Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Biol Chem ; 299(5): 104606, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36924941

RESUMEN

L1 is a dizinc subclass B3 metallo-ß-lactamase (MBL) that hydrolyzes most ß-lactam antibiotics and is a key resistance determinant in the Gram-negative pathogen Stenotrophomonas maltophilia, an important cause of nosocomial infections in immunocompromised patients. L1 is not usefully inhibited by MBL inhibitors in clinical trials, underlying the need for further studies on L1 structure and mechanism. We describe kinetic studies and crystal structures of L1 in complex with hydrolyzed ß-lactams from the penam (mecillinam), cephem (cefoxitin/cefmetazole), and carbapenem (tebipenem, doripenem, and panipenem) classes. Despite differences in their structures, all the ß-lactam-derived products hydrogen bond to Tyr33, Ser221, and Ser225 and are stabilized by interactions with a conserved hydrophobic pocket. The carbapenem products were modeled as Δ1-imines, with (2S)-stereochemistry. Their binding mode is determined by the presence of a 1ß-methyl substituent: the Zn-bridging hydroxide either interacts with the C-6 hydroxyethyl group (1ß-hydrogen-containing carbapenems) or is displaced by the C-6 carboxylate (1ß-methyl-containing carbapenems). Unexpectedly, the mecillinam product is a rearranged N-formyl amide rather than penicilloic acid, with the N-formyl oxygen interacting with the Zn-bridging hydroxide. NMR studies imply mecillinam rearrangement can occur nonenzymatically in solution. Cephem-derived imine products are bound with (3R)-stereochemistry and retain their 3' leaving groups, likely representing stable endpoints, rather than intermediates, in MBL-catalyzed hydrolysis. Our structures show preferential complex formation by carbapenem- and cephem-derived species protonated on the equivalent (ß) faces and so identify interactions that stabilize diverse hydrolyzed antibiotics. These results may be exploited in developing antibiotics, and ß-lactamase inhibitors, that form long-lasting complexes with dizinc MBLs.


Asunto(s)
Antibacterianos , Inhibidores de beta-Lactamasas , beta-Lactamas , Humanos , Antibacterianos/farmacología , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/química , beta-Lactamas/química , beta-Lactamas/metabolismo , beta-Lactamas/farmacología , Carbapenémicos/metabolismo , Cristalografía , Cinética , Stenotrophomonas maltophilia/efectos de los fármacos , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico
2.
J Am Chem Soc ; 145(13): 7166-7180, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-36972204

RESUMEN

KPC-2 (Klebsiella pneumoniae carbapenemase-2) is a globally disseminated serine-ß-lactamase (SBL) responsible for extensive ß-lactam antibiotic resistance in Gram-negative pathogens. SBLs inactivate ß-lactams via a mechanism involving a hydrolytically labile covalent acyl-enzyme intermediate. Carbapenems, the most potent ß-lactams, evade the activity of many SBLs by forming long-lived inhibitory acyl-enzymes; however, carbapenemases such as KPC-2 efficiently deacylate carbapenem acyl-enzymes. We present high-resolution (1.25-1.4 Å) crystal structures of KPC-2 acyl-enzymes with representative penicillins (ampicillin), cephalosporins (cefalothin), and carbapenems (imipenem, meropenem, and ertapenem) obtained utilizing an isosteric deacylation-deficient mutant (E166Q). The mobility of the Ω-loop (residues 165-170) negatively correlates with antibiotic turnover rates (kcat), highlighting the role of this region in positioning catalytic residues for efficient hydrolysis of different ß-lactams. Carbapenem-derived acyl-enzyme structures reveal the predominance of the Δ1-(2R) imine rather than the Δ2 enamine tautomer. Quantum mechanics/molecular mechanics molecular dynamics simulations of KPC-2:meropenem acyl-enzyme deacylation used an adaptive string method to differentiate the reactivity of the two isomers. These identify the Δ1-(2R) isomer as having a significantly (7 kcal/mol) higher barrier than the Δ2 tautomer for the (rate-determining) formation of the tetrahedral deacylation intermediate. Deacylation is therefore likely to proceed predominantly from the Δ2, rather than the Δ1-(2R) acyl-enzyme, facilitated by tautomer-specific differences in hydrogen-bonding networks involving the carbapenem C-3 carboxylate and the deacylating water and stabilization by protonated N-4, accumulating a negative charge on the Δ2 enamine-derived oxyanion. Taken together, our data show how the flexible Ω-loop helps confer broad-spectrum activity upon KPC-2, while carbapenemase activity stems from efficient deacylation of the Δ2-enamine acyl-enzyme tautomer.


Asunto(s)
Antibacterianos , Carbapenémicos , Carbapenémicos/química , Carbapenémicos/farmacología , Meropenem , Isomerismo , Antibacterianos/farmacología , Antibacterianos/química , beta-Lactamasas/metabolismo , Proteínas Bacterianas , beta-Lactamas , Klebsiella pneumoniae
3.
J Biol Chem ; 296: 100126, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33257320

RESUMEN

Class A serine ß-lactamases (SBLs) are key antibiotic resistance determinants in Gram-negative bacteria. SBLs neutralize ß-lactams via a hydrolytically labile covalent acyl-enzyme intermediate. Klebsiella pneumoniae carbapenemase (KPC) is a widespread SBL that hydrolyzes carbapenems, the most potent ß-lactams; known KPC variants differ in turnover of expanded-spectrum oxyimino-cephalosporins (ESOCs), for example, cefotaxime and ceftazidime. Here, we compare ESOC hydrolysis by the parent enzyme KPC-2 and its clinically observed double variant (P104R/V240G) KPC-4. Kinetic analyses show that KPC-2 hydrolyzes cefotaxime more efficiently than the bulkier ceftazidime, with improved ESOC turnover by KPC-4 resulting from enhanced turnover (kcat), rather than altered KM values. High-resolution crystal structures of ESOC acyl-enzyme complexes with deacylation-deficient (E166Q) KPC-2 and KPC-4 mutants show that ceftazidime acylation causes rearrangement of three loops; the Ω, 240, and 270 loops, which border the active site. However, these rearrangements are less pronounced in the KPC-4 than the KPC-2 ceftazidime acyl-enzyme and are not observed in the KPC-2:cefotaxime acyl-enzyme. Molecular dynamics simulations of KPC:ceftazidime acyl-enyzmes reveal that the deacylation general base E166, located on the Ω loop, adopts two distinct conformations in KPC-2, either pointing "in" or "out" of the active site; with only the "in" form compatible with deacylation. The "out" conformation was not sampled in the KPC-4 acyl-enzyme, indicating that efficient ESOC breakdown is dependent upon the ordering and conformation of the KPC Ω loop. The results explain how point mutations expand the activity spectrum of the clinically important KPC SBLs to include ESOCs through their effects on the conformational dynamics of the acyl-enzyme intermediate.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Ceftazidima/farmacología , Farmacorresistencia Microbiana , Klebsiella pneumoniae/enzimología , Mutación , beta-Lactamasas/química , beta-Lactamasas/metabolismo , Acilación , Antibacterianos/farmacología , Proteínas Bacterianas/genética , Dominio Catalítico , Hidrólisis , Cinética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación Proteica , beta-Lactamasas/genética
4.
Proteins ; 90(2): 372-384, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34455628

RESUMEN

Antibiotic resistance is a major threat to global public health. ß-lactamases, which catalyze breakdown of ß-lactam antibiotics, are a principal cause. Metallo ß-lactamases (MBLs) represent a particular challenge because they hydrolyze almost all ß-lactams and to date no MBL inhibitor has been approved for clinical use. Molecular simulations can aid drug discovery, for example, predicting inhibitor complexes, but empirical molecular mechanics (MM) methods often perform poorly for metalloproteins. Here we present a multiscale approach to model thiol inhibitor binding to IMP-1, a clinically important MBL containing two catalytic zinc ions, and predict the binding mode of a 2-mercaptomethyl thiazolidine (MMTZ) inhibitor. Inhibitors were first docked into the IMP-1 active site, testing different docking programs and scoring functions on multiple crystal structures. Complexes were then subjected to molecular dynamics (MD) simulations and subsequently refined through QM/MM optimization with a density functional theory (DFT) method, B3LYP/6-31G(d), increasing the accuracy of the method with successive steps. This workflow was tested on two IMP-1:MMTZ complexes, for which it reproduced crystallographically observed binding, and applied to predict the binding mode of a third MMTZ inhibitor for which a complex structure was crystallographically intractable. We also tested a 12-6-4 nonbonded interaction model in MD simulations and optimization with a SCC-DFTB QM/MM approach. The results show the limitations of empirical models for treating these systems and indicate the need for higher level calculations, for example, DFT/MM, for reliable structural predictions. This study demonstrates a reliable computational pipeline that can be applied to inhibitor design for MBLs and other zinc-metalloenzyme systems.


Asunto(s)
Antibacterianos/química , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química , beta-Lactamas/química , Dominio Catalítico , Modelos Moleculares , Zinc
5.
Org Biomol Chem ; 19(17): 3813-3819, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33606866

RESUMEN

The mcr-1 gene encodes a membrane-bound Zn2+-metalloenzyme, MCR-1, which catalyses phosphoethanolamine transfer onto bacterial lipid A, making bacteria resistant to colistin, a last-resort antibiotic. Mechanistic understanding of this process remains incomplete. Here, we investigate possible catalytic pathways using DFT and ab initio calculations on cluster models and identify a complete two-step reaction mechanism. The first step, formation of a covalent phosphointermediate via transfer of phosphoethanolamine from a membrane phospholipid donor to the acceptor Thr285, is rate-limiting and proceeds with a single Zn2+ ion. The second step, transfer of the phosphoethanolamine group to lipid A, requires an additional Zn2+. The calculations suggest the involvement of the Zn2+ orbitals directly in the reaction is limited, with the second Zn2+ acting to bind incoming lipid A and direct phosphoethanolamine addition. The new level of mechanistic detail obtained here, which distinguishes these enzymes from other phosphotransferases, will aid in the development of inhibitors specific to MCR-1 and related bacterial phosphoethanolamine transferases.


Asunto(s)
Farmacorresistencia Bacteriana
6.
J Chem Inf Model ; 61(12): 5988-5999, 2021 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-34637298

RESUMEN

Widespread bacterial resistance to carbapenem antibiotics is an increasing global health concern. Resistance has emerged due to carbapenem-hydrolyzing enzymes, including metallo-ß-lactamases (MßLs), but despite their prevalence and clinical importance, MßL mechanisms are still not fully understood. Carbapenem hydrolysis by MßLs can yield alternative product tautomers with the potential to access different binding modes. Here, we show that a combined approach employing crystallography and quantum mechanics/molecular mechanics (QM/MM) simulations allow tautomer assignment in MßL:hydrolyzed antibiotic complexes. Molecular simulations also examine (meta)stable species of alternative protonation and tautomeric states, providing mechanistic insights into ß-lactam hydrolysis. We report the crystal structure of the hydrolyzed carbapenem ertapenem bound to the L1 MßL from Stenotrophomonas maltophilia and model alternative tautomeric and protonation states of both hydrolyzed ertapenem and faropenem (a related penem antibiotic), which display different binding modes with L1. We show how the structures of both complexed ß-lactams are best described as the (2S)-imine tautomer with the carboxylate formed after ß-lactam ring cleavage deprotonated. Simulations show that enamine tautomer complexes are significantly less stable (e.g., showing partial loss of interactions with the L1 binuclear zinc center) and not consistent with experimental data. Strong interactions of Tyr32 and one zinc ion (Zn1) with ertapenem prevent a C6 group rotation, explaining the different binding modes of the two ß-lactams. Our findings establish the relative stability of different hydrolyzed (carba)penem forms in the L1 active site and identify interactions important to stable complex formation, information that should assist inhibitor design for this important antibiotic resistance determinant.


Asunto(s)
Antibacterianos , Carbapenémicos , Antibacterianos/química , Carbapenémicos/farmacología , Cristalografía por Rayos X , Iminas , Meropenem , beta-Lactamasas/química
7.
J Chem Inf Model ; 60(1): 226-234, 2020 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-31820972

RESUMEN

The enoyl-acyl carrier protein reductase InhA of Mycobacterium tuberculosis is an attractive, validated target for antituberculosis drug development. Moreover, direct inhibitors of InhA remain effective against InhA variants with mutations associated with isoniazid resistance, offering the potential for activity against MDR isolates. Here, structure-based virtual screening supported by biological assays was applied to identify novel InhA inhibitors as potential antituberculosis agents. High-speed Glide SP docking was initially performed against two conformations of InhA differing in the orientation of the active site Tyr158. The resulting hits were filtered for drug-likeness based on Lipinski's rule and avoidance of PAINS-like properties and finally subjected to Glide XP docking to improve accuracy. Sixteen compounds were identified and selected for in vitro biological assays, of which two (compounds 1 and 7) showed MIC of 12.5 and 25 µg/mL against M. tuberculosis H37Rv, respectively. Inhibition assays against purified recombinant InhA determined IC50 values for these compounds of 0.38 and 0.22 µM, respectively. A crystal structure of the most potent compound, compound 7, bound to InhA revealed the inhibitor to occupy a hydrophobic pocket implicated in binding the aliphatic portions of InhA substrates but distant from the NADH cofactor, i.e., in a site distinct from those occupied by the great majority of known InhA inhibitors. This compound provides an attractive starting template for ligand optimization aimed at discovery of new and effective compounds against M. tuberculosis that act by targeting InhA.


Asunto(s)
Antituberculosos/química , Proteínas Bacterianas/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Oxidorreductasas/antagonistas & inhibidores , Antituberculosos/farmacología , Sitios de Unión , Cristalografía por Rayos X , Descubrimiento de Drogas , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Estructura Molecular , Reproducibilidad de los Resultados , Relación Estructura-Actividad
8.
Artículo en Inglés | MEDLINE | ID: mdl-31383664

RESUMEN

ß-Lactamase production is the major ß-lactam resistance mechanism in Gram-negative bacteria. ß-Lactamase inhibitors (BLIs) efficacious against serine ß-lactamase (SBL) producers, especially strains carrying the widely disseminated class A enzymes, are required. Relebactam, a diazabicyclooctane (DBO) BLI, is in phase 3 clinical trials in combination with imipenem for the treatment of infections by multidrug-resistant Enterobacteriaceae We show that relebactam inhibits five clinically important class A SBLs (despite their differing spectra of activity), representing both chromosomal and plasmid-borne enzymes, i.e., the extended-spectrum ß-lactamases L2 (inhibition constant 3 µM) and CTX-M-15 (21 µM) and the carbapenemases KPC-2, -3, and -4 (1 to 5 µM). Against purified class A SBLs, relebactam is an inferior inhibitor compared with the clinically approved DBO avibactam (9- to 120-fold differences in half maximal inhibitory concentration [IC50]). MIC assays indicate relebactam potentiates ß-lactam (imipenem) activity against KPC-producing Klebsiella pneumoniae, with similar potency to avibactam (with ceftazidime). Relebactam is less effective than avibactam in combination with aztreonam against Stenotrophomonas maltophilia K279a. X-ray crystal structures of relebactam bound to CTX-M-15, L2, KPC-2, KPC-3, and KPC-4 reveal its C2-linked piperidine ring can sterically clash with Asn104 (CTX-M-15) or His/Trp105 (L2 and KPCs), rationalizing its poorer inhibition activity than that of avibactam, which has a smaller C2 carboxyamide group. Mass spectrometry and crystallographic data show slow, pH-dependent relebactam desulfation by KPC-2, -3, and -4. This comprehensive comparison of relebactam binding across five clinically important class A SBLs will inform the design of future DBOs, with the aim of improving clinical efficacy of BLI-ß-lactam combinations.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Stenotrophomonas maltophilia/efectos de los fármacos , Resistencia betalactámica/genética , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/química , Compuestos de Azabiciclo/química , Compuestos de Azabiciclo/metabolismo , Aztreonam/química , Aztreonam/metabolismo , Aztreonam/farmacología , Sitios de Unión , Ceftazidima/química , Ceftazidima/metabolismo , Ceftazidima/farmacología , Cromosomas Bacterianos/química , Cromosomas Bacterianos/enzimología , Ensayos Clínicos Fase III como Asunto , Clonación Molecular , Combinación de Medicamentos , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Imipenem/química , Imipenem/metabolismo , Imipenem/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Klebsiella pneumoniae/enzimología , Klebsiella pneumoniae/genética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Plásmidos/química , Plásmidos/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Stenotrophomonas maltophilia/enzimología , Stenotrophomonas maltophilia/genética , Inhibidores de beta-Lactamasas/química , Inhibidores de beta-Lactamasas/metabolismo , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
9.
Bioorg Med Chem Lett ; 29(15): 1981-1984, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31171422

RESUMEN

ß-Lactams are the most successful antibacterials, yet their use is threatened by resistance, importantly as caused by ß-lactamases. ß-Lactamases fall into two mechanistic groups: the serine ß-lactamases that utilise a covalent acyl-enzyme mechanism and the metallo ß-lactamases that utilise a zinc-bound water nucleophile. Achieving simultaneous inhibition of both ß-lactamase classes remains a challenge in the field. Vaborbactam is a boronate-based inhibitor that reacts with serine-ß-lactamases to form covalent complexes that mimic tetrahedral intermediates in catalysis. Vaborbactam has recently been approved for clinical use in combination with the carbapenem meropenem. Here we show that vaborbactam moderately inhibits metallo-ß-lactamases from all 3 subclasses (B1, B2 and B3), with a potency of around 20-100 fold below that by which it inhibits its current clinical targets, the Class A serine ß-lactamases. This result contrasts with recent investigations of bicyclic boronate inhibitors, which potently inhibit subclass B1 MBLs but which presently lack activity against B2 and B3 enzymes. These findings indicate that cyclic boronate scaffolds have the potential to inhibit the full range of ß-lactamases and justify further work on the development of boronates as broad-spectrum ß-lactamase inhibitors.


Asunto(s)
Antibacterianos/uso terapéutico , Ácidos Borónicos/uso terapéutico , beta-Lactamasas/uso terapéutico , Antibacterianos/farmacología , Ácidos Borónicos/farmacología , Humanos , beta-Lactamasas/farmacología
10.
Proc Natl Acad Sci U S A ; 113(26): E3745-54, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27303030

RESUMEN

Metallo-ß-lactamases (MBLs) hydrolyze almost all ß-lactam antibiotics and are unaffected by clinically available ß-lactamase inhibitors (ßLIs). Active-site architecture divides MBLs into three classes (B1, B2, and B3), complicating development of ßLIs effective against all enzymes. Bisthiazolidines (BTZs) are carboxylate-containing, bicyclic compounds, considered as penicillin analogs with an additional free thiol. Here, we show both l- and d-BTZ enantiomers are micromolar competitive ßLIs of all MBL classes in vitro, with Kis of 6-15 µM or 36-84 µM for subclass B1 MBLs (IMP-1 and BcII, respectively), and 10-12 µM for the B3 enzyme L1. Against the B2 MBL Sfh-I, the l-BTZ enantiomers exhibit 100-fold lower Kis (0.26-0.36 µM) than d-BTZs (26-29 µM). Importantly, cell-based time-kill assays show BTZs restore ß-lactam susceptibility of Escherichia coli-producing MBLs (IMP-1, Sfh-1, BcII, and GOB-18) and, significantly, an extensively drug-resistant Stenotrophomonas maltophilia clinical isolate expressing L1. BTZs therefore inhibit the full range of MBLs and potentiate ß-lactam activity against producer pathogens. X-ray crystal structures reveal insights into diverse BTZ binding modes, varying with orientation of the carboxylate and thiol moieties. BTZs bind the di-zinc centers of B1 (IMP-1; BcII) and B3 (L1) MBLs via the free thiol, but orient differently depending upon stereochemistry. In contrast, the l-BTZ carboxylate dominates interactions with the monozinc B2 MBL Sfh-I, with the thiol uninvolved. d-BTZ complexes most closely resemble ß-lactam binding to B1 MBLs, but feature an unprecedented disruption of the D120-zinc interaction. Cross-class MBL inhibition therefore arises from the unexpected versatility of BTZ binding.


Asunto(s)
Antibacterianos/química , Proteínas Bacterianas/química , Tiazolidinas/química , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química , Dominio Catalítico , Diseño de Fármacos , Hidrólisis , Cinética , Modelos Moleculares
11.
Biochemistry ; 57(12): 1880-1892, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29485857

RESUMEN

There are currently no clinically available inhibitors of metallo-ß-lactamases (MBLs), enzymes that hydrolyze ß-lactam antibiotics and confer resistance to Gram-negative bacteria. Here we present 6-phosphonomethylpyridine-2-carboxylates (PMPCs) as potent inhibitors of subclass B1 (IMP-1, VIM-2, and NDM-1) and B3 (L1) MBLs. Inhibition followed a competitive, slow-binding model without an isomerization step (IC50 values of 0.3-7.2 µM; Ki values of 0.03-1.5 µM). Minimum inhibitory concentration assays demonstrated potentiation of ß-lactam (Meropenem) activity against MBL-producing bacteria, including clinical isolates, at concentrations at which eukaryotic cells remain viable. Crystal structures revealed unprecedented modes of binding of inhibitor to B1 (IMP-1) and B3 (L1) MBLs. In IMP-1, binding does not replace the nucleophilic hydroxide, and the PMPC carboxylate and pyridine nitrogen interact closely (2.3 and 2.7 Å, respectively) with the Zn2 ion of the binuclear metal site. The phosphonate group makes limited interactions but is 2.6 Å from the nucleophilic hydroxide. Furthermore, the presence of a water molecule interacting with the PMPC phosphonate and pyridine N-C2 π-bond, as well as the nucleophilic hydroxide, suggests that the PMPC binds to the MBL active site as its hydrate. Binding is markedly different in L1, with the phosphonate displacing both Zn2, forming a monozinc enzyme, and the nucleophilic hydroxide, while also making multiple interactions with the protein main chain and Zn1. The carboxylate and pyridine nitrogen interact with Ser221 and -223, respectively (3 Å distance). The potency, low toxicity, cellular activity, and amenability to further modification of PMPCs indicate these and similar phosphonate compounds can be further considered for future MBL inhibitor development.


Asunto(s)
Bacterias/enzimología , Proteínas Bacterianas/química , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química
12.
Mol Microbiol ; 106(3): 492-504, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28876489

RESUMEN

Clavulanic acid and avibactam are clinically deployed serine ß-lactamase inhibitors, important as a defence against antibacterial resistance. Bicyclic boronates are recently discovered inhibitors of serine and some metallo ß-lactamases. Here, we show that avibactam and a bicyclic boronate inhibit L2 (serine ß-lactamase) but not L1 (metallo ß-lactamase) from the extensively drug resistant human pathogen Stenotrophomonas maltophilia. X-ray crystallography revealed that both inhibitors bind L2 by covalent attachment to the nucleophilic serine. Both inhibitors reverse ceftazidime resistance in S. maltophilia because, unlike clavulanic acid, they do not induce L1 production. Ceftazidime/inhibitor resistant mutants hyperproduce L1, but retain aztreonam/inhibitor susceptibility because aztreonam is not an L1 substrate. Importantly, avibactam, but not the bicyclic boronate is deactivated by L1 at a low rate; the utility of avibactam might be compromised by mutations that increase this deactivation rate. These data rationalize the observed clinical efficacy of ceftazidime/avibactam plus aztreonam as combination therapy for S. maltophilia infections and confirm that aztreonam-like ß-lactams plus nonclassical ß-lactamase inhibitors, particularly avibactam-like and bicyclic boronate compounds, have potential for treating infections caused by this most intractable of drug resistant pathogens.


Asunto(s)
Stenotrophomonas maltophilia/metabolismo , Inhibidores de beta-Lactamasas/metabolismo , Antibacterianos/farmacología , Compuestos de Azabiciclo/metabolismo , Compuestos de Azabiciclo/farmacología , Aztreonam , Proteínas Bacterianas/metabolismo , Ceftazidima , Cristalografía por Rayos X/métodos , Farmacorresistencia Bacteriana , Pruebas de Sensibilidad Microbiana , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
13.
Annu Rev Microbiol ; 67: 221-42, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23808339

RESUMEN

In bacteria such as Pseudomonas aeruginosa and Escherichia coli, tripartite membrane machineries, or pumps, determine the efflux of small noxious molecules, such as detergents, heavy metals, and antibiotics, and the export of large proteins including toxins. They are therefore influential in bacterial survival, particularly during infections caused by multidrug-resistant pathogens. In these tripartite pumps an inner membrane transporter, typically an ATPase or proton antiporter, binds and translocates export or efflux substrates. In cooperation with a periplasmic adaptor protein it recruits and opens a TolC family cell exit duct, which is anchored in the outer membrane and projects across the periplasmic space between inner and outer membranes. Assembled tripartite pumps thus span the entire bacterial cell envelope. We review the atomic structures of each of the three pump components and discuss how these have allowed high-resolution views of tripartite pump assembly, operation, and possible inhibition.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Escherichia coli/metabolismo , Bombas Iónicas/química , Bombas Iónicas/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/genética , Membrana Celular/química , Membrana Celular/genética , Membrana Celular/metabolismo , Escherichia coli/química , Escherichia coli/genética , Bombas Iónicas/genética , Modelos Moleculares , Periplasma/química , Periplasma/genética , Periplasma/metabolismo , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/genética
14.
Chemistry ; 24(22): 5734-5737, 2018 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-29250863

RESUMEN

The most important resistance mechanism to ß-lactam antibiotics involves hydrolysis by two ß-lactamase categories: the nucleophilic serine and the metallo-ß-lactamases (SBLs and MBLs, respectively). Cyclobutanones are hydrolytically stable ß-lactam analogues with potential to inhibit both SBLs and MBLs. We describe solution and crystallographic studies on the interaction of a cyclobutanone penem analogue with the clinically important MBL SPM-1. NMR experiments using 19 F-labeled SPM-1 imply the cyclobutanone binds to SPM-1 with micromolar affinity. A crystal structure of the SPM-1:cyclobutanone complex reveals binding of the hydrated cyclobutanone through interactions with one of the zinc ions, stabilisation of the hydrate by hydrogen bonding to zinc-bound water, and hydrophobic contacts with aromatic residues. NMR analyses using a 13 C-labeled cyclobutanone support assignment of the bound species as the hydrated ketone. The results inform on how MBLs bind substrates and stabilize tetrahedral intermediates. They support further investigations on the use of transition-state and/or intermediate analogues as inhibitors of all ß-lactamase classes.


Asunto(s)
Ciclobutanos/química , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo , beta-Lactamas/química , Catálisis , Meropenem , Imitación Molecular , Resonancia Magnética Nuclear Biomolecular , Tienamicinas/química , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química , beta-Lactamasas/clasificación
15.
Angew Chem Int Ed Engl ; 56(14): 3862-3866, 2017 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-28252254

RESUMEN

Resistance to ß-lactam antibiotics mediated by metallo-ß-lactamases (MBLs) is a growing problem. We describe the use of protein-observe 19 F-NMR (PrOF NMR) to study the dynamics of the São Paulo MBL (SPM-1) from ß-lactam-resistant Pseudomonas aeruginosa. Cysteinyl variants on the α3 and L3 regions, which flank the di-ZnII active site, were selectively 19 F-labeled using 3-bromo-1,1,1-trifluoroacetone. The PrOF NMR results reveal roles for the mobile α3 and L3 regions in the binding of both inhibitors and hydrolyzed ß-lactam products to SPM-1. These results have implications for the mechanisms and inhibition of MBLs by ß-lactams and non-ß-lactams and illustrate the utility of PrOF NMR for efficiently analyzing metal chelation, identifying new binding modes, and studying protein binding from a mixture of equilibrating isomers.


Asunto(s)
Imagen por Resonancia Magnética con Fluor-19 , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo , Sitios de Unión/efectos de los fármacos , Modelos Moleculares , Conformación Molecular , Inhibidores de beta-Lactamasas/síntesis química , Inhibidores de beta-Lactamasas/química
16.
Antimicrob Agents Chemother ; 60(10): 5828-40, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431213

RESUMEN

ß-Lactamase production increasingly threatens the effectiveness of ß-lactams, which remain a mainstay of antimicrobial chemotherapy. New activities emerge through both mutation of previously known ß-lactamases and mobilization from environmental reservoirs. The spread of metallo-ß-lactamases (MBLs) represents a particular challenge because of their typically broad-spectrum activities encompassing carbapenems, in addition to other ß-lactam classes. Increasingly, genomic and metagenomic studies have revealed the distribution of putative MBLs in the environment, but in most cases their activity against clinically relevant ß-lactams and, hence, the extent to which they can be considered a resistance reservoir remain uncharacterized. Here we characterize the product of one such gene, blaRm3, identified through functional metagenomic sampling of an environment with high levels of biocide exposure. blaRm3 encodes a subclass B3 MBL that, when expressed in a recombinant Escherichia coli strain, is exported to the bacterial periplasm and hydrolyzes clinically used penicillins, cephalosporins, and carbapenems with an efficiency limited by high Km values. An Rm3 crystal structure reveals the MBL superfamily αß/ßα fold, which more closely resembles that in mobilized B3 MBLs (AIM-1 and SMB-1) than other chromosomal enzymes (L1 or FEZ-1). A binuclear zinc site sits in a deep channel that is in part defined by a relatively extended N terminus. Structural comparisons suggest that the steric constraints imposed by the N terminus may limit its affinity for ß-lactams. Sequence comparisons identify Rm3-like MBLs in numerous other environmental samples and species. Our data suggest that Rm3-like enzymes represent a distinct group of B3 MBLs with a wide distribution and can be considered an environmental reservoir of determinants of ß-lactam resistance.


Asunto(s)
Metagenómica/métodos , beta-Lactamasas/química , beta-Lactamasas/genética , Cristalografía por Rayos X , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Filogenia , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , beta-Lactamasas/metabolismo
17.
Antimicrob Agents Chemother ; 60(7): 4170-5, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27139464

RESUMEN

Acetamido derivatives of the naturally antibacterial non-ß-lactam lactivicin (LTV) have improved activity against their penicillin binding protein targets and reduced hydrolysis by ß-lactamases, but penetration into Gram-negative bacteria is still relatively poor. Here we report that modification of the LTV lactone with a catechol-type siderophore increases potency 1,000-fold against Stenotrophomonas maltophilia, a species renowned for its insusceptibility to antimicrobials. The MIC90 of modified lactone compound 17 (LTV17) against a global collection of extensively drug-resistant clinical S. maltophilia isolates was 0.063 µg · ml(-1) Sideromimic modification does not reduce the ability of LTVs to induce production of the L1 and L2 ß-lactamases in S. maltophilia and does not reduce the rate at which LTVs are hydrolyzed by L1 or L2. We conclude, therefore, that lactivicin modification with a siderophore known to be preferentially used by S. maltophilia substantially increases penetration via siderophore uptake. LTV17 has the potential to be developed as a novel antimicrobial for treatment of infections by S. maltophilia More generally, our work shows that sideromimic modification in a species-targeted manner might prove useful for the development of narrow-spectrum antimicrobials that have reduced collateral effects.


Asunto(s)
Antibacterianos/farmacología , Péptidos/farmacología , Stenotrophomonas maltophilia/efectos de los fármacos , Antibacterianos/química , Farmacorresistencia Bacteriana , Pruebas de Sensibilidad Microbiana , Péptidos/química , Péptidos Cíclicos , Relación Estructura-Actividad
18.
Proc Natl Acad Sci U S A ; 108(5): 2112-7, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245342

RESUMEN

In bacterial drug resistance and virulence pumps, an inner membrane (IM) transporter and periplasmic adaptor recruit an outer membrane (OM) trimeric TolC exit duct that projects an α-helical tunnel across the periplasm. The TolC periplasmic entrance is closed by densely packed α-helical coiled coils, inner H7/H8, and outer H3/H4, constrained by a hydrogen bond network. On recruitment, these coiled coils must undergo transition to the open state. We present 2.9 Å resolution crystal structures of two sequential TolC open states in which the network is incrementally disrupted and channel conductances defined in lipid bilayers. Superimposition of TolC(RS) (370 pS) and TolC(YFRS) (1,000 pS) on the TolC(WT) closed state (80 pS) showed that in the initial open-state TolC(RS), relaxation already causes approximately 14° twisting and expansion of helix H7 at the periplasmic tip, increasing interprotomer distances from 12.2 Å in TolC(WT) to 18.9 Å. However, in the crystal structure, the weakened Asp(374) pore constriction was maintained at the closed state 11.3 Å(2). In the advanced open-state TolC(YFRS), there was little further expansion at the tip, to interprotomer 21.3 Å, but substantial movement of inner and outer coiled coils dilated the pore constriction. In particular, upon abolition of the TolC(YFRS) intraprotomer Tyr(362)-Asp(153) link, a redirection of Tyr(362) and "bulge" in H3 allowed a simple movement outward of H8, establishing a 50.3 Å(2) opening. Root mean square deviations (rmsds) over the coiled coils of the three protomers of TolC(RS) and TolC(YFRS) illustrate that, whereas independent movement at the periplasmic tips may feature in the initial stages of opening, full dilation of the pore constriction is entirely symmetrical.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/fisiología , Proteínas de Escherichia coli/fisiología , Proteínas de Transporte de Membrana/fisiología , Proteínas de la Membrana Bacteriana Externa/química , Transporte Biológico , Escherichia coli/fisiología , Proteínas de Escherichia coli/química , Membrana Dobles de Lípidos , Proteínas de Transporte de Membrana/química
19.
J Med Chem ; 67(5): 3795-3812, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38373290

RESUMEN

Antimicrobial resistance is a global public health threat. Metallo-ß-lactamases (MBLs) inactivate ß-lactam antibiotics, including carbapenems, are disseminating among Gram-negative bacteria, and lack clinically useful inhibitors. The evolving bisthiazolidine (BTZ) scaffold inhibits all three MBL subclasses (B1-B3). We report design, synthesis, and evaluation of BTZ analogues. Structure-activity relationships identified the BTZ thiol as essential, while carboxylate is replaceable, with its removal enhancing potency by facilitating hydrophobic interactions within the MBL active site. While the introduction of a flexible aromatic ring is neutral or detrimental for inhibition, a rigid (fused) ring generated nM benzobisheterocycle (BBH) inhibitors that potentiated carbapenems against MBL-producing strains. Crystallography of BBH:MBL complexes identified hydrophobic interactions as the basis of potency toward B1 MBLs. These data underscore BTZs as versatile, potent broad-spectrum MBL inhibitors (with activity extending to enzymes refractory to other inhibitors) and provide a rational approach to further improve the tricyclic BBH scaffold.


Asunto(s)
Antibacterianos , Inhibidores de beta-Lactamasas , Inhibidores de beta-Lactamasas/farmacología , Inhibidores de beta-Lactamasas/química , Antibacterianos/farmacología , Antibacterianos/química , beta-Lactamasas/química , Carbapenémicos , Bacterias Gramnegativas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA