteria to maintain peptidoglycan biosynthesis, a core component of the bacterial cell wall. Consequently, even in the presence of methicillin or other antibiotics, bacteria can develop resistance. Due to genes responsible for resistance, S. aureus becomes MRSA. The fundamental premise of this resistance mechanism is well-understood. Given the therapeutic concerns posed by resistant microorganisms, there is a legitimate demand for novel antibiotics. This review primarily focuses on PBP2a scaffolds and the various screening approaches used to identify PBP2a inhibitors. The following classes of compounds and their biological activities are discussed: Penicillin, Cephalosporins, Pyrazole-Benzimidazole-based derivatives, Oxadiazole-containing derivatives, non-ß-lactam allosteric inhibitors, 4-(3H)-Quinazolinones, Pyrrolylated chalcone, Bis-2-Oxoazetidinyl macrocycles (ß-lactam antibiotics with 1,3-Bridges), Macrocycle-embedded ß-lactams as novel inhibitors, Pyridine-Coupled Pyrimidinones, novel Naphthalimide corbelled aminothiazoximes, non-covalent inhibitors, Investigational-ß-lactam antibiotics, Carbapenem, novel Benzoxazole derivatives, Pyrazolylpyridine analogues, and other miscellaneous classes of scaffolds for PBP2a. Additionally, we discuss the penicillin-binding protein, a crucial target in the MRSA cell wall. Various aspects of PBP2a, bacterial cell walls, peptidoglycans, different crystal structures of PBP2a, synthetic routes for PBP2a inhibitors, and future perspectives on MRSA inhibitors are also explored.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Humanos , Proteínas de Unión a las Penicilinas/química , Staphylococcus aureus Resistente a Meticilina/metabolismo , Meticilina/metabolismo , Meticilina/farmacología , Staphylococcus aureus/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Monobactamas/metabolismo , Proteínas Bacterianas/química , Pruebas de Sensibilidad Microbiana
3.
PLoS Pathog ; 19(7): e1011536, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37486930

RESUMEN

Central metabolic pathways control virulence and antibiotic resistance, and constitute potential targets for antibacterial drugs. In Staphylococcus aureus the role of the pentose phosphate pathway (PPP) remains largely unexplored. Mutation of the 6-phosphogluconolactonase gene pgl, which encodes the only non-essential enzyme in the oxidative phase of the PPP, significantly increased MRSA resistance to ß-lactam antibiotics, particularly in chemically defined media with physiologically-relevant concentrations of glucose, and reduced oxacillin (OX)-induced lysis. Expression of the methicillin-resistance penicillin binding protein 2a and peptidoglycan architecture were unaffected. Carbon tracing and metabolomics revealed extensive metabolic reprogramming in the pgl mutant including increased flux to glycolysis, the TCA cycle, and several cell envelope precursors, which was consistent with increased ß-lactam resistance. Morphologically, pgl mutant cells were smaller than wild-type with a thicker cell wall and ruffled surface when grown in OX. The pgl mutation reduced resistance to Congo Red, sulfamethoxazole and oxidative stress, and increased resistance to targocil, fosfomycin and vancomycin. Levels of lipoteichoic acids (LTAs) were significantly reduced in pgl, which may limit cell lysis, while the surface charge of pgl cells was significantly more positive. A vraG mutation in pgl reversed the increased OX resistance phenotype, and partially restored wild-type surface charge, but not LTA levels. Mutations in vraF or graRS from the VraFG/GraRS complex that regulates DltABCD-mediated d-alanylation of teichoic acids (which in turn controls ß-lactam resistance and surface charge), also restored wild-type OX susceptibility. Collectively these data show that reduced levels of LTAs and OX-induced lysis combined with a VraFG/GraRS-dependent increase in cell surface positive charge are accompanied by significantly increased OX resistance in an MRSA pgl mutant.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/metabolismo , Vía de Pentosa Fosfato/genética , Antibacterianos/farmacología , Antibacterianos/metabolismo , Oxacilina/farmacología , Pared Celular/metabolismo , Monobactamas/metabolismo , Resistencia betalactámica/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Pruebas de Sensibilidad Microbiana
4.
Front Cell Infect Microbiol ; 13: 1139796, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37234778

RESUMEN

Introduction: Biofilm formation is the major pathogenicity of Staphylococcus epidermidis (S. epidermidis), which enhances bacterial resistance to antibiotics. Isookanin has potential inhibitory activity on biofilm. Method: The inhibiting mechanisms of isookanin against biofilm formation through surface hydrophobicity assay, exopolysaccharides, eDNA, gene expression analysis, microscopic visualization, and molecular docking were explored. Additionally, the combination of isookanin and ß-lactam antibiotics were evaluated by the broth micro-checkerboard assay. Results: The results showed that isookanin could decrease the biofilm formation of S. epidermidis by ≥85% at 250 µg/mL. The exopolysaccharides, eDNA and surface hydrophobicity were reduced after treatment with isookanin. Microscopic visualization analysis showed that there were fewer bacteria on the surface of the microscopic coverslip and the bacterial cell membrane was damaged after treatment with isookanin. The down-regulation of icaB and up-regulation of icaR were observed after treatment with isookanin. Additionally, the RNAIII gene was significantly up-regulated (p < 0.0001) at the mRNA level. Molecular docking showed that isookanin could bind to biofilm-related proteins. This indicated that isookanin can affect biofilm formation at the initial attachment phase and the aggregation phase. The FICI index showed that the combination of isookanin and ß-lactam antibiotics were synergistic and could reduce doses of antibiotics by inhibiting biofilm formation. Discussion: This study improved the antibiotic susceptibility of S. epidermidis through inhibition of the biofilm formation, and provided a guidance for the treatment of antibiotic resistance caused by biofilm.


Asunto(s)
Antibacterianos , Staphylococcus epidermidis , Staphylococcus epidermidis/genética , Antibacterianos/farmacología , Antibacterianos/metabolismo , Regulación hacia Abajo , Simulación del Acoplamiento Molecular , Biopelículas , Monobactamas/metabolismo , Pruebas de Sensibilidad Microbiana
5.
J Bacteriol ; 205(3): e0047622, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36840595

RESUMEN

Antibiotic tolerance, the ability of bacteria to sustain viability in the presence of typically bactericidal antibiotics for extended time periods, is an understudied contributor to treatment failure. The Gram-negative pathogen Vibrio cholerae, the causative agent of cholera, becomes highly tolerant to ß-lactam antibiotics (penicillin and related compounds) in a process requiring the two-component system VxrAB. VxrAB is induced by exposure to cell wall damaging conditions, which results in the differential regulation of >100 genes. While the effectors of VxrAB are relatively well known, VxrAB environment-sensing and activation mechanisms remain a mystery. Here, we used transposon mutagenesis to screen for mutants that spontaneously upregulate VxrAB signaling. This screen was answered by genes known to be required for proper cell envelope homeostasis, validating the approach. Unexpectedly, we also uncovered a new connection between central carbon metabolism and antibiotic tolerance in Vibrio cholerae. Inactivation of pgi (vc0374, coding for glucose-6-phosphate isomerase) resulted in an intracellular accumulation of glucose-6-phosphate and fructose-6-phosphate, concomitant with a marked cell envelope defect, resulting in VxrAB induction. Deletion of pgi also increased sensitivity to ß-lactams and conferred a growth defect on salt-free LB, phenotypes that could be suppressed by deleting sugar uptake systems and by supplementing cell wall precursors in the growth medium. Our data suggest an important connection between central metabolism and cell envelope integrity and highlight a potential new target for developing novel antimicrobial agents. IMPORTANCE Antibiotic tolerance (the ability to survive exposure to antibiotics) is a stepping stone toward antibiotic resistance (the ability to grow in the presence of antibiotics), an increasingly common cause of antibiotic treatment failure. The mechanisms promoting tolerance are poorly understood. Here, we identified central carbon metabolism as a key contributor to antibiotic tolerance and resistance. A strain with a mutation in a sugar utilization pathway accumulates metabolites that likely shut down the synthesis of cell wall precursors, which weakens the cell wall and thus increases susceptibility to cell wall-active drugs. Our results illuminate the connection between central carbon metabolism and cell wall homeostasis in V. cholerae and suggest that interfering with metabolism may be a fruitful future strategy for the development of antibiotic adjuvants.


Asunto(s)
Vibrio cholerae , Vibrio cholerae/metabolismo , Mutación , Penicilinas/farmacología , Antibacterianos/farmacología , Antibacterianos/metabolismo , Monobactamas/metabolismo , beta-Lactamas/farmacología
6.
Microbiol Spectr ; 11(1): e0297822, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36602386

RESUMEN

The frequent resistance associated with ß-lactam antibiotics and the high frequency of mutations in ß-lactamases constitute a major clinical challenge that can no longer be ignored. Andrographolide (AP), a natural active compound, has been shown to restore susceptibility to ß-lactam antibiotics. Fluorescence quenching and molecular simulation showed that AP quenched the intrinsic fluorescence of ß-lactamase BlaZ and stably bound to the residues in the catalytic cavity of BlaZ. Of note, AP was found to reduce the stability of the cell wall (CW) in methicillin-resistant Staphylococcus aureus (MRSA), and in combination with penicillin G (PEN), it significantly induced CW roughness and dispersion and even caused its disintegration, while the same concentration of PEN did not. In addition, transcriptome sequencing revealed that AP induced a significant stress response and increased peptidoglycan (PG) synthesis but disrupted its cross-linking, and it repressed the expression of critical genes such as mecA, blaZ, and sarA. We also validated these findings by quantitative reverse transcription-PCR (qRT-PCR). Association analysis using the GEO database showed that the alterations caused by AP were similar to those caused by mutations in the sarA gene. In summary, AP was able to restore the susceptibility of MRSA to ß-lactam antibiotics, mainly by inhibiting the ß-lactamase BlaZ, by downregulating the expression of critical resistance genes such as mecA and blaZ, and by disrupting CW homeostasis. In addition, restoration of susceptibility to antibiotics could be achieved by inhibiting the global regulator SarA, providing an effective solution to alleviate the problem of bacterial resistance. IMPORTANCE Increasingly, alternatives to antibiotics are being used to mitigate the rapid onset and development of bacterial resistance, and the combination of natural compounds with traditional antibiotics has become an effective therapeutic strategy. Therefore, we attempted to discover more mechanisms to restore susceptibility and effective dosing strategies. Andrographolide (AP), as a natural active ingredient, can mediate recovery of susceptibility of MRSA to ß-lactam antibiotics. AP bound stably to the ß-lactamase BlaZ and impaired its hydrolytic activity. Notably, AP was able to downregulate the expression of critical resistance genes such as mecA, blaZ, and sarA. Meanwhile, it disrupted the CW cross-linking and homeostasis, while the same concentration of penicillin could not. The multiple inhibitory effect of AP resensitizes intrinsically resistant bacteria to ß-lactam antibiotics, effectively prolonging the use cycle of these antibiotics and providing an effective solution to reduce the dosage of antibiotics and providing a theoretical reference for the prevention and control of MRSA.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Antibacterianos/metabolismo , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Penicilinas , Monobactamas/metabolismo , Monobactamas/farmacología
7.
mBio ; 14(1): e0247822, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36507833

RESUMEN

The purine-derived signaling molecules c-di-AMP and (p)ppGpp control mecA/PBP2a-mediated ß-lactam resistance in methicillin-resistant Staphylococcus aureus (MRSA) raise the possibility that purine availability can control antibiotic susceptibility. Consistent with this, exogenous guanosine and xanthosine, which are fluxed through the GTP branch of purine biosynthesis, were shown to significantly reduce MRSA ß-lactam resistance. In contrast, adenosine (fluxed to ATP) significantly increased oxacillin resistance, whereas inosine (which can be fluxed to ATP and GTP via hypoxanthine) only marginally increased oxacillin susceptibility. Furthermore, mutations that interfere with de novo purine synthesis (pur operon), transport (NupG, PbuG, PbuX) and the salvage pathway (DeoD2, Hpt) increased ß-lactam resistance in MRSA strain JE2. Increased resistance of a nupG mutant was not significantly reversed by guanosine, indicating that NupG is required for guanosine transport, which is required to reduce ß-lactam resistance. Suppressor mutants resistant to oxacillin/guanosine combinations contained several purine salvage pathway mutations, including nupG and hpt. Guanosine significantly increased cell size and reduced levels of c-di-AMP, while inactivation of GdpP, the c-di-AMP phosphodiesterase negated the impact of guanosine on ß-lactam susceptibility. PBP2a expression was unaffected in nupG or deoD2 mutants, suggesting that guanosine-induced ß-lactam susceptibility may result from dysfunctional c-di-AMP-dependent osmoregulation. These data reveal the therapeutic potential of purine nucleosides, as ß-lactam adjuvants that interfere with the normal activation of c-di-AMP are required for high-level ß-lactam resistance in MRSA. IMPORTANCE The clinical burden of infections caused by antimicrobial resistant (AMR) pathogens is a leading threat to public health. Maintaining the effectiveness of existing antimicrobial drugs or finding ways to reintroduce drugs to which resistance is widespread is an important part of efforts to address the AMR crisis. Predominantly, the safest and most effective class of antibiotics are the ß-lactams, which are no longer effective against methicillin-resistant Staphylococcus aureus (MRSA). Here, we report that the purine nucleosides guanosine and xanthosine have potent activity as adjuvants that can resensitize MRSA to oxacillin and other ß-lactam antibiotics. Mechanistically, exposure of MRSA to these nucleosides significantly reduced the levels of the cyclic dinucleotide c-di-AMP, which is required for ß-lactam resistance. Drugs derived from nucleotides are widely used in the treatment of cancer and viral infections highlighting the clinical potential of using purine nucleosides to restore or enhance the therapeutic effectiveness of ß-lactams against MRSA and potentially other AMR pathogens.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Nucleósidos de Purina/metabolismo , Nucleósidos de Purina/farmacología , Proteínas Bacterianas/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Oxacilina/farmacología , beta-Lactamas/farmacología , Monobactamas/metabolismo , Monobactamas/farmacología , Guanosina/metabolismo , Guanosina/farmacología , Adenosina Trifosfato/metabolismo , Guanosina Trifosfato/metabolismo , Pruebas de Sensibilidad Microbiana , Proteínas de Unión a las Penicilinas/genética , Proteínas de Unión a las Penicilinas/metabolismo , Resistencia betalactámica/genética
8.
Transplant Cell Ther ; 29(1): 54.e1-54.e6, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36208727

RESUMEN

The mechanism(s) of acquisition of extended-spectrum cephalosporin-resistant Enterobacteriaceae (ESCRE) on inpatient hospital units dedicated to hematopoietic stem cell transplantation (HSCT) is unclear. The objectives of this study were to determine whether ESCRE organisms are transmitted among patients housed on a HSCT unit, clarify the mechanisms involved, and determine whether routine surveillance for ESCRE carriage and contact isolation for ESCRE carriers is beneficial. The study was conducted on a 30-bed inpatient unit dedicated to the care of patients with hematologic malignancies and HSCT recipients. To investigate whether ESCRE organisms may be transmitted vertically to subsequent room occupants, presumably through contamination of room surfaces, we (1) cultured 6 high touch areas in 10 rooms before and 9 rooms after terminal cleaning that had been occupied by patients with ESCRE carriage, (2) determined the in vitro survivals of our most common clinical ESCRE species, and (3) followed the subsequent room occupants of 54 consecutive ESCRE colonized patients for the development of inpatient acquired ESCRE carriage. To investigate whether ESCRE organisms are transmitted horizontally among inpatients we (1) sequenced 60 available ESCRE Escherichia coli isolates obtained from unit inpatients and searched for identities using complete-genome multisequence locus typing (cgMLST) and (2) retrospectively tabulated the cumulative rates of acquired ESCRE carriage in 356 patients admitted for a first HSCT before (200 patients) or after (156 patients) institution of universal ESCRE stool surveillance and contact isolation for carriers. No ESCRE organisms were cultured from patient rooms before or after terminal cleaning. In vitro, few, if any, ESCRE organisms survived longer than 2 hours. Nine of the subsequent occupants of a room in which a patient with ESCRE carriage had resided were detected with ESCRE carriage, only 2 of whom carried the same species as that of the prior occupant. DNA sequencing and cgMLST determination of the 60 E. coli isolates showed 53 cgMLST strains. Seven of the 53 strains were shared by 2 patients. After institution of universal ESCRE surveillance/isolation there was a significant decline in acquired ESCRE carriage among HSCT recipients. We conclude that vertical transmission of ESCRE organisms through room contamination appears to be uncommon on modern HSCT units. Conversely, our results are consistent with the horizontal spread of ESCRE organisms, probably mediated by intermediate vectors such as personnel or shared equipment. Further studies are needed to better define the magnitude of and risk factors for ESCRE horizontal transfers and the benefits of ESCRE surveillance/isolation.


Asunto(s)
Infección Hospitalaria , Trasplante de Células Madre Hematopoyéticas , Humanos , Enterobacteriaceae/genética , Enterobacteriaceae/metabolismo , Cefalosporinas/uso terapéutico , Cefalosporinas/metabolismo , Escherichia coli/metabolismo , Estudios Retrospectivos , Infección Hospitalaria/prevención & control , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Monobactamas/metabolismo , Trasplante de Células Madre Hematopoyéticas/efectos adversos
9.
Commun Biol ; 5(1): 1059, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36198902

RESUMEN

Gram-negative porins are the main entry for small hydrophilic molecules. We studied translocation of structurally related cephalosporins, ceftazidime (CAZ), cefotaxime (CTX) and cefepime (FEP). CAZ is highly active on E. coli producing OmpF (Outer membrane protein F) but less efficient on cells expressing OmpC (Outer membrane protein C), whereas FEP and CTX kill bacteria regardless of the porin expressed. This matches with the different capacity of CAZ and FEP to accumulate into bacterial cells as quantified by LC-MS/MS (Liquid Chromatography Tandem Mass Spectrometry). Furthermore, porin reconstitution into planar lipid bilayer and zero current assays suggest permeation of ≈1,000 molecules of CAZ per sec and per channel through OmpF versus ≈500 through OmpC. Here, the instant killing is directly correlated to internal drug concentration. We propose that the net negative charge of CAZ represents a key advantage for permeation through OmpF porins that are less cation-selective than OmpC. These data could explain the decreased susceptibility to some cephalosporins of enterobacteria that exclusively express OmpC porins.


Asunto(s)
Cefalosporinas , Enterobacteriaceae , Cefepima/metabolismo , Cefotaxima/metabolismo , Ceftazidima , Cefalosporinas/farmacología , Cromatografía Liquida , Escherichia coli/metabolismo , Membrana Dobles de Lípidos/metabolismo , Monobactamas/metabolismo , Porinas/química , Porinas/metabolismo , Espectrometría de Masas en Tándem
10.
Antimicrob Agents Chemother ; 66(10): e0087922, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36102663

RESUMEN

Xeruborbactam (formerly QPX7728) is a cyclic boronate inhibitor of numerous serine and metallo-beta-lactamases. At concentrations generally higher than those required for beta-lactamase inhibition, xeruborbactam has direct antibacterial activity against some Gram-negative bacteria, with MIC50/MIC90 values of 16/32 µg/mL and 16/64 µg/mL against carbapenem-resistant Enterobacterales and carbapenem-resistant Acinetobacter baumannii, respectively (the MIC50/MIC90 values against Pseudomonas aeruginosa are >64 µg/mL). In Klebsiella pneumoniae, inactivation of OmpK36 alone or in combination with OmpK35 resulted in 2- to 4-fold increases in the xeruborbactam MIC. In A. baumannii and P. aeruginosa, AdeIJK and MexAB-OprM, respectively, affected xeruborbactam's antibacterial potency (the MICs were 4- to 16-fold higher in efflux-proficient strains). In Escherichia coli and K. pneumoniae, the 50% inhibitory concentrations (IC50s) of xeruborbactam's binding to penicillin-binding proteins (PBPs) PBP1a/PBP1b, PBP2, and PBP3 were in the 40 to 70 µM range; in A. baumannii, xeruborbactam bound to PBP1a, PBP2, and PBP3 with IC50s of 1.4 µM, 23 µM, and 140 µM, respectively. Treating K. pneumoniae and P. aeruginosa with xeruborbactam at 1× and 2× MIC resulted in changes of cellular morphology similar to those observed with meropenem; the morphological changes observed after treatment of A. baumannii were consistent with inhibition of multiple PBPs but were unique to xeruborbactam compared to the results for control beta-lactams. No single-step xeruborbactam resistance mutants were obtained after selection at 4× MIC of xeruborbactam using wild-type strains of E. coli, K. pneumoniae, and A. baumannii; mutations selected at 2× MIC in K. pneumoniae did not affect antibiotic potentiation by xeruborbactam through beta-lactamase inhibition. Consistent with inhibition of PBPs, xeruborbactam enhanced the potencies of beta-lactam antibiotics even against strains that lacked beta-lactamase. In a large panel of KPC-producing clinical isolates, the MIC90 values of meropenem tested with xeruborbactam (8 µg/mL) were at least 4-fold lower than those in combination with vaborbactam at 64 µg/mL, the concentration of vaborbactam that is associated with complete inhibition of KPC. The additional enhancement of the potency of beta-lactam antibiotics beyond beta-lactamase inhibition may contribute to the potentiation of beta-lactam antibiotics by xeruborbactam.


Asunto(s)
Antibacterianos , Escherichia coli , Meropenem/farmacología , Meropenem/metabolismo , Proteínas de Unión a las Penicilinas/genética , Escherichia coli/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , beta-Lactamasas/genética , beta-Lactamas/farmacología , Pruebas de Sensibilidad Microbiana , Klebsiella pneumoniae/genética , Carbapenémicos/farmacología , Carbapenémicos/metabolismo , Monobactamas/metabolismo , Pseudomonas aeruginosa/metabolismo , Serina/metabolismo
11.
Antimicrob Agents Chemother ; 66(7): e0059022, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35762796

RESUMEN

Tebipenem pivoxil hydrobromide (TBP-PI-HBr) is an oral carbapenem prodrug being developed for the treatment of serious bacterial infections. The active moiety, tebipenem, has broad-spectrum activity against common Enterobacterales pathogens, including extended-spectrum-ß-lactamase (ESBL)-producing multidrug-resistant strains. This study evaluated the intrapulmonary pharmacokinetics (PK) and epithelial lining fluid (ELF) and alveolar macrophage (AM) concentrations of tebipenem relative to plasma levels in nonsmoking, healthy adult subjects. Thirty subjects received oral TBP-PI-HBr at 600 mg every 8 h for five doses. Serial blood samples were collected following the last dose. Each subject underwent one standardized bronchoscopy with bronchoalveolar lavage (BAL) 1, 2, 4, 6, or 8 h after the fifth dose of TBP-PI-HBr. The tebipenem area under the concentration-time curve for the 8-h dosing interval (AUC0-8) values in plasma, ELF, and AMs were calculated using the mean concentration at each BAL sampling time. Ratios of AUC0-8 values for total ELF and AMs to those for unbound plasma were determined, using a plasma protein binding value of 42%. Mean values ± standard deviations (SD) of tebipenem maximum (Cmax) and minimum (Cmin) total plasma concentrations were 11.37 ± 3.87 mg/L and 0.043 ± 0.039 mg/L, respectively. Peak tebipenem concentrations in plasma, ELF, and AMs occurred at 1 h and then decreased over 8 h. Ratios of tebipenem AUC0-8 values for ELF and AMs to those for unbound plasma were 0.191 and 0.047, respectively. Four (13.3%) subjects experienced adverse events (diarrhea, fatigue, papule, and coronavirus disease 2019 [COVID-19]); all resolved, and none were severe or serious. Tebipenem is distributed into the lungs of healthy adults, which supports the further evaluation of TBP-PI-HBr for the treatment of lower respiratory tract bacterial infections caused by susceptible pathogens. (This study has been registered at ClinicalTrials.gov under identifier NCT04710407.).


Asunto(s)
Antibacterianos , COVID-19 , Administración Oral , Adulto , Antibacterianos/farmacocinética , Líquido del Lavado Bronquioalveolar , Carbapenémicos/metabolismo , Humanos , Pulmón/metabolismo , Monobactamas/metabolismo
12.
Biomolecules ; 11(7)2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34356681

RESUMEN

Novel antimicrobial strategies are urgently required because of the rising threat of multi drug resistant bacterial strains and the infections caused by them. Among the available target structures, the so-called penicillin binding proteins are of particular interest, owing to their good accessibility in the periplasmic space, and the lack of homologous proteins in humans, reducing the risk of side effects of potential drugs. In this report, we focus on the interaction of the innovative ß-lactam antibiotic AIC499 with penicillin binding protein 3 (PBP3) from Escherichia coli and Pseudomonas aeruginosa. This recently developed monobactam displays broad antimicrobial activity, against Gram-negative strains, and improved resistance to most classes of ß-lactamases. By analyzing crystal structures of the respective complexes, we were able to explore the binding mode of AIC499 to its target proteins. In addition, the apo structures determined for PBP3, from P. aeruginosa and the catalytic transpeptidase domain of the E. coli orthologue, provide new insights into the dynamics of these proteins and the impact of drug binding.


Asunto(s)
Monobactamas/metabolismo , Proteínas de Unión a las Penicilinas/química , Proteínas de Unión a las Penicilinas/metabolismo , Cristalografía por Rayos X , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Monobactamas/química , Proteínas de Unión a las Penicilinas/genética , Conformación Proteica , Pseudomonas aeruginosa
13.
Artículo en Inglés | MEDLINE | ID: mdl-29555629

RESUMEN

The conjugation of siderophores to antimicrobial molecules is an attractive strategy to overcome the low outer membrane permeability of Gram-negative bacteria. In this Trojan horse approach, the transport of drug conjugates is redirected via TonB-dependent receptors (TBDR), which are involved in the uptake of essential nutrients, including iron. Previous reports have demonstrated the involvement of the TBDRs PiuA and PirA from Pseudomonas aeruginosa and their orthologues in Acinetobacter baumannii in the uptake of siderophore-beta-lactam drug conjugates. By in silico screening, we further identified a PiuA orthologue, termed PiuD, present in clinical isolates, including strain LESB58. The piuD gene in LESB58 is located at the same genetic locus as piuA in strain PAO1. PiuD has a similar crystal structure as PiuA and is involved in the transport of the siderophore-drug conjugates BAL30072, MC-1, and cefiderocol in strain LESB58. To screen for additional siderophore-drug uptake systems, we overexpressed 28 of the 34 TBDRs of strain PAO1 and identified PfuA, OptE, OptJ, and the pyochelin receptor FptA as novel TBDRs conferring increased susceptibility to siderophore-drug conjugates. The existence of a TBDR repertoire in P. aeruginosa able to transport siderophore-drug molecules potentially decreases the likelihood of resistance emergence during therapy.


Asunto(s)
Antibacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas Bacterianas/genética , Transporte Biológico/efectos de los fármacos , Proteínas Portadoras/genética , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cefalosporinas/metabolismo , Proteínas de la Membrana/genética , Monobactamas/metabolismo , Pseudomonas aeruginosa/genética , Tiazoles/metabolismo , beta-Lactamas/metabolismo , Cefiderocol
14.
Bioorg Med Chem Lett ; 28(4): 748-755, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29336873

RESUMEN

Metallo-ß-lactamases (MBLs), such as New Delhi metallo-ß-lactamase (NDM-1) have spread world-wide and present a serious threat. Expression of MBLs confers resistance in Gram-negative bacteria to all classes of ß-lactam antibiotics, with the exception of monobactams, which are intrinsically stable to MBLs. However, existing first generation monobactam drugs like aztreonam have limited clinical utility against MBL-expressing strains because they are impacted by serine ß-lactamases (SBLs), which are often co-expressed in clinical isolates. Here, we optimized novel monobactams for stability against SBLs, which led to the identification of LYS228 (compound 31). LYS228 is potent in the presence of all classes of ß-lactamases and shows potent activity against carbapenem-resistant isolates of Enterobacteriaceae (CRE).


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Enterobacteriaceae Resistentes a los Carbapenémicos/efectos de los fármacos , Monobactamas/farmacología , Resistencia betalactámica/efectos de los fármacos , beta-Lactamasas/metabolismo , Animales , Antibacterianos/efectos adversos , Antibacterianos/química , Antibacterianos/metabolismo , Aztreonam/farmacología , Células CHO , Cricetulus , Estabilidad de Medicamentos , Escherichia coli/efectos de los fármacos , Femenino , Humanos , Meropenem , Ratones , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Monobactamas/efectos adversos , Monobactamas/química , Monobactamas/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Receptores de GABA-A/metabolismo , Convulsiones/inducido químicamente , Relación Estructura-Actividad , Tienamicinas/farmacología
15.
Antimicrob Agents Chemother ; 59(9): 5159-64, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26055384

RESUMEN

Three Klebsiella pneumoniae clinical isolates demonstrating carbapenem resistance were recovered from different patients hospitalized at two medical centers in São Paulo, Brazil. Resistance to all ß-lactams, quinolones, and some aminoglycosides was observed for these isolates that were susceptible to polymyxin B. Carbapenem hydrolysis, which was inhibited by clavulanic acid, was observed for all K. pneumoniae isolates that belonged to the same pulsed-field gel electrophoresis (PFGE) type and a novel sequence type (ST), ST1781 (clonal complex 442 [CC442]). A 10-kb nonconjugative incompatibility group Q (IncQ) plasmid, denominated p60136, was transferred to Escherichia coli strain TOP10 cells by electroporation. The full sequencing of p60136 showed that it was composed of a mobilization system, ISKpn23, the phosphotransferase aph3A-VI, and a 941-bp open reading frame (ORF) that codified a 313-amino acid protein. This ORF was named bla BKC-1. Brazilian Klebsiella carbapenemase-1 (BKC-1) showed a pI of 6.0 and possessed the highest identity (63%) with a ß-lactamase of Sinorhizobium meliloti, an environmental bacterium. Hydrolysis studies demonstrated that purified BKC-1 not only hydrolyzed carbapenems but also penicillins, cephalosporins, and monobactams. However, the carbapenems were less efficiently hydrolyzed due to their very low kcat values (0.0016 to 0.031 s(-1)). In fact, oxacillin was the best substrate for BKC-1 (kcat /Km , 53,522.6 mM(-1) s(-1)). Here, we report a new class A carbapenemase, confirming the diversity and rapid evolution of ß-lactamases in K. pneumoniae clinical isolates.


Asunto(s)
Proteínas Bacterianas/metabolismo , Klebsiella pneumoniae/enzimología , beta-Lactamasas/metabolismo , Antibacterianos/metabolismo , Antibacterianos/farmacología , Brasil , Carbapenémicos/metabolismo , Carbapenémicos/farmacología , Cefalosporinas/metabolismo , Cefalosporinas/farmacología , Farmacorresistencia Bacteriana , Electroforesis en Gel de Campo Pulsado , Pruebas de Sensibilidad Microbiana , Monobactamas/metabolismo , Monobactamas/farmacología , Penicilinas/metabolismo , Penicilinas/farmacología , Sinorhizobium meliloti/efectos de los fármacos , Sinorhizobium meliloti/metabolismo
16.
Int J Antimicrob Agents ; 38(2): 157-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21596528

RESUMEN

Burkholderia pseudomallei is an intrinsically antibiotic-resistant Category B priority pathogen and the aetiological agent of melioidosis. Treatment of B. pseudomallei infection is biphasic and lengthy in order to combat the acute and chronic phases of the disease. Acute-phase treatment preferably involves an intravenous cephalosporin (ceftazidime) or a carbapenem (imipenem or meropenem). In this study, the anti-B. pseudomallei efficacy of a new monosulfactam, BAL30072, was tested against laboratory strains 1026b and 1710b and several isogenic mutant derivatives as well as a collection of clinical and environmental B. pseudomallei strains from Thailand. More than 93% of the isolates had minimal inhibitory concentrations (MICs) in the range 0.004-0.016 µg/mL. For the laboratory strain 1026b, the MIC of BAL30072 was 0.008 µg/mL, comparable with the MICs of 1.5 µg/mL for ceftazidime, 0.5 µg/mL for imipenem and 1 µg/mL for meropenem. Time-kill curves revealed that BAL30072 was rapidly bactericidal, killing >99% of bacteria in 2 h. BAL30072 activity was not significantly affected by efflux, it was only a marginal substrate of PenA ß-lactamase, and activity was independent of malleobactin production and transport and the ability to transport pyochelin. In summary, BAL30072 has superior in vitro activity against B. pseudomallei compared with ceftazidime, meropenem or imipenem and it is rapidly bactericidal.


Asunto(s)
Antibacterianos/farmacología , Burkholderia pseudomallei/efectos de los fármacos , Monobactamas/farmacología , Tiazoles/farmacología , Antibacterianos/metabolismo , Transporte Biológico , Burkholderia pseudomallei/aislamiento & purificación , Microbiología Ambiental , Humanos , Melioidosis/microbiología , Pruebas de Sensibilidad Microbiana , Viabilidad Microbiana/efectos de los fármacos , Monobactamas/metabolismo , Tailandia , Tiazoles/metabolismo , Factores de Tiempo , beta-Lactamasas/metabolismo
17.
Pathol Biol (Paris) ; 56(7-8): 435-8, 2008.
Artículo en Francés | MEDLINE | ID: mdl-19019568

RESUMEN

Breakpoints harmonization concerning six European committees is being conducted within the European Committee for Antimicrobial Susceptibility Testing for a few years and is now finalized for beta-lactam antibiotics. This article describes the impact of breakpoint modifications on percentage of susceptibility of P. aeruginosa to beta-lactams in comparison with previous French National Committee breakpoints (CA-SFM). This harmonization leads to the need for new recommendations about diameter breakpoints and for updating breakpoints in antibiotic susceptibility testing automated devices. Moreover, it points out the importance of MICs and quantitative diameter data in order to follow the evolution of antibiotic susceptibility.


Asunto(s)
Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana/normas , Pseudomonas aeruginosa/efectos de los fármacos , Resistencia betalactámica , beta-Lactamas/farmacología , Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Carbapenémicos/metabolismo , Carbapenémicos/farmacología , Resistencia a las Cefalosporinas , Cefalosporinas/metabolismo , Cefalosporinas/farmacología , Relación Dosis-Respuesta a Droga , Europa (Continente)/epidemiología , Pruebas de Sensibilidad Microbiana/instrumentación , Monobactamas/metabolismo , Monobactamas/farmacología , Resistencia a las Penicilinas , Penicilinas/metabolismo , Penicilinas/farmacología , Pseudomonas aeruginosa/enzimología , beta-Lactamasas/metabolismo , beta-Lactamas/metabolismo
18.
J Med Microbiol ; 57(Pt 8): 974-979, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18628498

RESUMEN

Limited beta-lactams show antipseudomonal activity. The rapid spread of IMP-type metallo-beta-lactamases (MBLs), which have a broad spectrum of substrates and a poor susceptibility to clinically available inhibitors, further restricts beta-lactam use. In the present study, we evaluated the potency of IMP-10 MBL in hydrolysing antipseudomonal beta-lactams currently available in the clinic. Crude IMP-10 MBL was prepared from two clinical isolates of Pseudomonas aeruginosa harbouring the bla(IMP-10) gene. The sensitivity of beta-lactams to hydrolysis by IMP-10 MBL was determined by comparing the MICs of 14 antipseudomonal beta-lactams against a susceptible strain of P. aeruginosa in the presence and absence of IMP-10 MBL. Carbapenems (imipenem, meropenem and panipenem) and extended-spectrum cephems (ceftazidime, cefoperazone, cefsulodin and cefepime) were sensitive to the hydrolysing activity of IMP-10 MBL. By comparison, the fourth-generation cephem (cefpirome), the extended-spectrum penicillins (carbenicillin, ticarcillin, piperacillin and mezlocillin) and monobactams (aztreonam and carumonam) were relatively resistant to IMP-10 MBL. The sensitivity profile of antipseudomonal beta-lactams to IMP-10 MBL generated in the present study provides a valuable reference for antibiotic selection by medical professionals.


Asunto(s)
Monobactamas/metabolismo , beta-Lactamasas/metabolismo , beta-Lactamas/metabolismo , Cefalosporinas/metabolismo , Hidrólisis , Pruebas de Sensibilidad Microbiana , Monobactamas/farmacología , Penicilinas/metabolismo , Sensibilidad y Especificidad , Especificidad por Sustrato , beta-Lactamas/farmacología
19.
Enferm Infecc Microbiol Clin ; 23(9): 525-8, 2005 Nov.
Artículo en Español | MEDLINE | ID: mdl-16324563

RESUMEN

INTRODUCTION: Expanded-spectrum betalactamases (ESBLs) are the main source of resistance to oxyimino cephalosporins and monobactams in Enterobacteriaceae. Most of them derive from TEM or SHV, however the incidence of other families like CTX-M, OXA and PER has increased. In Argentina, the most frequent ESBL in Enterobacteriaceae is CTX-M-2. This specific circumstance, which differs from the situation in the Northern Hemisphere, motivated us to study new diagnostic strategies for the detection of ESBLs in our region. METHOD: Microbiological ESBL detection was performed by double-disk synergy tests, cefotaxime and ceftazidime disks with and without clavulanic acid (NCCLS), and cefotaxime and ceftazidime disks in Müeller-Hinton agar supplemented with lithium clavulanate (MH-cla). Betalactamases were characterized by isoelectric focusing, hydrolysis profile and PCR amplification. RESULTS: Among 575 clinical isolates of Enterobacteriaceae, 14% were oxyimino cephalosporin-resistant. Two different ESBLs were detected in 31 resistant strains: CTX-M-2 (28) and PER-2 groups (3). The double-disk synergy test was the least sensitive method for ESBL detection. ESBLs were detected by the other two methods in all isolates with the use of cefotaxime disks, but not with ceftazidime disks. CONCLUSION: The microbiological method employing MH-cla with cefotaxime disks had a sensitivity and specificity comparable to the referral test using the same antibiotic proposed by the NCCLS for the detection of ESBLs.


Asunto(s)
Proteínas Bacterianas/análisis , Cefalosporinas/metabolismo , Infecciones por Enterobacteriaceae/microbiología , Enterobacteriaceae/enzimología , Pruebas de Sensibilidad Microbiana/métodos , Monobactamas/metabolismo , Resistencia betalactámica , beta-Lactamasas/análisis , Argentina/epidemiología , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/metabolismo , Cefotaxima/metabolismo , Cefotaxima/farmacología , Ceftazidima/metabolismo , Ceftazidima/farmacología , Cefalosporinas/clasificación , Cefalosporinas/farmacología , Enterobacteriaceae/genética , Enterobacteriaceae/aislamiento & purificación , Infecciones por Enterobacteriaceae/epidemiología , Humanos , Hidrólisis , Focalización Isoeléctrica , Monobactamas/clasificación , Monobactamas/farmacología , Reacción en Cadena de la Polimerasa , Sensibilidad y Especificidad , Especificidad por Sustrato , Resistencia betalactámica/genética , beta-Lactamasas/clasificación , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
20.
Can J Microbiol ; 50(3): 137-65, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15105882

RESUMEN

Among the extended-spectrum beta-lactamases, the cefotaximases (CTX-M-ases) constitute a rapidly growing cluster of enzymes that have disseminated geographically. The CTX-M-ases, which hydrolyze cefotaxime efficiently, are mostly encoded by transferable plasmids, and the enzymes have been found predominantly in Enterobacteriaceae, most prevalently in Escherichia coli, Salmonella typhimurium, Klebsiella pneumoniae, and Proteus mirabilis. Isolates of Vibrio cholerae, Acinetobacter baumannii, and Aeromonas hydrophila encoding CTX-M-ases have also been reported. The CTX-M-ases belong to the molecular class A beta-lactamases, and the enzymes are functionally characterized as extended-spectrum beta-lactamases. This group of beta-lactamases confers resistance to penicillins, extended-spectrum cephalosporins, and monobactams, and the enzymes are inhibited by clavulanate, sulbactam, and tazobactam. Typically, the CTX-M-ases hydrolyze cefotaxime more efficiently than ceftazidime, which is reflected in substantially higher MICs to cefotaxime than to ceftazidime. Phylogenetically, the CTX-M-ases are divided into four subfamilies that seem to have descended from chromosomal beta-lactamases of Kluyvera spp. Insertion sequences, especially ISEcp1, have been found adjacent to genes encoding enzymes of all four subfamilies. The class I integron-associated orf513 also seems to be involved in the mobilization of blaCTX-M genes. This review discusses the phylogeny and the hydrolytic properties of the CTX-M-ases, as well as their geographic occurrence and mode of spread.


Asunto(s)
Cefotaxima/metabolismo , Farmacorresistencia Bacteriana/genética , Ácido Penicilánico/análogos & derivados , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Cefalosporinas/metabolismo , Ácido Clavulánico/farmacología , Enterobacteriaceae/efectos de los fármacos , Enterobacteriaceae/enzimología , Enterobacteriaceae/genética , Inhibidores Enzimáticos/farmacología , Transferencia de Gen Horizontal , Monobactamas/metabolismo , Ácido Penicilánico/farmacología , Penicilinas/metabolismo , Filogenia , Sulbactam/farmacología , Tazobactam
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...