Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Mol Biol ; 436(12): 168603, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38729259

RESUMEN

OXA-66 is a member of the OXA-51 subfamily of class D ß-lactamases native to the Acinetobacter genus that includes Acinetobacter baumannii, one of the ESKAPE pathogens and a major cause of drug-resistant nosocomial infections. Although both wild type OXA-66 and OXA-51 have low catalytic activity, they are ubiquitous in the Acinetobacter genomes. OXA-51 is also remarkably thermostable. In addition, newly emerging, single and double amino acid variants show increased activity against carbapenems, indicating that the OXA-51 subfamily is growing and gaining clinical significance. In this study, we used molecular dynamics simulations, X-ray crystallography, and thermal denaturation data to examine and compare the dynamics of OXA-66 wt and its gain-of-function variants: I129L (OXA-83), L167V (OXA-82), P130Q (OXA-109), P130A, and W222L (OXA-234). Our data indicate that OXA-66 wt also has a high melting temperature, and its remarkable stability is due to an extensive and rigid hydrophobic bridge formed by a number of residues around the active site and harbored by the three loops, P, Ω, and ß5-ß6. Compared to the WT enzyme, the mutants exhibit higher flexibility only in the loop regions, and are more stable than other robust carbapenemases, such as OXA-23 and OXA-24/40. All the mutants show increased rotational flexibility of residues I129 and W222, which allows carbapenems to bind. Overall, our data support the hypothesis that structural features in OXA-51 and OXA-66 promote evolution of multiple highly stable variants with increased clinical relevance in A. baumannii.

2.
J Med Chem ; 66(13): 8510-8525, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37358467

RESUMEN

Class C Acinetobacter-derived cephalosporinases (ADCs) represent an important target for inhibition in the multidrug-resistant pathogen Acinetobacter baumannii. Many ADC variants have emerged, and characterization of their structural and functional differences is essential. Equally as important is the development of compounds that inhibit all prevalent ADCs despite these differences. The boronic acid transition state inhibitor, MB076, a novel heterocyclic triazole with improved plasma stability, was synthesized and inhibits seven different ADC ß-lactamase variants with Ki values <1 µM. MB076 acted synergistically in combination with multiple cephalosporins to restore susceptibility. ADC variants containing an alanine duplication in the Ω-loop, specifically ADC-33, exhibited increased activity for larger cephalosporins, such as ceftazidime, cefiderocol, and ceftolozane. X-ray crystal structures of ADC variants in this study provide a structural context for substrate profile differences and show that the inhibitor adopts a similar conformation in all ADC variants, despite small changes near their active sites.


Asunto(s)
Acinetobacter baumannii , Cefalosporinasa , Cefalosporinasa/genética , Cefalosporinasa/química , Cefalosporinasa/farmacología , Ácidos Borónicos/farmacología , Ácidos Borónicos/química , Cefalosporinas/farmacología , beta-Lactamasas/genética , beta-Lactamasas/química , Antibacterianos/farmacología , Pruebas de Sensibilidad Microbiana
3.
Antibiotics (Basel) ; 12(4)2023 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-37107006

RESUMEN

Acinetobacter baumannii is a Gram-negative organism listed as an urgent threat pathogen by the World Health Organization (WHO). Carbapenem-resistant A. baumannii (CRAB), especially, present therapeutic challenges due to complex mechanisms of resistance to ß-lactams. One of the most important mechanisms is the production of ß-lactamase enzymes capable of hydrolyzing ß-lactam antibiotics. Co-expression of multiple classes of ß-lactamases is present in CRAB; therefore, the design and synthesis of "cross-class" inhibitors is an important strategy to preserve the efficacy of currently available antibiotics. To identify new, nonclassical ß-lactamase inhibitors, we previously identified a sulfonamidomethaneboronic acid CR167 active against Acinetobacter-derived class C ß-lactamases (ADC-7). The compound demonstrated affinity for ADC-7 with a Ki = 160 nM and proved to be able to decrease MIC values of ceftazidime and cefotaxime in different bacterial strains. Herein, we describe the activity of CR167 against other ß-lactamases in A. baumannii: the cefepime-hydrolysing class C extended-spectrum ß-lactamase (ESAC) ADC-33 and the carbapenem-hydrolyzing OXA-24/40 (class D). These investigations demonstrate CR167 as a valuable cross-class (C and D) inhibitor, and the paper describes our attempts to further improve its activity. Five chiral analogues of CR167 were rationally designed and synthesized. The structures of OXA-24/40 and ADC-33 in complex with CR167 and select chiral analogues were obtained. The structure activity relationships (SARs) are highlighted, offering insights into the main determinants for cross-class C/D inhibitors and impetus for novel drug design.

4.
J Biol Chem ; 298(7): 102127, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35709986

RESUMEN

The evolution of multidrug resistance in Acinetobacter spp. increases the risk of our best antibiotics losing their efficacy. From a clinical perspective, the carbapenem-hydrolyzing class D ß-lactamase subfamily present in Acinetobacter spp. is particularly concerning because of its ability to confer resistance to carbapenems. The kinetic profiles of class D ß-lactamases exhibit variability in carbapenem hydrolysis, suggesting functional differences. To better understand the structure-function relationship between the carbapenem-hydrolyzing class D ß-lactamase OXA-24/40 found in Acinetobacter baumannii and carbapenem substrates, we analyzed steady-state kinetics with the carbapenem antibiotics meropenem and ertapenem and determined the structures of complexes of OXA-24/40 bound to imipenem, meropenem, doripenem, and ertapenem, as well as the expanded-spectrum cephalosporin cefotaxime, using X-ray crystallography. We show that OXA-24/40 exhibits a preference for ertapenem compared with meropenem, imipenem, and doripenem, with an increase in catalytic efficiency of up to fourfold. We suggest that superposition of the nine OXA-24/40 complexes will better inform future inhibitor design efforts by providing insight into the complicated and varying ways in which carbapenems are selected and bound by class D ß-lactamases.


Asunto(s)
Proteínas Bacterianas , Carbapenémicos , beta-Lactamasas , Acinetobacter baumannii/enzimología , Antibacterianos/química , Antibacterianos/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Carbapenémicos/química , Carbapenémicos/metabolismo , Hidrólisis , Pruebas de Sensibilidad Microbiana , Conformación Proteica , Especificidad por Sustrato , beta-Lactamasas/química , beta-Lactamasas/metabolismo
5.
Diagn Microbiol Infect Dis ; 99(2): 115242, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33248392

RESUMEN

Successful treatment of Acinetobacter baumannii infections require early and appropriate antimicrobial therapy. One of the first steps in this process is understanding which ß-lactamase (bla) alleles are present and in what combinations. Thus, we performed WGS on 98 carbapenem-resistant A. baumannii (CR Ab). In most isolates, an acquired blaOXA carbapenemase was found in addition to the intrinsic blaOXA allele. The most commonly found allele was blaOXA-23 (n = 78/98). In some isolates, blaOXA-23 was found in addition to other carbapenemase alleles: blaOXA-82 (n = 12/78), blaOXA-72 (n = 2/78) and blaOXA-24/40 (n = 1/78). Surprisingly, 20% of isolates carried carbapenemases not routinely assayed for by rapid molecular diagnostic platforms, i.e., blaOXA-82 and blaOXA-172; all had ISAba1 elements. In 8 CR Ab, blaOXA-82 or blaOXA-172 was the only carbapenemase. Both blaOXA-24/40 and its variant blaOXA-72 were each found in 6/98 isolates. The most prevalent ADC variants were blaADC-30 (21%), blaADC-162 (21%), and blaADC-212 (26%). Complete combinations are reported.


Asunto(s)
Acinetobacter baumannii/genética , Proteínas Bacterianas/genética , Carbapenémicos/farmacología , Resistencia betalactámica/genética , beta-Lactamasas/genética , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/enzimología , Acinetobacter baumannii/aislamiento & purificación , Genoma Bacteriano/genética , Humanos
6.
Antimicrob Agents Chemother ; 64(12)2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-32988830

RESUMEN

Extended-spectrum class C ß-lactamases have evolved to rapidly inactivate expanded-spectrum cephalosporins, a class of antibiotics designed to be resistant to hydrolysis by ß-lactamase enzymes. To better understand the mechanism by which Acinetobacter-derived cephalosporinase-7 (ADC-7), a chromosomal AmpC enzyme, hydrolyzes these molecules, we determined the X-ray crystal structure of ADC-7 in an acyl-enzyme complex with the cephalosporin ceftazidime (2.40 Å) as well as in complex with a boronic acid transition state analog inhibitor that contains the R1 side chain of ceftazidime (1.67 Å). In the acyl-enzyme complex, the carbonyl oxygen is situated in the oxyanion hole where it makes key stabilizing interactions with the main chain nitrogens of Ser64 and Ser315. The boronic acid O1 hydroxyl group is similarly positioned in this area. Conserved residues Gln120 and Asn152 form hydrogen bonds with the amide group of the R1 side chain in both complexes. These complexes represent two steps in the hydrolysis of expanded-spectrum cephalosporins by ADC-7 and offer insight into the inhibition of ADC-7 by ceftazidime through displacement of the deacylating water molecule as well as blocking its trajectory to the acyl carbonyl carbon. In addition, the transition state analog inhibitor, LP06, was shown to bind with high affinity to ADC-7 (Ki , 50 nM) and was able to restore ceftazidime susceptibility, offering the potential for optimization efforts of this type of inhibitor.


Asunto(s)
Acinetobacter , Ácidos Borónicos , Ceftazidima , Cefalosporinasa , Antibacterianos/farmacología , Ácidos Borónicos/farmacología , Ceftazidima/farmacología , Cefalosporinasa/efectos de los fármacos , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas
7.
ACS Infect Dis ; 6(7): 1965-1975, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32502340

RESUMEN

Boronic acid transition state inhibitors (BATSIs) are known reversible covalent inhibitors of serine ß-lactamases. The selectivity and high potency of specific BATSIs bearing an amide side chain mimicking the ß-lactam's amide side chain are an established and recognized synthetic strategy. Herein, we describe a new class of BATSIs where the amide group is replaced by a bioisostere triazole; these compounds were designed as molecular probes. To this end, a library of 26 α-triazolylmethaneboronic acids was synthesized and tested against the clinically concerning Acinetobacter-derived cephalosporinase, ADC-7. In steady state analyses, these compounds demonstrated Ki values ranging from 90 nM to 38 µM (±10%). Five compounds were crystallized in complex with ADC-7 ß-lactamase, and all the crystal structures reveal the triazole is in the putative amide binding site, thus confirming the triazole-amide bioisosterism. The easy synthetic access of these new inhibitors as prototype scaffolds allows the insertion of a wide range of chemical groups able to explore the enzyme binding site and provides insights on the importance of specific residues in recognition and catalysis. The best inhibitor identified, compound 6q (Ki 90 nM), places a tolyl group near Arg340, making favorable cation-π interactions. Notably, the structure of 6q does not resemble the natural substrate of the ß-lactamase yet displays a pronounced inhibition activity, in addition to lowering the minimum inhibitory concentration (MIC) of ceftazidime against three bacterial strains expressing class C ß-lactamases. In summary, these observations validate the α-triazolylboronic acids as a promising template for further inhibitor design.


Asunto(s)
Acinetobacter baumannii , Inhibidores de beta-Lactamasas , Acinetobacter baumannii/metabolismo , Cefalosporinasa/genética , Cefalosporinasa/metabolismo , Relación Estructura-Actividad , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo
8.
ACS Infect Dis ; 4(3): 325-336, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29144724

RESUMEN

Acinetobacter baumannii is a multidrug resistant pathogen that infects more than 12 000 patients each year in the US. Much of the resistance to ß-lactam antibiotics in Acinetobacter spp. is mediated by class C ß-lactamases known as Acinetobacter-derived cephalosporinases (ADCs). ADCs are unaffected by clinically used ß-lactam-based ß-lactamase inhibitors. In this study, five boronic acid transition state analog inhibitors (BATSIs) were evaluated for inhibition of the class C cephalosporinase ADC-7. Our goal was to explore the properties of BATSIs designed to probe the R1 binding site. Ki values ranged from low micromolar to subnanomolar, and circular dichroism (CD) demonstrated that each inhibitor stabilizes the ß-lactamase-inhibitor complexes. Additionally, X-ray crystal structures of ADC-7 in complex with five inhibitors were determined (resolutions from 1.80 to 2.09 Å). In the ADC-7/CR192 complex, the BATSI with the lowest Ki (0.45 nM) and greatest Δ Tm (+9 °C), a trifluoromethyl substituent, interacts with Arg340. Arg340 is unique to ADCs and may play an important role in the inhibition of ADC-7. The ADC-7/BATSI complexes determined in this study shed light into the unique recognition sites in ADC enzymes and also offer insight into further structure-based optimization of these inhibitors.


Asunto(s)
Acinetobacter/enzimología , Ácidos Borónicos/química , Ácidos Borónicos/farmacología , Cefalosporinasa/química , Cefalosporinasa/metabolismo , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/farmacología , Sitios de Unión , Dicroismo Circular , Cristalografía por Rayos X , Modelos Moleculares , Unión Proteica , Conformación Proteica
9.
ACS Infect Dis ; 4(3): 337-348, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29144725

RESUMEN

Boronic acids are attracting a lot of attention as ß-lactamase inhibitors, and in particular, compound S02030 ( Ki = 44 nM) proved to be a good lead compound against ADC-7 ( Acinetobacter-derived cephalosporinase), one of the most significant resistance determinants in A. baumannii. The atomic structure of the ADC-7/S02030 complex highlighted the importance of critical structural determinants for recognition of the boronic acids. Herein, to elucidate the role in recognition of the R2-carboxylate, which mimics the C3/C4 found in ß-lactams, we designed, synthesized, and characterized six derivatives of S02030 (3a). Out of the six compounds, the best inhibitors proved to be those with an explicit negative charge (compounds 3a-c, 3h, and 3j, Ki = 44-115 nM), which is in contrast to the derivatives where the negative charge is omitted, such as the amide derivative 3d ( Ki = 224 nM) and the hydroxyamide derivative 3e ( Ki = 155 nM). To develop a structural characterization of inhibitor binding in the active site, the X-ray crystal structures of ADC-7 in a complex with compounds 3c, SM23, and EC04 were determined. All three compounds share the same structural features as in S02030 but only differ in the carboxy-R2 side chain, thereby providing the opportunity of exploring the distinct binding mode of the negatively charged R2 side chain. This cephalosporinase demonstrates a high degree of versatility in recognition, employing different residues to directly interact with the carboxylate, thus suggesting the existence of a "carboxylate binding region" rather than a binding site in ADC enzymes. Furthermore, this class of compounds was tested against resistant clinical strains of A. baumannii and are effective at inhibiting bacterial growth in conjunction with a ß-lactam antibiotic.


Asunto(s)
Acinetobacter/enzimología , Ácidos Borónicos/química , Ácidos Borónicos/farmacología , Cefalosporinasa/química , Cefalosporinasa/metabolismo , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/farmacología , Sitios de Unión , Ácidos Borónicos/síntesis química , Cristalografía por Rayos X , Modelos Moleculares , Estructura Molecular , Unión Proteica , Conformación Proteica , Inhibidores de beta-Lactamasas/síntesis química
10.
Biochem J ; 475(1): 273-288, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29229762

RESUMEN

OXA-239 is a class D carbapenemase isolated from an Acinetobacter baumannii strain found in Mexico. This enzyme is a variant of OXA-23 with three amino acid substitutions in or near the active site. These substitutions cause OXA-239 to hydrolyze late-generation cephalosporins and the monobactam aztreonam with greater efficiency than OXA-23. OXA-239 activity against the carbapenems doripenem and imipenem is reduced ∼3-fold and 20-fold, respectively. Further analysis demonstrated that two of the substitutions (P225S and D222N) are largely responsible for the observed alteration of kinetic parameters, while the third (S109L) may serve to stabilize the protein. Structures of OXA-239 with cefotaxime, doripenem and imipenem bound as acyl-intermediates were determined. These structures reveal that OXA-239 has increased flexibility in a loop that contains P225S and D222N. When carbapenems are bound, the conformation of this loop is essentially identical with that observed previously for OXA-23, with a narrow active site that makes extensive contacts to the ligand. When cefotaxime is bound, the loop can adopt a different conformation that widens the active site to allow binding of that bulky drug. This alternate conformation is made possible by P225S and further stabilized by D222N. Taken together, these results suggest that the three substitutions were selected to expand the substrate specificity profile of OXA-23 to cephalosporins and monobactams. The loss of activity against imipenem, however, suggests that there may be limits to the plasticity of class D enzymes with regard to evolving active sites that can effectively bind multiple classes of ß-lactam drugs.


Asunto(s)
Acinetobacter baumannii/enzimología , Sustitución de Aminoácidos , Proteínas Bacterianas/química , Carbapenémicos/química , Cefotaxima/química , Imipenem/química , beta-Lactamasas/química , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Carbapenémicos/metabolismo , Carbapenémicos/farmacología , Dominio Catalítico , Cefotaxima/metabolismo , Cefotaxima/farmacología , Clonación Molecular , Cristalografía por Rayos X , Doripenem , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Imipenem/metabolismo , Imipenem/farmacología , Cinética , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
11.
Protein Sci ; 26(3): 515-526, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27997706

RESUMEN

ß-lactam antibiotics are crucial to the management of bacterial infections in the medical community. Due to overuse and misuse, clinically significant bacteria are now resistant to many commercially available antibiotics. The most widespread resistance mechanism to ß-lactams is the expression of ß-lactamase enzymes. To overcome ß-lactamase mediated resistance, inhibitors were designed to inactivate these enzymes. However, current inhibitors (clavulanic acid, tazobactam, and sulbactam) for ß-lactamases also contain the characteristic ß-lactam ring, making them susceptible to resistance mechanisms employed by bacteria. This presents a critical need for novel, non-ß-lactam inhibitors that can circumvent these resistance mechanisms. The carbapenem-hydrolyzing class D ß-lactamases (CHDLs) are of particular concern, given that they efficiently hydrolyze potent carbapenem antibiotics. Unfortunately, these enzymes are not inhibited by clinically available ß-lactamase inhibitors, nor are they effectively inhibited by the newest, non-ß-lactam inhibitor, avibactam. Boronic acids are known transition state analog inhibitors of class A and C ß-lactamases, and are not extensively characterized as inhibitors of class D ß-lactamases. Importantly, boronic acids provide a novel way to potentially inhibit class D ß-lactamases. Sixteen boronic acids were selected and tested for inhibition of the CHDL OXA-24/40. Several compounds were identified as effective inhibitors of OXA-24/40, with Ki values as low as 5 µM. The X-ray crystal structures of OXA-24/40 in complex with BA3, BA4, BA8, and BA16 were determined and revealed the importance of interactions with hydrophobic residues Tyr112 and Trp115. These boronic acids serve as progenitors in optimization efforts of a novel series of inhibitors for class D ß-lactamases.


Asunto(s)
Ácidos Borónicos/química , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química , Cristalografía por Rayos X , Dominios Proteicos
12.
Protein Sci ; 25(12): 2152-2163, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27636561

RESUMEN

OXA-51 is a class D ß-lactamase that is thought to be the native carbapenemase of Acinetobacter baumannii. Many variants of OXA-51 containing active site substitutions have been identified from A. baumannii isolates, and some of these substitutions increase hydrolytic activity toward carbapenem antibiotics. We have determined the high-resolution structures of apo OXA-51 and OXA-51 with one such substitution (I129L) with the carbapenem doripenem trapped in the active site as an acyl-intermediate. The structure shows that acyl-doripenem adopts an orientation very similar to carbapenem ligands observed in the active site of OXA-24/40 (doripenem) and OXA-23 (meropenem). In the OXA-51 variant/doripenem complex, the indole ring of W222 is oriented away from the doripenem binding site, thereby eliminating a clash that is predicted to occur in wildtype OXA-51. Similarly, in the OXA-51 variant complex, L129 adopts a different rotamer compared to I129 in wildtype OXA-51. This alternative position moves its side chain away from the hydroxyethyl moiety of doripenem and relieves another potential clash between the enzyme and carbapenem substrates. Molecular dynamics simulations of OXA-51 and OXA-51 I129L demonstrate that compared to isoleucine, a leucine at this position greatly favors a rotamer that accommodates the ligand. These results provide a molecular justification for how this substitution generates enhanced binding affinity for carbapenems, and therefore helps explain the prevalence of this substitution in clinical OXA-51 variants.


Asunto(s)
Acinetobacter baumannii/enzimología , Carbapenémicos/química , Simulación de Dinámica Molecular , beta-Lactamasas/química , Acinetobacter baumannii/genética , Sustitución de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Doripenem , Mutación Missense , beta-Lactamasas/genética
13.
Curr Drug Targets ; 17(9): 1051-60, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26073861

RESUMEN

ß-lactam antibiotics have revolutionized modern medicine, but resistance to these drugs is a major public health crisis. Traditionally, class C ß-lactamases were referred to as cephalosporinases due to their substrate preference for this particular class of ß-lactams. However, the emergence of AmpC enzymes with extended-spectrum activity (extended-spectrum cephalosporinases or ESACs) is particularly worrisome, especially given that most clinical ß-lactamase inhibitors are ineffective against these enzymes. This review summarizes structures of several extended spectrum class C ß-lactamases and analyzes the structure-function relationship observed among them.


Asunto(s)
Cefalosporinasa/química , Cefalosporinasa/metabolismo , Inhibidores de beta-Lactamasas/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Diseño de Fármacos , Humanos , Ligandos , Modelos Moleculares , Conformación Proteica , Resistencia betalactámica , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/química , beta-Lactamasas/metabolismo
14.
Biochemistry ; 54(10): 1976-87, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25710192

RESUMEN

The carbapenem-hydrolyzing class D ß-lactamases OXA-23 and OXA-24/40 have emerged worldwide as causative agents for ß-lactam antibiotic resistance in Acinetobacter species. Many variants of these enzymes have appeared clinically, including OXA-160 and OXA-225, which both contain a P → S substitution at homologous positions in the OXA-24/40 and OXA-23 backgrounds, respectively. We purified OXA-160 and OXA-225 and used steady-state kinetic analysis to compare the substrate profiles of these variants to their parental enzymes, OXA-24/40 and OXA-23. OXA-160 and OXA-225 possess greatly enhanced hydrolytic activities against aztreonam, ceftazidime, cefotaxime, and ceftriaxone when compared to OXA-24/40 and OXA-23. These enhanced activities are the result of much lower Km values, suggesting that the P → S substitution enhances the binding affinity of these drugs. We have determined the structures of the acylated forms of OXA-160 (with ceftazidime and aztreonam) and OXA-225 (ceftazidime). These structures show that the R1 oxyimino side-chain of these drugs occupies a space near the ß5-ß6 loop and the omega loop of the enzymes. The P → S substitution found in OXA-160 and OXA-225 results in a deviation of the ß5-ß6 loop, relieving the steric clash with the R1 side-chain carboxypropyl group of aztreonam and ceftazidime. These results reveal worrying trends in the enhancement of substrate spectrum of class D ß-lactamases but may also provide a map for ß-lactam improvement.


Asunto(s)
Acinetobacter baumannii/enzimología , Aztreonam/química , Proteínas Bacterianas/química , Cefalosporinas/química , beta-Lactamasas/química , Hidrólisis , Cinética , Estructura Secundaria de Proteína
15.
Biochemistry ; 53(48): 7670-9, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25380506

RESUMEN

ß-Lactam resistance in Acinetobacter baumannii presents one of the greatest challenges to contemporary antimicrobial chemotherapy. Much of this resistance to cephalosporins derives from the expression of the class C ß-lactamase enzymes, known as Acinetobacter-derived cephalosporinases (ADCs). Currently, ß-lactamase inhibitors are structurally similar to ß-lactam substrates and are not effective inactivators of this class C cephalosporinase. Herein, two boronic acid transition state inhibitors (BATSIs S02030 and SM23) that are chemically distinct from ß-lactams were designed and tested for inhibition of ADC enzymes. BATSIs SM23 and S02030 bind with high affinity to ADC-7, a chromosomal cephalosporinase from Acinetobacter baumannii (Ki = 21.1 ± 1.9 nM and 44.5 ± 2.2 nM, respectively). The X-ray crystal structures of ADC-7 were determined in both the apo form (1.73 Å resolution) and in complex with S02030 (2.0 Å resolution). In the complex, S02030 makes several canonical interactions: the O1 oxygen of S02030 is bound in the oxyanion hole, and the R1 amide group makes key interactions with conserved residues Asn152 and Gln120. In addition, the carboxylate group of the inhibitor is meant to mimic the C3/C4 carboxylate found in ß-lactams. The C3/C4 carboxylate recognition site in class C enzymes is comprised of Asn346 and Arg349 (AmpC numbering), and these residues are conserved in ADC-7. Interestingly, in the ADC-7/S02030 complex, the inhibitor carboxylate group is observed to interact with Arg340, a residue that distinguishes ADC-7 from the related class C enzyme AmpC. A thermodynamic analysis suggests that ΔH driven compounds may be optimized to generate new lead agents. The ADC-7/BATSI complex provides insight into recognition of non-ß-lactam inhibitors by ADC enzymes and offers a starting point for the structure-based optimization of this class of novel ß-lactamase inhibitors against a key resistance target.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/enzimología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/química , Cefalosporinasa/química , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/farmacología , Acinetobacter baumannii/genética , Proteínas Bacterianas/genética , Fenómenos Biofísicos , Ácidos Borónicos/química , Ácidos Borónicos/farmacología , Dominio Catalítico , Cefalosporinasa/genética , Cristalografía por Rayos X , Diseño de Fármacos , Cinética , Modelos Moleculares , Estructura Molecular , Electricidad Estática , Termodinámica , Resistencia betalactámica/genética
16.
Bioorg Med Chem ; 22(13): 3351-9, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24835785

RESUMEN

ß-Lactams are the most widely prescribed class of antibiotics, yet their efficacy is threatened by expression of ß-lactamase enzymes, which hydrolyze the defining lactam ring of these antibiotics. To overcome resistance due to ß-lactamases, inhibitors that do not resemble ß-lactams are needed. A novel, non-ß-lactam inhibitor for the class C ß-lactamase AmpC (3-[(4-chloroanilino)sulfonyl]thiophene-2-carboxylic acid; Ki 26µM) was previously identified. Based on this lead, a series of compounds with the potential to interact with residues at the edge of the active site were synthesized and tested for inhibition of AmpC. The length of the carbon chain spacer was extended by 1, 2, 3, and 4 carbons between the integral thiophene ring and the benzene ring (compounds 4, 5, 6, and 7). Compounds 4 and 6 showed minimal improvement over the lead compound (Ki 18 and 19µM, respectively), and compound 5 inhibited to the same extent as the lead. The X-ray crystal structures of AmpC in complexes with compounds 4, 5, and 6 were determined. The complexes provide insight into the structural reasons for the observed inhibition, and inform future optimization efforts in this series.


Asunto(s)
Compuestos de Anilina/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Tiofenos/farmacología , Compuestos de Anilina/síntesis química , Compuestos de Anilina/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Tiofenos/síntesis química , Tiofenos/química , beta-Lactamasas/metabolismo
17.
Antimicrob Agents Chemother ; 58(1): 333-41, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24165180

RESUMEN

Since the discovery and use of penicillin, the increase of antibiotic resistance among bacterial pathogens has become a major health concern. The most prevalent resistance mechanism in Gram-negative bacteria is due to ß-lactamase expression. Class D ß-lactamases are of particular importance due to their presence in multidrug-resistant Acinetobacter baumannii and Pseudomonas aeruginosa. The class D enzymes were initially characterized by their ability to efficiently hydrolyze isoxazolyl-type ß-lactams like oxacillin. Due to this substrate preference, these enzymes are traditionally referred to as oxacillinases or OXAs. However, this class is comprised of subfamilies characterized by diverse activities that include oxacillinase, carbapenemase, or cephalosporinase substrate specificity. OXA-1 represents one subtype of class D enzyme that efficiently hydrolyzes oxacillin, and OXA-24/40 represents another with weak oxacillinase, but increased carbapenemase, activity. To examine the structural basis for the substrate selectivity differences between OXA-1 and OXA-24/40, the X-ray crystal structures of deacylation-deficient mutants of these enzymes (Lys70Asp for OXA-1; Lys84Asp for OXA-24) in complexes with oxacillin were determined to 1.4 Å and 2.4 Å, respectively. In the OXA-24/40/oxacillin structure, the hydrophobic R1 side chain of oxacillin disrupts the bridge between Tyr112 and Met223 present in the apo OXA-24/40 structure, causing the main chain of the Met223-containing loop to adopt a completely different conformation. In contrast, in the OXA-1/oxacillin structure, a hydrophobic pocket consisting of Trp102, Met99, Phe217, Leu161, and Leu255 nicely complements oxacillin's nonpolar R1 side chain. Comparison of the OXA-1/oxacillin and OXA-24/40/oxacillin complexes provides novel insight on how substrate selectivity is achieved among subtypes of class D ß-lactamases. By elucidating important active site interactions, these findings can also inform the design of novel antibiotics and inhibitors.


Asunto(s)
beta-Lactamasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Cefalosporinasa/química , Cefalosporinasa/metabolismo , Cristalografía por Rayos X , Oxacilina/metabolismo , Especificidad por Sustrato , beta-Lactamasas/química
18.
Acc Chem Res ; 46(11): 2407-15, 2013 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-23902256

RESUMEN

Despite 70 years of clinical use, ß-lactam antibiotics still remain at the forefront of antimicrobial chemotherapy. The major challenge to these life-saving therapeutics is the presence of bacterial enzymes (i.e., ß-lactamases) that can hydrolyze the ß-lactam bond and inactivate the antibiotic. These enzymes can be grouped into four classes (A-D). Among the most genetically diverse are the class D ß-lactamases. In this class are ß-lactamases that can inactivate the entire spectrum of ß-lactam antibiotics (penicillins, cephalosporins, and carbapenems). Class D ß-lactamases are mostly found in Gram-negative bacteria such as Pseudomonas aeruginosa , Escherichia coli , Proteus mirabilis , and Acinetobacter baumannii . The active-sites of class D ß-lactamases contain an unusual N-carboxylated lysine post-translational modification. A strongly hydrophobic active-site helps create the conditions that allow the lysine to combine with CO2, and the resulting carbamate is stabilized by a number of hydrogen bonds. The carboxy-lysine plays a symmetric role in the reaction, serving as a general base to activate the serine nucleophile in the acylation reaction, and the deacylating water in the second step. There are more than 250 class D ß-lactamases described, and the full set of variants shows remarkable diversity with regard to substrate binding and turnover. Narrow-spectrum variants are most effective against the earliest generation penicillins and cephalosporins such as ampicillin and cephalothin. Extended-spectrum variants (also known as extended-spectrum ß-lactamases, ESBLs) pose a more dangerous clinical threat as they possess a small number of substitutions that allow them to bind and hydrolyze later generation cephalosporins that contain bulkier side-chain constituents (e.g., cefotaxime, ceftazidime, and cefepime). Mutations that permit this versatility seem to cluster in the area surrounding an active-site tryptophan resulting in a widened active-site to accommodate the oxyimino side-chains of these cephalosporins. More concerning are the class D ß-lactamases that hydrolyze clinically important carbapenem ß-lactam drugs (e.g., imipenem). Whereas carbapenems irreversibly acylate and inhibit narrow-spectrum ß-lactamases, class D carbapenemases are able to recruit and activate a deacylating water. The rotational orientation of the C6 hydroxyethyl group found on all carbapenem antibiotics likely plays a role in whether the deacylating water is effective or not. Inhibition of class D ß-lactamases is a current challenge. Commercially available inhibitors that are active against other classes of ß-lactamases are ineffective against class D enzymes. On the horizon are several compounds, consisting of both ß-lactam derivatives and non-ß-lactams, that have the potential of providing novel leads to design new mechanism-based inactivators that are effective against the class D enzymes. Several act synergistically when given in combination with a ß-lactam antibiotic, and others show a unique mechanism of inhibition that is distinct from the traditional ß-lactamase inhibitors. These studies will bolster structure-based inhibitor design efforts to facilitate the optimization and development of these compounds as class D inactivators.


Asunto(s)
Bacterias Gramnegativas/enzimología , beta-Lactamasas/metabolismo , Catálisis , Dominio Catalítico , Modelos Moleculares
19.
Antimicrob Agents Chemother ; 57(10): 4848-55, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23877677

RESUMEN

Class D ß-lactamases that hydrolyze carbapenems such as imipenem and doripenem are a recognized danger to the efficacy of these "last-resort" ß-lactam antibiotics. Like all known class D carbapenemases, OXA-23 cannot hydrolyze the expanded-spectrum cephalosporin ceftazidime. OXA-146 is an OXA-23 subfamily clinical variant that differs from the parent enzyme by a single alanine (A220) inserted in the loop connecting ß-strands ß5 and ß6. We discovered that this insertion enables OXA-146 to bind and hydrolyze ceftazidime with an efficiency comparable to those of other extended-spectrum class D ß-lactamases. OXA-146 also binds and hydrolyzes aztreonam, cefotaxime, ceftriaxone, and ampicillin with higher efficiency than OXA-23 and preserves activity against doripenem. In this study, we report the X-ray crystal structures of both the OXA-23 and OXA-146 enzymes at 1.6-Å and 1.2-Å resolution. A comparison of the two structures shows that the extra alanine moves a methionine (M221) out of its normal position, where it forms a bridge over the top of the active site. This single amino acid insertion also lengthens the ß5-ß6 loop, moving the entire backbone of this region further away from the active site. A model of ceftazidime bound in the active site reveals that these two structural alterations are both likely to relieve steric clashes between the bulky R1 side chain of ceftazidime and OXA-23. With activity against all four classes of ß-lactam antibiotics, OXA-146 represents an alarming new threat to the treatment of infections caused by Acinetobacter spp.


Asunto(s)
Antibacterianos/farmacología , Aztreonam/farmacología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Carbapenémicos/farmacología , Cefalosporinas/farmacología , beta-Lactamasas/química , beta-Lactamasas/metabolismo , Acinetobacter/efectos de los fármacos , Acinetobacter/enzimología , Secuencia de Aminoácidos , Ampicilina/farmacología , Cristalografía por Rayos X , Doripenem , Pruebas de Sensibilidad Microbiana , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
20.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 9): 1189-93, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22948919

RESUMEN

P99 cephalosporinase is a class C ß-lactamase that is responsible in part for the widespread bacterial resistance to ß-lactam antibiotics. Mutations of the conserved active-site residue Asn152 of the enzyme have been shown to alter ß-lactam substrate specificity in vivo. Mutation of Asn152 to a glycine is notable in that it exhibits in vivo substrate-selectivity switching. In order to better understand the structural basis for this observed switch, the X-ray crystal structure of the apo Asn152Gly mutant of P99 was determined to 1.95 Å resolution. Unexpectedly, the artificial C-terminal His(6) tag of a symmetrically-related molecule was observed bound in the active site. The His(6) tag makes several interactions with key active-site residues, as well as with several sulfate ions. Additionally, the overall C-terminus occupies the space left vacant upon the mutation of Asn152 to glycine.


Asunto(s)
Cefalosporinasa/química , Enterobacter cloacae/enzimología , Mutación , Cefalosporinasa/genética , Modelos Moleculares , Estructura Terciaria de Proteína , Homología Estructural de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...