Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Antimicrob Agents Chemother ; 66(4): e0212421, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35311523

RESUMEN

ß-Lactamase-mediated resistance to ceftazidime-avibactam (CZA) is a serious limitation in the treatment of Gram-negative bacteria harboring Klebsiella pneumoniae carbapenemase (KPC). Herein, the basis of susceptibility to carbapenems and resistance to ceftazidime (CAZ) and CZA of the D179Y variant of KPC-2 and -3 was explored. First, we determined that resistance to CZA in a laboratory strain of Escherichia coli DH10B was not due to increased expression levels of the variant enzymes, as demonstrated by reverse transcription PCR (RT-PCR). Using timed mass spectrometry, the D179Y variant formed prolonged acyl-enzyme complexes with imipenem (IMI) and meropenem (MEM) in KPC-2 and KPC-3, which could be detected up to 24 h, suggesting that IMI and MEM act as covalent ß-lactamase inhibitors more than as substrates for D179Y KPC-2 and -3. This prolonged acyl-enzyme complex of IMI and MEM by D179Y variants was not observed with wild-type (WT) KPCs. CAZ was studied and the D179Y variants also formed acyl-enzyme complexes (1 to 2 h). Thermal denaturation and differential scanning fluorimetry showed that the tyrosine substitution at position 179 destabilized the KPC ß-lactamases (KPC-2/3 melting temperature [Tm] of 54 to 55°C versus D179Y Tm of 47.5 to 51°C), and the D179Y protein was 3% disordered compared to KPC-2 at 318 K. Heteronuclear 1H/15N-heteronuclear single quantum coherence (HSQC) nuclear magnetic resonance (NMR) spectroscopy also revealed that the D179Y variant, compared to KPC-2, is partially disordered. Based upon these observations, we discuss the impact of disordering of the Ω loop as a consequence of the D179Y substitution. These conformational changes and disorder in the overall structure as a result of D179Y contribute to this unanticipated phenotype.


Asunto(s)
Ceftazidima , Infecciones por Klebsiella , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Compuestos de Azabiciclo/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ceftazidima/farmacología , Combinación de Medicamentos , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Imipenem/farmacología , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae , Espectroscopía de Resonancia Magnética , Meropenem/farmacología , Pruebas de Sensibilidad Microbiana , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
2.
Artículo en Inglés | MEDLINE | ID: mdl-32393499

RESUMEN

Mycobacterium abscessus is a highly drug-resistant nontuberculous mycobacterium (NTM). Efforts to discover new treatments for M. abscessus infections are accelerating, with a focus on cell wall synthesis proteins (M. abscessus l,d-transpeptidases 1 to 5 [LdtMab1 to LdtMab5] and d,d-carboxypeptidase) that are targeted by ß-lactam antibiotics. A challenge to this approach is the presence of chromosomally encoded ß-lactamase (BlaMab). Using a mechanism-based approach, we found that a novel ceftaroline-imipenem combination effectively lowered the MICs of M. abscessus isolates (MIC50 ≤ 0.25 µg/ml; MIC90 ≤ 0.5 µg/ml). Combining ceftaroline and imipenem with a ß-lactamase inhibitor, i.e., relebactam or avibactam, demonstrated only a modest effect on susceptibility compared to each of the ß-lactams alone. In steady-state kinetic assays, BlaMab exhibited a lower Ki app (0.30 ± 0.03 µM for avibactam and 136 ± 14 µM for relebactam) and a higher acylation rate for avibactam (k2/K = 3.4 × 105 ± 0.4 × 105 M-1 s-1 for avibactam and 6 × 102 ± 0.6 × 102 M-1 s-1 for relebactam). The kcat/Km was nearly 10-fold lower for ceftaroline fosamil (0.007 ± 0.001 µM-1 s-1) than for imipenem (0.056 ± 0.006 µM-1 s-1). Timed mass spectrometry captured complexes of avibactam and BlaMab, LdtMab1, LdtMab2, LdtMab4, and d,d-carboxypeptidase, whereas relebactam bound only BlaMab, LdtMab1, and LdtMab2 Interestingly, LdtMab1, LdtMab2, LdtMab4, LdtMab5, and d,d-carboxypeptidase bound only to imipenem when incubated with imipenem and ceftaroline fosamil. We next determined the binding constants of imipenem and ceftaroline fosamil for LdtMab1, LdtMab2, LdtMab4, and LdtMab5 and showed that imipenem bound >100-fold more avidly than ceftaroline fosamil to LdtMab1 and LdtMab2 (e.g., Ki app or Km of LdtMab1 = 0.01 ± 0.01 µM for imipenem versus 0.73 ± 0.08 µM for ceftaroline fosamil). Molecular modeling indicates that LdtMab2 readily accommodates imipenem, but the active site must widen to ≥8 Å for ceftaroline to enter. Our analysis demonstrates that ceftaroline and imipenem binding to multiple targets (l,d-transpeptidases and d,d-carboxypeptidase) and provides a mechanistic rationale for the effectiveness of this dual ß-lactam combination in M. abscessus infections.


Asunto(s)
Mycobacterium abscessus , Peptidil Transferasas , Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , Carboxipeptidasas , Cefalosporinas , Imipenem/farmacología , Pruebas de Sensibilidad Microbiana , Inhibidores de beta-Lactamasas , Ceftarolina
3.
Artículo en Inglés | MEDLINE | ID: mdl-31712217

RESUMEN

Unlike for classes A and B, a standardized amino acid numbering scheme has not been proposed for the class C (AmpC) ß-lactamases, which complicates communication in the field. Here, we propose a scheme developed through a collaborative approach that considers both sequence and structure, preserves traditional numbering of catalytically important residues (Ser64, Lys67, Tyr150, and Lys315), is adaptable to new variants or enzymes yet to be discovered and includes a variation for genetic and epidemiological applications.


Asunto(s)
Proteínas Bacterianas/clasificación , Bacterias Gramnegativas/genética , Bacterias Grampositivas/genética , Mutación , Terminología como Asunto , Resistencia betalactámica/genética , beta-Lactamasas/clasificación , Secuencia de Aminoácidos , Antibacterianos/química , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Expresión Génica , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/enzimología , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/enzimología , Cooperación Internacional , Estructura Secundaria de Proteína , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , beta-Lactamas/química , beta-Lactamas/farmacología
4.
Artículo en Inglés | MEDLINE | ID: mdl-30718248

RESUMEN

Burkholderia spp. are opportunistic human pathogens that infect persons with cystic fibrosis and the immunocompromised. Burkholderia spp. express class A and C ß-lactamases, which are transcriptionally regulated by PenRA through linkage to cell wall metabolism and ß-lactam exposure. The potency of temocillin, a 6-methoxy-ß-lactam, was tested against a panel of multidrug-resistant (MDR) Burkholderia spp. In addition, the mechanistic basis of temocillin activity was assessed and compared to that of ticarcillin. Susceptibility testing with temocillin and ticarcillin was conducted, as was biochemical analysis of the PenA1 class A ß-lactamase and AmpC1 class C ß-lactamase. Molecular dynamics simulations (MDS) were performed using PenA1 with temocillin and ticarcillin. The majority (86.7%) of 150 MDR Burkholderia strains were susceptible to temocillin, while only 4% of the strains were susceptible to ticarcillin. Neither temocillin nor ticarcillin induced bla expression. Ticarcillin was hydrolyzed by PenA1 (kcat/Km = 1.7 ± 0.2 µM-1 s-1), while temocillin was slow to form a favorable complex (apparent Ki [Ki app] = ∼2 mM). Ticarcillin and temocillin were both potent inhibitors of AmpC1, with Ki app values of 4.9 ± 1.0 µM and 4.3 ± 0.4 µM, respectively. MDS of PenA revealed that ticarcillin is in an advantageous position for acylation and deacylation. Conversely, with temocillin, active-site residues K73 and S130 are rotated and the catalytic water molecule is displaced, thereby slowing acylation and allowing the 6-methoxy of temocillin to block deacylation. Temocillin is a ß-lactam with potent activity against Burkholderia spp., as it does not induce bla expression and is poorly hydrolyzed by endogenous ß-lactamases.


Asunto(s)
Antibacterianos/farmacología , Burkholderia/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Penicilinas/farmacología , beta-Lactamas/farmacología , Burkholderia/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Ticarcilina/farmacología , Estados Unidos , beta-Lactamasas/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-31235626

RESUMEN

The diazabicyclooctane (DBO) avibactam (AVI) reversibly inactivates most serine-ß-lactamases. Previous investigations showed that inhibition constants of AVI toward class A PER-2 are reminiscent of values observed for class C and D ß-lactamases (i.e., k2/K of ≈103 M-1 s-1) but lower than other class A ß-lactamases (i.e., k2/K = 104 to 105 M-1 s-1). Herein, biochemical and structural studies were conducted with PER-2 and AVI to explore these differences. Furthermore, biochemical studies on Arg220 and Thr237 variants with AVI were conducted to gain deeper insight into the mechanism of PER-2 inactivation. The main biochemical and structural observations revealed the following: (i) both amino-acid substitutions in Arg220 and the rich hydrophobic content in the active site hinder the binding of catalytic waters and acylation, impairing AVI inhibition; (ii) movement of Ser130 upon binding of AVI favors the formation of a hydrogen bond with the sulfate group of AVI; and (iii) the Thr237Ala substitution alters the AVI inhibition constants. The acylation constant (k2/K) of PER-2 by AVI is primarily influenced by stabilizing hydrogen bonds involving AVI and important residues such as Thr237 and Arg220. (Variants in Arg220 demonstrate a dramatic reduction in k2/K) We also observed that displacement of Ser130 side chain impairs AVI acylation, an observation not made in other extended-spectrum ß-lactamases (ESBLs). Comparatively, relebactam combined with a ß-lactam is more potent against Escherichia coli producing PER-2 variants than ß-lactam-AVI combinations. Our findings provide a rationale for evaluating the utility of the currently available DBO inhibitors against unique ESBLs like PER-2 and anticipate the effectiveness of these inhibitors toward variants that may eventually be selected upon AVI usage.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/química , Sustitución de Aminoácidos , Arginina , Compuestos de Azabiciclo/química , Compuestos de Azabiciclo/metabolismo , Dominio Catalítico , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Mutación , Conformación Proteica , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/metabolismo , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
6.
Artículo en Inglés | MEDLINE | ID: mdl-30858223

RESUMEN

Impeding, as well as reducing, the burden of antimicrobial resistance in Gram-negative pathogens is an urgent public health endeavor. Our current antibiotic armamentarium is dwindling, while major resistance determinants (e.g., extended-spectrum ß-lactamases [ESBLs]) continue to evolve and disseminate around the world. One approach to attack this problem is to develop novel therapies that will protect our current agents. AAI101 is a novel penicillanic acid sulfone ß-lactamase inhibitor similar in structure to tazobactam, with one important difference. AAI101 possesses a strategically placed methyl group that gives the inhibitor a net neutral charge, enhancing bacterial cell penetration. AAI101 paired with cefepime, also a zwitterion, is in phase III of clinical development for the treatment of serious Gram-negative infections. Here, AAI101 was found to restore the activity of cefepime against class A ESBLs (e.g., CTX-M-15) and demonstrated increased potency compared to that of piperacillin-tazobactam when tested against an established isogenic panel. The enzymological properties of AAI101 further revealed that AAI101 possessed a unique mechanism of ß-lactamase inhibition compared to that of tazobactam. Additionally, upon reaction with AAI101, CTX-M-15 was modified to an inactive state. Notably, the in vivo efficacy of cefepime-AAI101 was demonstrated using a mouse septicemia model, indicating the ability of AAI101 to bolster significantly the therapeutic efficacy of cefepime in vivo The combination of AAI101 with cefepime represents a potential carbapenem-sparing treatment regimen for infections suspected to be caused by Enterobacteriaceae expressing ESBLs.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Cefepima/farmacología , Enterobacteriaceae/efectos de los fármacos , Enterobacteriaceae/metabolismo , Combinación Piperacilina y Tazobactam/farmacología , Sulbactam/farmacología , Triazoles/farmacología , Inhibidores de beta-Lactamasas/farmacología , Espectrometría de Masa por Ionización de Electrospray
7.
Artículo en Inglés | MEDLINE | ID: mdl-31182530

RESUMEN

Carbapenem-resistant Enterobacteriaceae (CRE) are resistant to most antibiotics, making CRE infections extremely difficult to treat with available agents. Klebsiella pneumoniae carbapenemases (KPC-2 and KPC-3) are predominant carbapenemases in CRE in the United States. Nacubactam is a bridged diazabicyclooctane (DBO) ß-lactamase inhibitor that inactivates class A and C ß-lactamases and exhibits intrinsic antibiotic and ß-lactam "enhancer" activity against Enterobacteriaceae In this study, we examined a collection of meropenem-resistant K. pneumoniae isolates carrying blaKPC-2 or blaKPC-3; meropenem-nacubactam restored susceptibility. Upon testing isogenic Escherichia coli strains producing KPC-2 variants with single-residue substitutions at important Ambler class A positions (K73, S130, R164, E166, N170, D179, K234, E276, etc.), the K234R variant increased the meropenem-nacubactam MIC compared to that for the strain producing KPC-2, without increasing the meropenem MIC. Correspondingly, nacubactam inhibited KPC-2 (apparent Ki [Ki app] = 31 ± 3 µM) more efficiently than the K234R variant (Ki app = 270 ± 27 µM) and displayed a faster acylation rate (k2/K), which was 5,815 ± 582 M-1 s-1 for KPC-2 versus 247 ± 25 M-1 s-1 for the K234R variant. Unlike avibactam, timed mass spectrometry revealed an intact sulfate on nacubactam and a novel peak (+337 Da) with the K234R variant. Molecular modeling of the K234R variant showed significant catalytic residue (i.e., S70, K73, and S130) rearrangements that likely interfere with nacubactam binding and acylation. Nacubactam's aminoethoxy tail formed unproductive interactions with the K234R variant's active site. Molecular modeling and docking observations were consistent with the results of biochemical analyses. Overall, the meropenem-nacubactam combination is effective against carbapenem-resistant K. pneumoniae Moreover, our data suggest that ß-lactamase inhibition by nacubactam proceeds through an alternative mechanism compared to that for avibactam.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Enterobacteriaceae Resistentes a los Carbapenémicos/efectos de los fármacos , Enterobacteriaceae Resistentes a los Carbapenémicos/metabolismo , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/metabolismo , Meropenem/farmacología , beta-Lactamasas/metabolismo , Acilación/efectos de los fármacos , Compuestos de Azabiciclo/farmacología , Carbapenémicos/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Humanos , Pruebas de Sensibilidad Microbiana/métodos , Inhibidores de beta-Lactamasas/farmacología
8.
J Clin Microbiol ; 57(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31167848

RESUMEN

In persons with cystic fibrosis (CF), airway infection with Burkholderia cepacia complex (Bcc) species or Burkholderia gladioli presents a significant challenge due to inherent resistance to multiple antibiotics. Two chromosomally encoded inducible ß-lactamases, a Pen-like class A and AmpC are produced in Bcc and B. gladioli Previously, ceftazidime-avibactam demonstrated significant potency against Bcc and B. gladioli isolated from the sputum of individuals with CF; however, 10% of the isolates tested resistant to ceftazidime-avibactam. Here, we describe an alternative antibiotic combination to overcome ceftazidime-avibactam resistance. Antimicrobial susceptibility testing was performed on Bcc and B. gladioli clinical and control isolates. Biochemical analysis was conducted on purified PenA1 and AmpC1 ß-lactamases from Burkholderia multivorans ATCC 17616. Analytic isoelectric focusing and immunoblotting were conducted on cellular extracts of B. multivorans induced by various ß-lactams or ß-lactam-ß-lactamase inhibitor combinations. Combinations of piperacillin-avibactam, as well as piperacillin-tazobactam plus ceftazidime-avibactam (the clinically available counterpart), were tested against a panel of ceftazidime-avibactam nonsusceptible Bcc and B. gladioli The piperacillin-avibactam and piperacillin-tazobactam-ceftazidime-avibactam combinations restored susceptibility to 99% of the isolates tested. Avibactam is a potent inhibitor of PenA1 (apparent inhibitory constant [Kiapp] = 0.5 µM), while piperacillin was found to inhibit AmpC1 (Kiapp = 2.6 µM). Moreover, piperacillin, tazobactam, ceftazidime, and avibactam, as well as combinations thereof, did not induce expression of blapenA1 and blaampC1 in the B. multivorans ATCC 17616 background. When ceftazidime-avibactam is combined with piperacillin-tazobactam, the susceptibility of Bcc and B. gladioli to ceftazidime and piperacillin is restored in vitro Both the lack of blapenA1 induction and potent inactivation of PenA1 by avibactam likely provide the major contributions toward susceptibility. With in vivo validation, piperacillin-tazobactam-ceftazidime-avibactam may represent salvage therapy for individuals with CF and highly drug-resistant Bcc and B. gladioli infections.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Complejo Burkholderia cepacia/efectos de los fármacos , Burkholderia gladioli/efectos de los fármacos , Fibrosis Quística/microbiología , Farmacorresistencia Bacteriana Múltiple , Piperacilina/farmacología , Antibacterianos/farmacología , Infecciones por Burkholderia/tratamiento farmacológico , Fibrosis Quística/complicaciones , Sustitución de Medicamentos , Humanos , Cinética , Pruebas de Sensibilidad Microbiana
9.
Artículo en Inglés | MEDLINE | ID: mdl-30012762

RESUMEN

Burkholderia multivorans is a member of the Burkholderia cepacia complex, a group of >20 related species of nosocomial pathogens that commonly infect individuals suffering from cystic fibrosis. ß-Lactam antibiotics are recommended as therapy for infections due to Bmultivorans, which possesses two ß-lactamase genes, blapenA and blaAmpC PenA is a carbapenemase with a substrate profile similar to that of the Klebsiella pneumoniae carbapenemase (KPC); in addition, expression of PenA is inducible by ß-lactams in Bmultivorans Here, we characterize AmpC from Bmultivorans ATCC 17616. AmpC possesses only 38 to 46% protein identity with non-Burkholderia AmpC proteins (e.g., PDC-1 and CMY-2). Among 49 clinical isolates of Bmultivorans, we identified 27 different AmpC variants. Some variants possessed single amino acid substitutions within critical active-site motifs (Ω loop and R2 loop). Purified AmpC1 demonstrated minimal measurable catalytic activity toward ß-lactams (i.e., nitrocefin and cephalothin). Moreover, avibactam was a poor inhibitor of AmpC1 (Kiapp > 600 µM), and acyl-enzyme complex formation with AmpC1 was slow, likely due to lack of productive interactions with active-site residues. Interestingly, immunoblotting using a polyclonal anti-AmpC antibody revealed that protein expression of AmpC1 was inducible in Bmultivorans ATCC 17616 after growth in subinhibitory concentrations of imipenem (1 µg/ml). AmpC is a unique inducible class C cephalosporinase that may play an ancillary role in Bmultivorans compared to PenA, which is the dominant ß-lactamase in Bmultivorans ATCC 17616.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Burkholderia/efectos de los fármacos , Burkholderia/enzimología , beta-Lactamasas/química , beta-Lactamasas/metabolismo , beta-Lactamas/farmacología , Secuencia de Aminoácidos , Compuestos de Azabiciclo/farmacología , Cefalosporinasa/química , Cefalosporinasa/metabolismo , Cefalosporinas/farmacología , Cefalotina/farmacología , Imipenem/farmacología , Pruebas de Sensibilidad Microbiana , Estructura Secundaria de Proteína
10.
Artículo en Inglés | MEDLINE | ID: mdl-29439972

RESUMEN

Ceftazidime-avibactam is a "second-generation" ß-lactam-ß-lactamase inhibitor combination that is effective against Enterobacteriaceae expressing class A extended-spectrum ß-lactamases, class A carbapenemases, and/or class C cephalosporinases. Knowledge of the interactions of avibactam, a diazabicyclooctane with different ß-lactamases, is required to anticipate future resistance threats. FOX family ß-lactamases possess unique hydrolytic properties with a broadened substrate profile to include cephamycins, partly as a result of an isoleucine at position 346, instead of the conserved asparagine found in most AmpCs. Interestingly, a single amino acid substitution at N346 in the Citrobacter AmpC is implicated in resistance to the aztreonam-avibactam combination. In order to understand how diverse active-site topologies affect avibactam inhibition, we tested a panel of clinical Enterobacteriaceae isolates producing blaFOX using ceftazidime-avibactam, determined the biochemical parameters for inhibition using the FOX-4 variant, and probed the atomic structure of avibactam with FOX-4. Avibactam restored susceptibility to ceftazidime for most isolates producing blaFOX; two isolates, one expressing blaFOX-4 and the other producing blaFOX-5, displayed an MIC of 16 µg/ml for the combination. FOX-4 possessed a k2/K value of 1,800 ± 100 M-1 · s-1 and an off rate (koff) of 0.0013 ± 0.0003 s-1 Mass spectrometry showed that the FOX-4-avibactam complex did not undergo chemical modification for 24 h. Analysis of the crystal structure of FOX-4 with avibactam at a 1.5-Å resolution revealed a unique characteristic of this AmpC ß-lactamase. Unlike in the Pseudomonas-derived cephalosporinase 1 (PDC-1)-avibactam crystal structure, interactions (e.g., hydrogen bonding) between avibactam and position I346 in FOX-4 are not evident. Furthermore, another residue is not observed to be close enough to compensate for the loss of these critical hydrogen-bonding interactions. This observation supports findings from the inhibition analysis of FOX-4; FOX-4 possessed the highest Kd (dissociation constant) value (1,600 nM) for avibactam compared to other AmpCs (7 to 660 nM). Medicinal chemists must consider the properties of extended-spectrum AmpCs, such as the FOX ß-lactamases, for the design of future diazabicyclooctanes.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Proteínas Bacterianas/metabolismo , beta-Lactamasas/metabolismo , Sustitución de Aminoácidos , Ceftazidima/farmacología , Combinación de Medicamentos , Activación Enzimática/efectos de los fármacos , Proteínas de Escherichia coli/metabolismo , Pruebas de Sensibilidad Microbiana , Pseudomonas/enzimología
11.
Artículo en Inglés | MEDLINE | ID: mdl-29530851

RESUMEN

Pseudomonas aeruginosa is a prevalent and life-threatening Gram-negative pathogen. Pseudomonas-derived cephlosporinase (PDC) is the major inducible cephalosporinase in P. aeruginosa In this investigation, we show that relebactam, a diazabicyclooctane ß-lactamase inhibitor, potently inactivates PDC-3, with a k2/K of 41,400 M-1 s-1 and a koff of 0.00095 s-1 Relebactam restored susceptibility to imipenem in 62% of multidrug-resistant P. aeruginosa clinical isolates, while only 21% of isolates were susceptible to imipenem-cilastatin alone. Relebactam promises to increase the efficacy of imipenem-cilastatin against P. aeruginosa.


Asunto(s)
Cefalosporinasa/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas/efectos de los fármacos , Compuestos de Azabiciclo/farmacología , Cilastatina/farmacología , Imipenem/farmacología , Pruebas de Sensibilidad Microbiana , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo
12.
Artículo en Inglés | MEDLINE | ID: mdl-29610205

RESUMEN

The imipenem-relebactam combination is in development as a potential treatment regimen for infections caused by Enterobacteriaceae possessing complex ß-lactamase backgrounds. Relebactam is a ß-lactamase inhibitor that possesses the diazabicyclooctane core, as in avibactam; however, the R1 side chain of relebactam also includes a piperidine ring, whereas that of avibactam is a carboxyamide. Here, we investigated the inactivation of the Klebsiella pneumoniae carbapenemase KPC-2, the most widespread class A carbapenemase, by relebactam and performed susceptibility testing with imipenem-relebactam using KPC-producing clinical isolates of Enterobacteriaceae MIC measurements using agar dilution methods revealed that all 101 clinical isolates of KPC-producing Enterobacteriaceae (K. pneumoniae, Klebsiella oxytoca, Enterobacter cloacae, Enterobacter aerogenes, Citrobacter freundii, Citrobacter koseri, and Escherichia coli) were highly susceptible to imipenem-relebactam (MICs ≤ 2 mg/liter). Relebactam inhibited KPC-2 with a second-order onset of acylation rate constant (k2/K) value of 24,750 M-1 s-1 and demonstrated a slow off-rate constant (koff) of 0.0002 s-1 Biochemical analysis using time-based mass spectrometry to map intermediates revealed that the KPC-2-relebactam acyl-enzyme complex was stable for up to 24 h. Importantly, desulfation of relebactam was not observed using mass spectrometry. Desulfation and subsequent deacylation have been observed during the reaction of KPC-2 with avibactam. Upon molecular dynamics simulations of relebactam in the KPC-2 active site, we found that the positioning of active-site water molecules is less favorable for desulfation in the KPC-2 active site than it is in the KPC-2-avibactam complex. In the acyl complexes, the water molecules are within 2.5 to 3 Å of the avibactam sulfate; however, they are more than 5 to 6 Å from the relebactam sulfate. As a result, we propose that the KPC-2-relebactam acyl complex is more stable than the KPC-2-avibactam complex. The clinical implications of this difference are not currently known.


Asunto(s)
Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Imipenem/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo , Enterobacteriaceae Resistentes a los Carbapenémicos/efectos de los fármacos , Dominio Catalítico/efectos de los fármacos , Combinación de Medicamentos , Humanos , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/aislamiento & purificación , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular
13.
Artículo en Inglés | MEDLINE | ID: mdl-28784669

RESUMEN

Stenotrophomonas maltophilia is an emerging opportunistic pathogen, classified by the World Health Organization as one of the leading multidrug-resistant organisms in hospital settings. The need to discover novel compounds and/or combination therapies for S. maltophilia is urgent. We demonstrate the in vitro efficacy of aztreonam-avibactam (ATM-AVI) against S. maltophilia and kinetically characterize the inhibition of the L2 ß-lactamase by avibactam. ATM-AVI overcomes aztreonam resistance in selected clinical strains of S. maltophilia, addressing an unmet medical need.


Asunto(s)
Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , Aztreonam/farmacología , Stenotrophomonas maltophilia/efectos de los fármacos , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/efectos de los fármacos , Combinación de Medicamentos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones por Bacterias Gramnegativas/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Stenotrophomonas maltophilia/aislamiento & purificación
14.
Antibiotics (Basel) ; 12(5)2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37237794

RESUMEN

BACKGROUND: Ceftazidime-avibactam was approved by the FDA to treat infections caused by Enterobacterales carrying blaKPC-2. However, variants of KPC-2 with amino acid substitutions at position 179 have emerged and confer resistance to ceftazidime-avibactam. METHODS: The activity of imipenem-relebactam was assessed against a panel of 19 KPC-2 D179 variants. KPC-2 and the D179N and D179Y variants were purified for biochemical analyses. Molecular models were constructed with imipenem to assess differences in kinetic profiles. RESULTS: All strains were susceptible to imipenem-relebactam, but resistant to ceftazidime (19/19) and ceftazidime-avibactam (18/19). KPC-2 and the D179N variant hydrolyzed imipenem, but the D179N variant's rate was much slower. The D179Y variant was unable to turnover imipenem. All three ß-lactamases hydrolyzed ceftazidime at varying rates. The acylation rate of relebactam for the D179N variant was ~2.5× lower than KPC-2. Poor catalytic turnover by the D179Y variant precluded the determination of inhibitory kinetic parameters. Acyl-complexes with imipenem and ceftazidime were less prevalent with the D179N variant compared to the D179Y variant, supporting the kinetic observations that the D179Y variant was not as active as the D179N variant. Relebactam was slower to form an acyl-complex with the D179Y variant compared to avibactam. The D179Y model with imipenem revealed that the catalytic water molecule was shifted, and the carbonyl of imipenem was not within the oxyanion hole. Conversely in the D179N model, imipenem was oriented favorably for deacylation. CONCLUSIONS: Imipenem-relebactam overcame the resistance of the D179 variants, suggesting that this combination will be active against clinical isolates harboring these derivatives of KPC-2.

15.
mBio ; 10(2)2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30862744

RESUMEN

Multidrug-resistant (MDR) Acinetobacter spp. poses a significant therapeutic challenge in part due to the presence of chromosomally encoded ß-lactamases, including class C Acinetobacter-derived cephalosporinases (ADC) and class D oxacillinases (OXA), as well as plasmid-mediated class A ß-lactamases. Importantly, OXA-like ß-lactamases represent a gap in the spectrum of inhibition by recently approved ß-lactamase inhibitors such as avibactam and vaborbactam. ETX2514 is a novel, rationally designed, diazabicyclooctenone inhibitor that effectively targets class A, C, and D ß-lactamases. We show that addition of ETX2514 significantly increased the susceptibility of clinical Acinetobacterbaumannii isolates to sulbactam. AdeB and AdeJ were identified to be key efflux constituents for ETX2514 in A. baumannii The combination of sulbactam and ETX2514 was efficacious against A. baumannii carrying blaTEM-1, blaADC-82, blaOXA-23, and blaOXA-66 in a neutropenic murine thigh infection model. We also show that, in vitro, ETX2514 inhibited ADC-7 (k2/Ki 1.0 ± 0.1 × 106 M-1 s-1) and OXA-58 (k2/Ki 2.5 ± 0.3 × 105 M-1 s-1). Cocrystallization of ETX2514 with OXA-24/40 revealed hydrogen bonding interactions between ETX2514 and residues R261, S219, and S128 of OXA-24/40 in addition to a chloride ion occupied in the active site. Further, the C3 methyl group of ETX2514 shifts the position of M223. In conclusion, the sulbactam-ETX2514 combination possesses a broadened inhibitory range to include class D ß-lactamases as well as class A and C ß-lactamases and is a promising therapeutic candidate for infections caused by MDR Acinetobacter spp.IMPORTANCE The number and diversity of ß-lactamases are steadily increasing. The emergence of ß-lactamases that hydrolyze carbapenems poses a significant threat to our antibiotic armamentarium. The explosion of OXA enzymes that are carbapenem hydrolyzers is a major challenge (carbapenem-hydrolyzing class D [CHD]). An urgent need exists to discover ß-lactamase inhibitors with class D activity. The sulbactam-ETX2514 combination demonstrates the potential to become a treatment regimen of choice for Acinetobacter spp. producing class D ß-lactamases.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/administración & dosificación , Compuestos de Azabiciclo/administración & dosificación , Sulbactam/administración & dosificación , Inhibidores de beta-Lactamasas/administración & dosificación , Infecciones por Acinetobacter/microbiología , Animales , Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Ratones , Unión Proteica , Conformación Proteica , Sulbactam/farmacología , Resultado del Tratamiento , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/química , beta-Lactamasas/metabolismo
16.
Sci Rep ; 8(1): 12916, 2018 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-30150677

RESUMEN

The rise of multi-drug resistance in bacterial pathogens is one of the grand challenges facing medical science. A major concern is the speed of development of ß-lactamase-mediated resistance in Gram-negative species, thus putting at risk the efficacy of the most recently approved antibiotics and inhibitors, including carbapenems and avibactam, respectively. New strategies to overcome resistance are urgently required, which will ultimately be facilitated by a deeper understanding of the mechanisms that regulate the function of ß-lactamases such as the Klebsiella Pneumoniae carbapenemases (KPCs). Using enhanced sampling computational methods together with site-directed mutagenesis, we report the identification of two "hydrophobic networks" in the KPC-2 enzyme, the integrity of which has been found to be essential for protein stability and corresponding resistance. Present throughout the structure, these networks are responsible for the structural integrity and allosteric signaling. Disruption of the networks leads to a loss of the KPC-2 mediated resistance phenotype, resulting in restored susceptibility to different classes of ß-lactam antibiotics including carbapenems and cephalosporins. The "hydrophobic networks" were found to be highly conserved among class-A ß-lactamases, which implies their suitability for exploitation as a potential target for therapeutic intervention.


Asunto(s)
Antibacterianos/farmacología , beta-Lactamasas/farmacología , Compuestos de Azabiciclo/farmacología , Carbapenémicos/farmacología , Farmacorresistencia Microbiana , Klebsiella pneumoniae/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Estructura Secundaria de Proteína
17.
mBio ; 9(6)2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30538183

RESUMEN

Pseudomonas aeruginosa produces a class C ß-lactamase (e.g., PDC-3) that robustly hydrolyzes early generation cephalosporins often at the diffusion limit; therefore, bacteria possessing these ß-lactamases are resistant to many ß-lactam antibiotics. In response to this significant clinical threat, ceftolozane, a 3' aminopyrazolium cephalosporin, was developed. Combined with tazobactam, ceftolozane promised to be effective against multidrug-resistant P. aeruginosa Alarmingly, Ω-loop variants of the PDC ß-lactamase (V213A, G216R, E221K, E221G, and Y223H) were identified in ceftolozane/tazobactam-resistant P. aeruginosa clinical isolates. Herein, we demonstrate that the Escherichia coli strain expressing the E221K variant of PDC-3 had the highest minimum inhibitory concentrations (MICs) against a panel of ß-lactam antibiotics, including ceftolozane and ceftazidime, a cephalosporin that differs in structure largely in the R2 side chain. The kcat values of the E221K variant for both substrates were equivalent, whereas the Km for ceftolozane (341 ± 64 µM) was higher than that for ceftazidime (174 ± 20 µM). Timed mass spectrometry, thermal stability, and equilibrium unfolding studies revealed key mechanistic insights. Enhanced sampling molecular dynamics simulations identified conformational changes in the E221K variant Ω-loop, where a hidden pocket adjacent to the catalytic site opens and stabilizes ceftolozane for efficient hydrolysis. Encouragingly, the diazabicyclooctane ß-lactamase inhibitor avibactam restored susceptibility to ceftolozane and ceftazidime in cells producing the E221K variant. In addition, a boronic acid transition state inhibitor, LP-06, lowered the ceftolozane and ceftazidime MICs by 8-fold for the E221K-expressing strain. Understanding these structural changes in evolutionarily selected variants is critical toward designing effective ß-lactam/ß-lactamase inhibitor therapies for P. aeruginosa infections.IMPORTANCE The presence of ß-lactamases (e.g., PDC-3) that have naturally evolved and acquired the ability to break down ß-lactam antibiotics (e.g., ceftazidime and ceftolozane) leads to highly resistant and potentially lethal Pseudomonas aeruginosa infections. We show that wild-type PDC-3 ß-lactamase forms an acyl enzyme complex with ceftazidime, but it cannot accommodate the structurally similar ceftolozane that has a longer R2 side chain with increased basicity. A single amino acid substitution from a glutamate to a lysine at position 221 in PDC-3 (E221K) causes the tyrosine residue at 223 to adopt a new position poised for efficient hydrolysis of both cephalosporins. The importance of the mechanism of action of the E221K variant, in particular, is underscored by its evolutionary recurrences in multiple bacterial species. Understanding the biochemical and molecular basis for resistance is key to designing effective therapies and developing new ß-lactam/ß-lactamase inhibitor combinations.


Asunto(s)
Antibacterianos/farmacología , Resistencia a las Cefalosporinas , Cefalosporinas/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Tazobactam/farmacología , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Expresión Génica , Cinética , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Conformación Proteica , Pliegue de Proteína , Estabilidad Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Temperatura , beta-Lactamasas/química , beta-Lactamasas/genética
18.
J Med Chem ; 61(9): 4067-4086, 2018 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-29627985

RESUMEN

Limited treatment options exist to combat infections caused by multidrug-resistant (MDR) Gram-negative bacteria possessing broad-spectrum ß-lactamases. The design of novel ß-lactamase inhibitors is of paramount importance. Here, three novel diazabicyclooctanes (DBOs), WCK 5153, zidebactam (WCK 5107), and WCK 4234 (compounds 1-3, respectively), were synthesized and biochemically characterized against clinically important bacteria. Compound 3 inhibited class A, C, and D ß-lactamases with unprecedented k2/ K values against OXA carbapenemases. Compounds 1 and 2 acylated class A and C ß-lactamses rapidly but not the tested OXAs. Compounds 1-3 formed highly stable acyl-complexes as demonstrated by mass spectrometry. Crystallography revealed that 1-3 complexed with KPC-2 adopted a "chair conformation" with the sulfate occupying the carboxylate binding region. The cefepime-2 and meropenem-3 combinations were effective in murine peritonitis and neutropenic lung infection models caused by MDR Acinetobacter baumannii. Compounds 1-3 are novel ß-lactamase inhibitors that demonstate potent cross-class inhibition, and clinical studies targeting MDR infections are warranted.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Compuestos Bicíclicos con Puentes/farmacología , Ciclooctanos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Octanos/farmacología , Piperidinas/farmacología , Resistencia betalactámica/efectos de los fármacos , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo , beta-Lactamas/farmacología , Animales , Compuestos de Azabiciclo/química , Compuestos de Azabiciclo/uso terapéutico , Compuestos Bicíclicos con Puentes/química , Compuestos Bicíclicos con Puentes/uso terapéutico , Ciclooctanos/química , Ciclooctanos/uso terapéutico , Sinergismo Farmacológico , Pulmón/efectos de los fármacos , Pulmón/microbiología , Ratones , Octanos/química , Octanos/uso terapéutico , Peritonitis/tratamiento farmacológico , Peritonitis/microbiología , Piperidinas/química , Piperidinas/uso terapéutico , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/uso terapéutico
19.
mBio ; 8(5)2017 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-29089425

RESUMEN

The emergence of Klebsiella pneumoniae carbapenemases (KPCs), ß-lactamases that inactivate "last-line" antibiotics such as imipenem, represents a major challenge to contemporary antibiotic therapies. The combination of ceftazidime (CAZ) and avibactam (AVI), a potent ß-lactamase inhibitor, represents an attempt to overcome this formidable threat and to restore the efficacy of the antibiotic against Gram-negative bacteria bearing KPCs. CAZ-AVI-resistant clinical strains expressing KPC variants with substitutions in the Ω-loop are emerging. We engineered 19 KPC-2 variants bearing targeted mutations at amino acid residue Ambler position 179 in Escherichia coli and identified a unique antibiotic resistance phenotype. We focus particularly on the CAZ-AVI resistance of the clinically relevant Asp179Asn variant. Although this variant demonstrated less hydrolytic activity, we demonstrated that there was a prolonged period during which an acyl-enzyme intermediate was present. Using mass spectrometry and transient kinetic analysis, we demonstrated that Asp179Asn "traps" ß-lactams, preferentially binding ß-lactams longer than AVI owing to a decreased rate of deacylation. Molecular dynamics simulations predict that (i) the Asp179Asn variant confers more flexibility to the Ω-loop and expands the active site significantly; (ii) the catalytic nucleophile, S70, is shifted more than 1.5 Å and rotated more than 90°, altering the hydrogen bond networks; and (iii) E166 is displaced by 2 Å when complexed with ceftazidime. These analyses explain the increased hydrolytic profile of KPC-2 and suggest that the Asp179Asn substitution results in an alternative complex mechanism leading to CAZ-AVI resistance. The future design of novel ß-lactams and ß-lactamase inhibitors must consider the mechanistic basis of resistance of this and other threatening carbapenemases.IMPORTANCE Antibiotic resistance is emerging at unprecedented rates and threatens to reach crisis levels. One key mechanism of resistance is the breakdown of ß-lactam antibiotics by ß-lactamase enzymes. KPC-2 is a ß-lactamase that inactivates carbapenems and ß-lactamase inhibitors (e.g., clavulanate) and is prevalent around the world, including in the United States. Resistance to the new antibiotic ceftazidime-avibactam, which was designed to overcome KPC resistance, had already emerged within a year. Using protein engineering, we uncovered a mechanism by which resistance to this new drug emerges, which could arm scientists with the ability to forestall such resistance to future drugs.


Asunto(s)
Antibacterianos/metabolismo , Compuestos de Azabiciclo/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ceftazidima/metabolismo , Farmacorresistencia Bacteriana Múltiple/genética , Klebsiella pneumoniae/enzimología , Klebsiella pneumoniae/genética , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Sustitución de Aminoácidos , Antibacterianos/farmacología , Asparagina/química , Asparagina/genética , Proteínas Bacterianas/química , Combinación de Medicamentos , Humanos , Cinética , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Simulación de Dinámica Molecular , Mutación , Fenotipo , Ingeniería de Proteínas/métodos , beta-Lactamasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA