Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
PLoS Pathog ; 20(6): e1012235, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38843111

RESUMEN

Amikacin and piperacillin/tazobactam are frequent antibiotic choices to treat bloodstream infection, which is commonly fatal and most often caused by bacteria from the family Enterobacterales. Here we show that two gene cassettes located side-by-side in and ancestral integron similar to In37 have been "harvested" by insertion sequence IS26 as a transposon that is widely disseminated among the Enterobacterales. This transposon encodes the enzymes AAC(6')-Ib-cr and OXA-1, reported, respectively, as amikacin and piperacillin/tazobactam resistance mechanisms. However, by studying bloodstream infection isolates from 769 patients from three hospitals serving a population of 1.2 million people in South West England, we show that increased enzyme production due to mutation in an IS26/In37-derived hybrid promoter or, more commonly, increased transposon copy number is required to simultaneously remove these two key therapeutic options; in many cases leaving only the last-resort antibiotic, meropenem. These findings may help improve the accuracy of predicting piperacillin/tazobactam treatment failure, allowing stratification of patients to receive meropenem or piperacillin/tazobactam, which may improve outcome and slow the emergence of meropenem resistance.


Asunto(s)
Antibacterianos , Elementos Transponibles de ADN , Humanos , Antibacterianos/farmacología , Elementos Transponibles de ADN/genética , Farmacorresistencia Bacteriana Múltiple/genética , Piperacilina/farmacología , Amicacina/farmacología , Pruebas de Sensibilidad Microbiana , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/tratamiento farmacológico , Infecciones por Enterobacteriaceae/genética , Enterobacteriaceae/genética , Enterobacteriaceae/efectos de los fármacos , Integrones/genética , Bacteriemia/microbiología , Bacteriemia/tratamiento farmacológico , Bacteriemia/genética
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.
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
5.
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
6.
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
7.
Angew Chem Int Ed Engl ; 58(7): 1990-1994, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30569575

RESUMEN

Enzymes often use nucleophilic serine, threonine, and cysteine residues to achieve the same type of reaction; the underlying reasons for this are not understood. While bacterial d,d-transpeptidases (penicillin-binding proteins) employ a nucleophilic serine, l,d-transpeptidases use a nucleophilic cysteine. The covalent complexes formed by l,d-transpeptidases with some ß-lactam antibiotics undergo non-hydrolytic fragmentation. This is not usually observed for penicillin-binding proteins, or for the related serine ß-lactamases. Replacement of the nucleophilic serine of serine ß-lactamases with cysteine yields enzymes which fragment ß-lactams via a similar mechanism as the l,d-transpeptidases, implying the different reaction outcomes are principally due to the formation of thioester versus ester intermediates. The results highlight fundamental differences in the reactivity of nucleophilic serine and cysteine enzymes, and imply new possibilities for the inhibition of nucleophilic enzymes.


Asunto(s)
Antibacterianos/metabolismo , Cisteína/metabolismo , Peptidil Transferasas/metabolismo , beta-Lactamasas/metabolismo , beta-Lactamas/metabolismo , Antibacterianos/química , Cisteína/química , Conformación Molecular , Peptidil Transferasas/química , beta-Lactamasas/química , beta-Lactamas/química
8.
Angew Chem Int Ed Engl ; 57(5): 1282-1285, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29236332

RESUMEN

ß-Lactamases threaten the clinical use of carbapenems, which are considered antibiotics of last resort. The classical mechanism of serine carbapenemase catalysis proceeds through hydrolysis of an acyl-enzyme intermediate. We show that class D ß-lactamases also degrade clinically used 1ß-methyl-substituted carbapenems through the unprecedented formation of a carbapenem-derived ß-lactone. ß-Lactone formation results from nucleophilic attack of the carbapenem hydroxyethyl side chain on the ester carbonyl of the acyl-enzyme intermediate. The carbapenem-derived lactone products inhibit both serine ß-lactamases (particularly class D) and metallo-ß-lactamases. These results define a new mechanism for the class D carbapenemases, in which a hydrolytic water molecule is not required.

9.
Antibiotics (Basel) ; 11(3)2022 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-35326858

RESUMEN

Carbapenems are important antibacterials and are both substrates and inhibitors of some ß-lactamases. We report studies on the reaction of the unusual carbapenem biapenem, with the subclass B1 metallo-ß-lactamases VIM-1 and VIM-2 and the class A serine-ß-lactamase KPC-2. X-ray diffraction studies with VIM-2 crystals treated with biapenem reveal the opening of the ß-lactam ring to form a mixture of the (2S)-imine and enamine complexed at the active site. NMR studies on the reactions of biapenem with VIM-1, VIM-2, and KPC-2 reveal the formation of hydrolysed enamine and (2R)- and (2S)-imine products. The combined results support the proposal that SBL/MBL-mediated carbapenem hydrolysis results in a mixture of tautomerizing enamine and (2R)- and (2S)-imine products, with the thermodynamically favoured (2S)-imine being the major observed species over a relatively long-time scale. The results suggest that prolonging the lifetimes of ß-lactamase carbapenem complexes by optimising tautomerisation of the nascently formed enamine to the (2R)-imine and likely more stable (2S)-imine tautomer is of interest in developing improved carbapenems.

10.
mBio ; 13(3): e0179321, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35612361

RESUMEN

ß-Lactamases hydrolyze ß-lactam antibiotics and are major determinants of antibiotic resistance in Gram-negative pathogens. Enmetazobactam (formerly AAI101) and tazobactam are penicillanic acid sulfone (PAS) ß-lactamase inhibitors that differ by an additional methyl group on the triazole ring of enmetazobactam, rendering it zwitterionic. In this study, ultrahigh-resolution X-ray crystal structures and mass spectrometry revealed the mechanism of PAS inhibition of CTX-M-15, an extended-spectrum ß-lactamase (ESBL) globally disseminated among Enterobacterales. CTX-M-15 crystals grown in the presence of enmetazobactam or tazobactam revealed loss of the Ser70 hydroxyl group and formation of a lysinoalanine cross-link between Lys73 and Ser70, two residues critical for catalysis. Moreover, the residue at position 70 undergoes epimerization, resulting in formation of a d-amino acid. Cocrystallization of enmetazobactam or tazobactam with CTX-M-15 with a Glu166Gln mutant revealed the same cross-link, indicating that this modification is not dependent on Glu166-catalyzed deacylation of the PAS-acylenzyme. A cocrystal structure of enmetazobactam with CTX-M-15 with a Lys73Ala mutation indicates that epimerization can occur without cross-link formation and positions the Ser70 Cß closer to Lys73, likely facilitating formation of the Ser70-Lys73 cross-link. A crystal structure of a tazobactam-derived imine intermediate covalently linked to Ser70, obtained after 30 min of exposure of CTX-M-15 crystals to tazobactam, supports formation of an initial acylenzyme by PAS inhibitors on reaction with CTX-M-15. These data rationalize earlier results showing CTX-M-15 deactivation by PAS inhibitors to involve loss of protein mass, and they identify a distinct mechanism of ß-lactamase inhibition by these agents. IMPORTANCE ß-Lactams are the most prescribed antibiotic class for treating bacterial diseases, but their continued efficacy is threatened by bacterial strains producing ß-lactamase enzymes that catalyze their inactivation. The CTX-M family of ESBLs are major contributors to ß-lactam resistance in Enterobacterales, preventing effective treatment with most penicillins and cephalosporins. Combining ß-lactams with ß-lactamase inhibitors (BLIs) is a validated route to overcome such resistance. Here, we describe how exposure to enmetazobactam and tazobactam, BLIs based on a penicillanic acid sulfone (PAS) scaffold, leads to a protein modification in CTX-M-15, resulting in irremediable inactivation of this most commonly encountered member of the CTX-M family. High-resolution X-ray crystal structures showed that PAS exposure induces formation of a cross-link between Ser70 and Lys73, two residues critical to ß-lactamase function. This previously undescribed mechanism of inhibition furthers our understanding of ß-lactamase inhibition by classical PAS inhibitors and provides a basis for further, rational inhibitor development.


Asunto(s)
Sulbactam , Inhibidores de beta-Lactamasas , Antibacterianos/farmacología , Lisina , Pruebas de Sensibilidad Microbiana , Serina , Sulbactam/farmacología , Tazobactam/farmacología , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/metabolismo
11.
Eur J Med Chem ; 215: 113257, 2021 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-33618159

RESUMEN

Penems have demonstrated potential as antibacterials and ß-lactamase inhibitors; however, their clinical use has been limited, especially in comparison with the structurally related carbapenems. Faropenem is an orally active antibiotic with a C-2 tetrahydrofuran (THF) ring, which is resistant to hydrolysis by some ß-lactamases. We report studies on the reactions of faropenem with carbapenem-hydrolysing ß-lactamases, focusing on the class A serine ß-lactamase KPC-2 and the metallo ß-lactamases (MBLs) VIM-2 (a subclass B1 MBL) and L1 (a B3 MBL). Kinetic studies show that faropenem is a substrate for all three ß-lactamases, though it is less efficiently hydrolysed by KPC-2. Crystallographic analyses on faropenem-derived complexes reveal opening of the ß-lactam ring with formation of an imine with KPC-2, VIM-2, and L1. In the cases of the KPC-2 and VIM-2 structures, the THF ring is opened to give an alkene, but with L1 the THF ring remains intact. Solution state studies, employing NMR, were performed on L1, KPC-2, VIM-2, VIM-1, NDM-1, OXA-23, OXA-10, and OXA-48. The solution results reveal, in all cases, formation of imine products in which the THF ring is opened; formation of a THF ring-closed imine product was only observed with VIM-1 and VIM-2. An enamine product with a closed THF ring was also observed in all cases, at varying levels. Combined with previous reports, the results exemplify the potential for different outcomes in the reactions of penems with MBLs and SBLs and imply further structure-activity relationship studies are worthwhile to optimise the interactions of penems with ß-lactamases. They also exemplify how crystal structures of ß-lactamase substrate/inhibitor complexes do not always reflect reaction outcomes in solution.


Asunto(s)
Antibacterianos/química , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química , beta-Lactamas/química , Antibacterianos/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Hidrólisis , Klebsiella pneumoniae/enzimología , Unión Proteica , Pseudomonas aeruginosa/enzimología , Stenotrophomonas maltophilia/enzimología , Inhibidores de beta-Lactamasas/metabolismo , beta-Lactamasas/metabolismo , beta-Lactamas/metabolismo
12.
Elife ; 102021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33755013

RESUMEN

Understanding allostery in enzymes and tools to identify it offer promising alternative strategies to inhibitor development. Through a combination of equilibrium and nonequilibrium molecular dynamics simulations, we identify allosteric effects and communication pathways in two prototypical class A ß-lactamases, TEM-1 and KPC-2, which are important determinants of antibiotic resistance. The nonequilibrium simulations reveal pathways of communication operating over distances of 30 Å or more. Propagation of the signal occurs through cooperative coupling of loop dynamics. Notably, 50% or more of clinically relevant amino acid substitutions map onto the identified signal transduction pathways. This suggests that clinically important variation may affect, or be driven by, differences in allosteric behavior, providing a mechanism by which amino acid substitutions may affect the relationship between spectrum of activity, catalytic turnover, and potential allosteric behavior in this clinically important enzyme family. Simulations of the type presented here will help in identifying and analyzing such differences.


Antibiotics are crucial drugs for treating and preventing bacterial infections, but some bacteria are evolving ways to resist their effects. This 'antibiotic resistance' threatens lives and livelihoods worldwide. ß-lactam antibiotics, like penicillin, are some of the most commonly used, but some bacteria can now make enzymes called ß-lactamases, which destroy these antibiotics. Dozens of different types of ß-lactamases now exist, each with different properties. Two of the most medically important are TEM-1 and KPC-2. One way to counteract ß-lactamases is with drugs called inhibitors that stop the activity of these enzymes. The approved ß-lactamase inhibitors work by blocking the part of the enzyme that binds and destroys antibiotics, known as the 'active site'. The ß-lactamases have evolved, some of which have the ability to resist the effects of known inhibitors. It is possible that targeting parts of ß-lactamases far from the active site, known as 'allosteric sites', might get around these new bacterial defences. A molecule that binds to an allosteric site might alter the enzyme's shape, or restrict its movement, making it unable to do its job. Galdadas, Qu et al. used simulations to understand how molecules binding at allosteric sites affect enzyme movement. The experiments examined the structures of both TEM-1 and KPC-2, looking at how their shapes changed as molecules were removed from the allosteric site. This revealed how the allosteric sites and the active site are linked together. When molecules were taken out of the allosteric sites, they triggered ripples of shape change that travelled via loop-like structures across the surface of the enzyme. These loops contain over half of the known differences between the different types of ß-lactamases, suggesting mutations here may be responsible for changing which antibiotics each enzyme can destroy. In other words, changes in the 'ripples' may be related to the ability of the enzymes to resist particular antibiotics. Understanding how changes in one part of a ß-lactamase enzyme reach the active site could help in the design of new inhibitors. It might also help to explain how ß-lactamases evolve new properties. Further work could show why different enzymes are more or less active against different antibiotics.


Asunto(s)
Farmacorresistencia Bacteriana , Simulación de Dinámica Molecular , beta-Lactamasas/química , Sustitución de Aminoácidos , Conformación Proteica
13.
Nat Commun ; 12(1): 4461, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294694

RESUMEN

Serial femtosecond crystallography has opened up many new opportunities in structural biology. In recent years, several approaches employing light-inducible systems have emerged to enable time-resolved experiments that reveal protein dynamics at high atomic and temporal resolutions. However, very few enzymes are light-dependent, whereas macromolecules requiring ligand diffusion into an active site are ubiquitous. In this work we present a drop-on-drop sample delivery system that enables the study of enzyme-catalyzed reactions in microcrystal slurries. The system delivers ligand solutions in bursts of multiple picoliter-sized drops on top of a larger crystal-containing drop inducing turbulent mixing and transports the mixture to the X-ray interaction region with temporal resolution. We demonstrate mixing using fluorescent dyes, numerical simulations and time-resolved serial femtosecond crystallography, which show rapid ligand diffusion through microdroplets. The drop-on-drop method has the potential to be widely applicable to serial crystallography studies, particularly of enzyme reactions with small molecule substrates.


Asunto(s)
Cristalografía por Rayos X/métodos , Enzimas/química , Enzimas/metabolismo , Animales , Proteínas Aviares/química , Proteínas Aviares/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Biocatálisis , Dominio Catalítico , Pollos , Cristalografía por Rayos X/instrumentación , Diseño de Equipo , Modelos Moleculares , Muramidasa/química , Muramidasa/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , beta-Lactamasas/química , beta-Lactamasas/metabolismo
14.
RSC Med Chem ; 11(4): 491-496, 2020 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33479650

RESUMEN

Klebsiella pneumoniae carbapenemase-2 (KPC-2) is a serine-ß-lactamase (SBL) capable of hydrolysing almost all ß-lactam antibiotics. We compare KPC-2 inhibition by vaborbactam, a clinically-approved monocyclic boronate, and VNRX-5133 (taniborbactam), a bicyclic boronate in late-stage clinical development. Vaborbactam inhibition is slowly reversible, whereas taniborbactam has an off-rate indicating essentially irreversible complex formation and a 15-fold higher on-rate, although both potentiate ß-lactam activity against KPC-2-expressing K. pneumoniae. High resolution X-ray crystal structures reveal closely related binding modes for both inhibitors to KPC-2, with differences apparent only in positioning of the endocyclic boronate ester oxygen. The results indicate the bicyclic boronate scaffold as both an efficient, long-lasting, KPC-2 inhibitor and capable of supporting further iterations that may improve potency against specific enzyme targets and pre-empt the emergence of inhibitor resistant KPC-2 variants.

15.
J Mol Biol ; 431(18): 3472-3500, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30959050

RESUMEN

The ß-lactams retain a central place in the antibacterial armamentarium. In Gram-negative bacteria, ß-lactamase enzymes that hydrolyze the amide bond of the four-membered ß-lactam ring are the primary resistance mechanism, with multiple enzymes disseminating on mobile genetic elements across opportunistic pathogens such as Enterobacteriaceae (e.g., Escherichia coli) and non-fermenting organisms (e.g., Pseudomonas aeruginosa). ß-Lactamases divide into four classes; the active-site serine ß-lactamases (classes A, C and D) and the zinc-dependent or metallo-ß-lactamases (MBLs; class B). Here we review recent advances in mechanistic understanding of each class, focusing upon how growing numbers of crystal structures, in particular for ß-lactam complexes, and methods such as neutron diffraction and molecular simulations, have improved understanding of the biochemistry of ß-lactam breakdown. A second focus is ß-lactamase interactions with carbapenems, as carbapenem-resistant bacteria are of grave clinical concern and carbapenem-hydrolyzing enzymes such as KPC (class A) NDM (class B) and OXA-48 (class D) are proliferating worldwide. An overview is provided of the changing landscape of ß-lactamase inhibitors, exemplified by the introduction to the clinic of combinations of ß-lactams with diazabicyclooctanone and cyclic boronate serine ß-lactamase inhibitors, and of progress and strategies toward clinically useful MBL inhibitors. Despite the long history of ß-lactamase research, we contend that issues including continuing unresolved questions around mechanism; opportunities afforded by new technologies such as serial femtosecond crystallography; the need for new inhibitors, particularly for MBLs; the likely impact of new ß-lactam:inhibitor combinations and the continuing clinical importance of ß-lactams mean that this remains a rewarding research area.


Asunto(s)
Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/efectos de los fármacos , beta-Lactamasas/metabolismo , beta-Lactamas/metabolismo , beta-Lactamas/farmacología , Antibacterianos/farmacología , Enterobacteriaceae Resistentes a los Carbapenémicos , Carbapenémicos/química , Carbapenémicos/metabolismo , Dominio Catalítico , Combinación de Medicamentos , Farmacorresistencia Bacteriana/genética , Enterobacteriaceae/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/enzimología , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/metabolismo , Humanos , Secuencias Repetitivas Esparcidas , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química , beta-Lactamasas/clasificación , beta-Lactamas/química
16.
Sci Rep ; 9(1): 13608, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31541180

RESUMEN

ß-Lactamases are a major threat to the clinical use of carbapenems, which are often antibiotics of last resort. Despite this, the reaction outcomes and mechanisms by which ß-lactamases degrade carbapenems are still not fully understood. The carbapenem bicyclic core consists of a ß-lactam ring fused to a pyrroline ring. Following ß-lactamase-mediated opening of the ß-lactam, the pyrroline may interconvert between an enamine (2-pyrroline) form and two epimeric imine (1-pyrroline) forms; previous crystallographic and spectroscopic studies have reported all three of these forms in the contexts of hydrolysis by different ß-lactamases. As we show by NMR spectroscopy, the serine ß-lactamases (KPC-2, SFC-1, CMY-10, OXA-23, and OXA-48) and metallo-ß-lactamases (NDM-1, VIM-1, BcII, CphA, and L1) tested all degrade carbapenems to preferentially give the Δ2 (enamine) and/or (R)-Δ1 (imine) products. Rapid non-enzymatic tautomerisation of the Δ2 product to the (R)-Δ1 product prevents assignment of the nascent enzymatic product by NMR. The observed stereoselectivity implies that carbapenemases control the form of their pyrroline ring intermediate(s)/product(s), thereby preventing pyrroline tautomerisation from inhibiting catalysis.

17.
J Med Chem ; 62(9): 4411-4425, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-31009558

RESUMEN

Expression of ß-lactamase is the single most prevalent determinant of antibiotic resistance, rendering bacteria resistant to ß-lactam antibiotics. In this article, we describe the development of an antibiotic prodrug that combines ciprofloxacin with a ß-lactamase-cleavable motif. The prodrug is only bactericidal after activation by ß-lactamase. Bactericidal activity comparable to ciprofloxacin is demonstrated against clinically relevant E. coli isolates expressing diverse ß-lactamases; bactericidal activity was not observed in strains without ß-lactamase. These findings demonstrate that it is possible to exploit antibiotic resistance to selectively target ß-lactamase-producing bacteria using our prodrug approach, without adversely affecting bacteria that do not produce ß-lactamase. This paves the way for selective targeting of drug-resistant pathogens without disrupting or selecting for resistance within the microbiota, reducing the rate of secondary infections and subsequent antibiotic use.


Asunto(s)
Antibacterianos/farmacología , Cefalosporinas/farmacología , Ciprofloxacina/análogos & derivados , Ciprofloxacina/farmacología , Profármacos/farmacología , beta-Lactamasas/metabolismo , Antibacterianos/síntesis química , Antibacterianos/metabolismo , Cefalosporinas/síntesis química , Cefalosporinas/metabolismo , Ciprofloxacina/metabolismo , Farmacorresistencia Microbiana/fisiología , Escherichia coli/efectos de los fármacos , Hidrólisis , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Profármacos/síntesis química , Profármacos/metabolismo , Relación Estructura-Actividad , Inhibidores de Topoisomerasa II/síntesis química , Inhibidores de Topoisomerasa II/metabolismo , Inhibidores de Topoisomerasa II/farmacología
18.
Sci Rep ; 7: 39392, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28059088

RESUMEN

The polymixin colistin is a "last line" antibiotic against extensively-resistant Gram-negative bacteria. Recently, the mcr-1 gene was identified as a plasmid-mediated resistance mechanism in human and animal Enterobacteriaceae, with a wide geographical distribution and many producer strains resistant to multiple other antibiotics. mcr-1 encodes a membrane-bound enzyme catalysing phosphoethanolamine transfer onto bacterial lipid A. Here we present crystal structures revealing the MCR-1 periplasmic, catalytic domain to be a zinc metalloprotein with an alkaline phosphatase/sulphatase fold containing three disulphide bonds. One structure captures a phosphorylated form representing the first intermediate in the transfer reaction. Mutation of residues implicated in zinc or phosphoethanolamine binding, or catalytic activity, restores colistin susceptibility of recombinant E. coli. Zinc deprivation reduces colistin MICs in MCR-1-producing laboratory, environmental, animal and human E. coli. Conversely, over-expression of the disulphide isomerase DsbA increases the colistin MIC of laboratory E. coli. Preliminary density functional theory calculations on cluster models suggest a single zinc ion may be sufficient to support phosphoethanolamine transfer. These data demonstrate the importance of zinc and disulphide bonds to MCR-1 activity, suggest that assays under zinc-limiting conditions represent a route to phenotypic identification of MCR-1 producing E. coli, and identify key features of the likely catalytic mechanism.


Asunto(s)
Antibacterianos/farmacología , Colistina/farmacología , Farmacorresistencia Bacteriana , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Análisis Mutacional de ADN , Disulfuros/metabolismo , Escherichia coli/efectos de los fármacos , Proteínas de Escherichia coli/genética , Metaloproteínas/química , Metaloproteínas/genética , Metaloproteínas/metabolismo , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación Proteica , Proteína Disulfuro Isomerasas/metabolismo , Zinc/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA