Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 233
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Antimicrob Agents Chemother ; 68(3): e0110823, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38259088

RESUMEN

Klebsiella pneumoniae carbapenemase (KPC) variants have been described that confer resistance to both ceftazidime-avibactam and cefiderocol. Of these, KPC-33 and KPC-31 are D179Y-containing variants derived from KPC-2 and KPC-3, respectively. To better understand this atypical phenotype, the catalytic mechanism of ceftazidime and cefiderocol hydrolysis by KPC-33 and KPC-31 as well as the ancestral KPC-2 and KPC-3 enzymes was studied. Steady-state kinetics showed that the D179Y substitution in either KPC-2 or KPC-3 is associated with a large decrease in both kcat and KM such that kcat/KM values were largely unchanged for both ceftazidime and cefiderocol substrates. A decrease in both kcat and KM is consistent with a decreased and rate-limiting deacylation step. We explored this hypothesis by performing pre-steady-state kinetics and showed that the acylation step is rate-limiting for KPC-2 and KPC-3 for both ceftazidime and cefiderocol hydrolysis. In contrast, we observed a burst of acyl-enzyme formation followed by a slow steady-state rate for the D179Y variants of KPC-2 and KPC-3 with either ceftazidime or cefiderocol, indicating that deacylation of the covalent intermediate is the rate-limiting step for catalysis. Finally, we show that the low KM value for ceftazidime or cefiderocol hydrolysis of the D179Y variants is not an indication of tight binding affinity for the substrates but rather is a reflection of the deacylation reaction becoming rate-limiting. Thus, the hydrolysis mechanism of ceftazidime and cefiderocol by the D179Y variants is very similar and involves the formation of a long-lived covalent intermediate that is associated with resistance to the drugs.


Asunto(s)
Antibacterianos , Ceftazidima , Ceftazidima/farmacología , Ceftazidima/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Cefiderocol , Klebsiella pneumoniae , Hidrólisis , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Combinación de Medicamentos , Compuestos de Azabiciclo/farmacología , Pruebas de Sensibilidad Microbiana
2.
Angew Chem Int Ed Engl ; 63(12): e202317315, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38227422

RESUMEN

The amino acid substitutions in Klebsiella pneumoniae carbapenemase 2 (KPC-2) that have arisen in the clinic are observed to lead to the development of resistance to ceftazidime-avibactam, a preferred treatment for KPC bearing Gram-negative bacteria. Specific substitutions in the omega loop (R164-D179) result in changes in the structure and function of the enzyme, leading to alterations in substrate specificity, decreased stability, and more recently observed, increased resistance to ceftazidime/avibactam. Using accelerated rare-event sampling well-tempered metadynamics simulations, we explored in detail the structural role of R164 and D179 variants that are described to confer ceftazidime/avibactam resistance. The buried conformation of D179 substitutions produce a pronounced structural disorder in the omega loop - more than R164 mutants, where the crystallographic omega loop structure remains mostly intact. Our findings also reveal that the conformation of N170 plays an underappreciated role impacting drug binding and restricting deacylation. The results further support the hypothesis that KPC-2 D179 variants employ substrate-assisted catalysis for ceftazidime hydrolysis, involving the ring amine of the aminothiazole group to promote deacylation and catalytic turnover. Moreover, the shift in the WT conformation of N170 contributes to reduced deacylation and an altered spectrum of enzymatic activity.


Asunto(s)
Antibacterianos , Ceftazidima , Ceftazidima/química , Ceftazidima/metabolismo , Antibacterianos/química , beta-Lactamasas/metabolismo , Proteínas Bacterianas/metabolismo , Compuestos de Azabiciclo , Sustitución de Aminoácidos , Pruebas de Sensibilidad Microbiana , Inhibidores de beta-Lactamasas
3.
Antimicrob Agents Chemother ; 67(6): e0160322, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37199612

RESUMEN

The ß-lactam antibiotics have been successfully used for decades to combat susceptible Pseudomonas aeruginosa, which has a notoriously difficult to penetrate outer membrane (OM). However, there is a dearth of data on target site penetration and covalent binding of penicillin-binding proteins (PBP) for ß-lactams and ß-lactamase inhibitors in intact bacteria. We aimed to determine the time course of PBP binding in intact and lysed cells and estimate the target site penetration and PBP access for 15 compounds in P. aeruginosa PAO1. All ß-lactams (at 2 × MIC) considerably bound PBPs 1 to 4 in lysed bacteria. However, PBP binding in intact bacteria was substantially attenuated for slow but not for rapid penetrating ß-lactams. Imipenem yielded 1.5 ± 0.11 log10 killing at 1h compared to <0.5 log10 killing for all other drugs. Relative to imipenem, the rate of net influx and PBP access was ~ 2-fold slower for doripenem and meropenem, 7.6-fold for avibactam, 14-fold for ceftazidime, 45-fold for cefepime, 50-fold for sulbactam, 72-fold for ertapenem, ~ 249-fold for piperacillin and aztreonam, 358-fold for tazobactam, ~547-fold for carbenicillin and ticarcillin, and 1,019-fold for cefoxitin. At 2 × MIC, the extent of PBP5/6 binding was highly correlated (r2 = 0.96) with the rate of net influx and PBP access, suggesting that PBP5/6 acted as a decoy target that should be avoided by slowly penetrating, future ß-lactams. This first comprehensive assessment of the time course of PBP binding in intact and lysed P. aeruginosa explained why only imipenem killed rapidly. The developed novel covalent binding assay in intact bacteria accounts for all expressed resistance mechanisms.


Asunto(s)
Antibacterianos , Pseudomonas aeruginosa , Proteínas de Unión a las Penicilinas/genética , Proteínas de Unión a las Penicilinas/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas Bacterianas/metabolismo , Farmacología en Red , Pruebas de Sensibilidad Microbiana , beta-Lactamas/farmacología , beta-Lactamas/metabolismo , Imipenem/farmacología , Imipenem/metabolismo , Ceftazidima/metabolismo , beta-Lactamasas/metabolismo
4.
Int Arch Allergy Immunol ; 184(9): 932-948, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37321197

RESUMEN

INTRODUCTION: Neutrophilic granulocytes represent the first line of defense against microorganisms. Granulocytes phagocytose microorganisms and specifically synthesize oxygen radicals against them, which eventually kills the invaders. METHODS: Neutrophilic granulocytes were isolated from peripheral blood of healthy volunteer donors. Putative interference of new-generation antibiotics with neutrophil function was tested using a collection of granulocyte-stimulating agents and Amplex™ Red-based plate assay and flow cytometry-based respiratory burst assays. In addition, phagocytosis of E. coli, IL-8 production, bactericidal activity, and CD62L expression of granulocytes were evaluated. RESULTS: Of note, we found that the two glycopeptide antibiotics dalbavancin and teicoplanin inhibited ROS production upon granulocyte activation via different signaling pathways in a dose-dependent manner. Dalbavancin also blocked the PMA-induced shedding of CD62L. In contrast, the oxazolidinone antibiotics tedizolid and linezolid had no effect on neutrophil function, while the combination of ceftazidime/avibactam dose dependently inhibited the fMLP/Cytochalasin B-induced granulocyte burst in a dose-dependent manner. Additionally, we showed that dalbavancin and teicoplanin as well as sulfametrole/trimethoprim and ceftazidime/avibactam inhibited baseline and PMA-induced IL-8 production by neutrophilic granulocytes. Moreover, dalbavancin impaired the bactericidal activity of neutrophilic granulocytes. CONCLUSION: We here identified hitherto unknown inhibitory effects of several classes of antibiotics on the effector functions of neutrophilic granulocytes.


Asunto(s)
Ceftazidima , Neutrófilos , Humanos , Ceftazidima/metabolismo , Ceftazidima/farmacología , Teicoplanina/farmacología , Teicoplanina/metabolismo , Escherichia coli , Interleucina-8/metabolismo , Antibacterianos/farmacología
5.
J Basic Microbiol ; 63(9): 1007-1015, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36086811

RESUMEN

The aim of this study was to investigate the effect of zinc oxide nanoparticles (ZnO-NPs) on the expression of genes involved in toxin-antitoxin (TA) systems in multidrug-resistant (MDR) Acinetobacter baumannii. Seventy clinical isolates of A. baumannii were collected from variuos clinical samples. Antimicrobial susceptibility test was determined by disk diffusion. Type II TA system-related genes including GNAT, XRE-like, hipA, hipB, hicA, hicB were screened using polymerase chain reaction (PCR). ZnO-NPs prepared and characterized by field emission scanning electron microscopy and X-ray diffraction. MIC of ZnO-NPs of A. baumannii isolates was performed using the microdilution method. The expression of type II TA systems-related genes were assessed with and without exposure to ZnO-NPs using real-time PCR. The highest rate of resistance and sensitivity was observed against cefepime (77.14%), and ampicillin/sulbactam (42.85%), respectively. All A. baumannii isolates were considered as MDR. In this study, three TA loci were identified for A. baumannii including GNAT/XRE-like, HicA/HicB, and HipA/HipB and their prevalence was 100%, 42%, and 27.1%, respectively. There was no significant relationship between the prevalence of these systems and the origin of A. baumannii. Our data showed significant correlations between the presence of HicA/HicB system and resistance to ceftazidime, meropenem, imipenem, and cefepime (p < 0.05), and the presence of HipA/HipB system and resistance to ceftazidime, meropenem, imipenem, and cefepime (p < 0.05). In presence of ZnO-NPs, the expression of all studied genes decreased. GNAT and hicB showed the highest and lowest expression changes by 2.4 folds (p < 0.001) and 1.3 folds (p < 0.05), respectively. This study demonstrates the promising potential of nanoparticles to impact the expression of the genes involved in TA Systems. So, the application of ZnO-NPs may be helpful to design target-based strategies towards MDRs pathogens for empowered clinical applications by microbiologists and nanotechnologists.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Nanopartículas , Sistemas Toxina-Antitoxina , Óxido de Zinc , Humanos , Antibacterianos/farmacología , Antibacterianos/metabolismo , Óxido de Zinc/farmacología , Ceftazidima/metabolismo , Ceftazidima/farmacología , Cefepima/metabolismo , Cefepima/farmacología , Meropenem/metabolismo , Meropenem/farmacología , Imipenem/metabolismo , Imipenem/farmacología , Pruebas de Sensibilidad Microbiana , Farmacorresistencia Bacteriana Múltiple/genética
6.
Xenobiotica ; 50(7): 815-821, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31755347

RESUMEN

Recent approvals of beta-lactamase inhibitor (BLI) drug in combination with cephalosporins/penems have provided the right impetus for novel BLIs. One important research question, hitherto not addressed, is pertaining to the relevance of preclinical pharmacokinetics for pairing the antibiotic with existing/novel BLI.Two BLI combination drugs: (a) approved (i.e. ceftazidime/avibactam); (b) clinical development (i.e. cefepime/zidebactam) were explored to provide insights to address the research question.Individual intravenous dosing of ceftazidime, avibactam, cefepime and zidebactam was done at 1 mg/kg by intravenous route in Balb/c mice and Wistar rats. Serial blood samples were collected and analysed by LC-MS/MS method.Examination of the ratios of pharmacokinetic parameters (CL, VSS and T1/2) for individual drugs in combinations (for instance, CL (ceftazidime)/CL (avibactam); CL (cefepime)/CL (zidebactam)) suggested that the pharmacokinetic data gathered in rats were generally within 0.5- to 2-fold; but mouse data revealed larger disparity for VSS (0.11- to 8.25-fold) or CL (0.49- to 4.03-fold).The observed ratio for CL/VSS observed in rats agreed with corresponding human ratios for the pairwise comparison of the individual drugs in the combinations.Retrospectively, current pharmacokinetic findings suggest rat pharmacokinetic data may aid the combination of BLI with an appropriate antibiotic.


Asunto(s)
Compuestos de Azabiciclo/metabolismo , Ceftazidima/metabolismo , Inhibidores de beta-Lactamasas/metabolismo , Animales , Ciclooctanos , Combinación de Medicamentos , Ratones , Pruebas de Sensibilidad Microbiana , Piperidinas , Ratas , Roedores
7.
Artículo en Inglés | MEDLINE | ID: mdl-30348667

RESUMEN

Carbapenems are "last resort" ß-lactam antibiotics used to treat serious and life-threatening health care-associated infections caused by multidrug-resistant Gram-negative bacteria. Unfortunately, the worldwide spread of genes coding for carbapenemases among these bacteria is threatening these life-saving drugs. Metallo-ß-lactamases (MßLs) are the largest family of carbapenemases. These are Zn(II)-dependent hydrolases that are active against almost all ß-lactam antibiotics. Their catalytic mechanism and the features driving substrate specificity have been matter of intense debate. The active sites of MßLs are flanked by two loops, one of which, loop L3, was shown to adopt different conformations upon substrate or inhibitor binding, and thus are expected to play a role in substrate recognition. However, the sequence heterogeneity observed in this loop in different MßLs has limited the generalizations about its role. Here, we report the engineering of different loops within the scaffold of the clinically relevant carbapenemase NDM-1. We found that the loop sequence dictates its conformation in the unbound form of the enzyme, eliciting different degrees of active-site exposure. However, these structural changes have a minor impact on the substrate profile. Instead, we report that the loop conformation determines the protonation rate of key reaction intermediates accumulated during the hydrolysis of different ß-lactams in all MßLs. This study demonstrates the existence of a direct link between the conformation of this loop and the mechanistic features of the enzyme, bringing to light an unexplored function of active-site loops on MßLs.


Asunto(s)
Antibacterianos/química , Ceftazidima/química , Imipenem/química , Meropenem/química , Zinc/química , beta-Lactamasas/química , Secuencia de Aminoácidos , Antibacterianos/metabolismo , Dominio Catalítico , Cefepima/química , Cefepima/metabolismo , Cefotaxima/química , Cefotaxima/metabolismo , Ceftazidima/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Imipenem/metabolismo , Cinética , Meropenem/metabolismo , Modelos Moleculares , Piperacilina/química , Piperacilina/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Ingeniería de Proteínas , 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 , Zinc/metabolismo , Resistencia betalactámica , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
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.
Artículo en Inglés | MEDLINE | ID: mdl-29632016

RESUMEN

The natural diversification of CTX-M ß-lactamases led to the emergence of Asp240Gly variants in the clinic that confer reduced susceptibility to ceftazidime (CAZ). In this study, we compared the impact of this substitution on CAZ and ceftazidime-avibactam (CZA) MICs against isogenic Escherichia coli strains with different porin deficiencies. Our results show a noticeable increase in CAZ resistance in clones expressing Asp240Gly-harboring CTX-M when combined with OmpF porin deficiency. Kinetic analysis revealed that the kcat/Km for CAZ was 5- to 15-fold higher for all Asp240Gly variants but remained 200- to 725-fold lower than that for cefotaxime (CTX). In vitro selection of CAZ-resistant clones yielded nonsusceptible CTX-M producers (MIC of >16 µg/ml) only after overnight incubation; the addition of avibactam (AVI) decreased MICs to a susceptible range against these variants. In contrast, the use of CZA as a selective agent did not yield resistant clones. AVI inactivated both CTX-M-12 and CTX-M-96, with an apparent inhibition constant comparable to that of SHV-2 and 1,000-fold greater than that of PER-2 and CMY-2, and k2/K for CTX-M-12 was 24- and 35-fold higher than that for CTX-M-96 and CTX-M-15, respectively. Molecular modeling suggests that AVI interacts similarly with CTX-M-96 and CTX-M-15. We conclude that the impact of Asp240Gly in resistance may arise when other mechanisms are also present (i.e., OmpF deficiency). Additionally, CAZ selection could favor the emergence of CAZ-resistant subpopulations. These results define the role of Asp240 and the impact of the -Gly substitution and allow us to hypothesize that the use of CZA is an effective preventive strategy to delay the development of resistance in this family of extended-spectrum ß-lactamases.


Asunto(s)
Sustitución de Aminoácidos/genética , Compuestos de Azabiciclo/metabolismo , Ceftazidima/metabolismo , Proteínas de Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Porinas/genética , beta-Lactamasas/genética , Compuestos de Azabiciclo/farmacología , Ceftazidima/farmacología , Combinación de Medicamentos , Farmacorresistencia Bacteriana/genética , Proteínas de Escherichia coli/metabolismo , Hidrólisis , Pruebas de Sensibilidad Microbiana , Especificidad por Sustrato , beta-Lactamasas/metabolismo
10.
Biochemistry ; 56(27): 3443-3453, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28613873

RESUMEN

CTX-M ß-lactamases provide resistance against the ß-lactam antibiotic, cefotaxime, but not a related antibiotic, ceftazidime. ß-Lactamases that carry the P167S substitution, however, provide ceftazidime resistance. In this study, CTX-M-14 was used as a model to study the structural changes caused by the P167S mutation that accelerate ceftazidime turnover. X-ray crystallography was used to determine the structures of the P167S apoenzyme along with the structures of the S70G/P167S, E166A/P167S, and E166A mutant enzymes complexed with ceftazidime as well as the E166A/P167S apoenzyme. The S70G and E166A mutations allow capture of the enzyme-substrate complex and the acylated form of ceftazidime, respectively. The results showed a large conformational change in the Ω-loop of the ceftazidime acyl-enzyme complex of the P167S mutant but not in the enzyme-substrate complex, suggesting the change occurs upon acylation. The change results in a larger active site that prevents steric clash between the aminothiazole ring of ceftazidime and the Asn170 residue in the Ω-loop, allowing accommodation of ceftazidime for hydrolysis. In addition, the conformational change was not observed in the E166A/P167S apoenzyme, suggesting the presence of acylated ceftazidime influences the conformational change. Finally, the E166A acyl-enzyme structure with ceftazidime did not exhibit the altered conformation, indicating the P167S substitution is required for the change. Taken together, the results reveal that the P167S substitution and the presence of acylated ceftazidime both drive the structure toward a conformational change in the Ω-loop and that in CTX-M P167S enzymes found in drug-resistant bacteria this will lead to an increased level of ceftazidime hydrolysis.


Asunto(s)
Antibacterianos/metabolismo , Ceftazidima/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Modelos Moleculares , beta-Lactamasas/metabolismo , Acilación , Sustitución de Aminoácidos , Antibacterianos/química , Antibacterianos/farmacología , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Dominio Catalítico , Ceftazidima/química , Ceftazidima/farmacología , Cefalosporinas/química , Cefalosporinas/metabolismo , Cefalosporinas/farmacología , Cristalografía por Rayos X , Farmacorresistencia Bacteriana Múltiple , Estabilidad de Enzimas , Escherichia coli/efectos de los fármacos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Hidrólisis , Ligandos , Mutagénesis Sitio-Dirigida , Oximas/química , Oximas/metabolismo , Oximas/farmacología , Mutación Puntual , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , beta-Lactamasas/química , beta-Lactamasas/genética
11.
J Antimicrob Chemother ; 72(4): 1109-1117, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28077672

RESUMEN

Objectives: This study evaluated the in vitro pharmacodynamics of combinations of ceftazidime and the non-ß-lactam ß-lactamase inhibitor, avibactam, against ceftazidime-, piperacillin/tazobactam- and meropenem-multiresistant Pseudomonas aeruginosa by a quantitative time-kill method. Methods: MICs of ceftazidime plus 0-16 mg/L avibactam were determined against eight isolates of P. aeruginosa . Single-compartment, 24 h time-kill kinetics were investigated for three isolates at 0-16 mg/L avibactam with ceftazidime at 0.25-4-fold the MIC as measured at the respective avibactam concentration. Ceftazidime and avibactam concentrations were measured by LC-MS/MS during the time-kill kinetic studies to evaluate drug degradation. Results: Avibactam alone displayed no antimicrobial activity. MICs of ceftazidime decreased by 8-16-fold in the presence of avibactam at 4 mg/L. The changes in log 10 cfu/mL at both the 10 h and 24 h timepoints (versus 0 h) revealed bacterial killing at ≥1-fold MIC. Significantly higher concentrations of ceftazidime alone, as compared with those of ceftazidime in combination, were required to produce any given kill. Without avibactam, ceftazidime degradation was significant (defined as degradation t 1/2 < 24 h), with as little as 19% ± 18% of the original concentration remaining at 8 h for the most resistant strain. In combination with avibactam, ceftazidime degradation at ≥ 1-fold MIC was negligible. Conclusion: The addition of avibactam protected ceftazidime from degradation in a dose-dependent manner and restored its cidal and static activity at concentrations in combination well below the MIC of ceftazidime alone.


Asunto(s)
Antibacterianos/farmacología , Compuestos de Azabiciclo/farmacología , Ceftazidima/farmacología , Ácido Penicilánico/análogos & derivados , Pseudomonas aeruginosa/efectos de los fármacos , Resistencia betalactámica , Inhibidores de beta-Lactamasas/farmacología , Ceftazidima/metabolismo , Sinergismo Farmacológico , Cinética , Meropenem , Pruebas de Sensibilidad Microbiana/métodos , Ácido Penicilánico/farmacología , Piperacilina/farmacología , Combinación Piperacilina y Tazobactam , Espectrometría de Masas en Tándem/métodos , Tienamicinas/farmacología
12.
J Antimicrob Chemother ; 72(7): 1930-1936, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28333323

RESUMEN

Objectives: To investigate mechanisms for the decreased susceptibility to ceftazidime/avibactam in KPC-producing Klebsiella pneumoniae (KPC-KP). Methods: A total of 24 isolates, 8 each with ceftazidime/avibactam MICs of 4-8, 1-2 and ≤0.5 mg/L, were randomly selected from 214 clinical isolates of KPC-KP, and the ß-lactamase hydrolysis activity and porin expression profiles were determined. Plasmid profile and relative expression and copy number of the bla KPC gene were also analysed. Results: Ceftazidime/avibactam MIC 50 and MIC 90 were 2 and 4 mg/L, respectively, for the 214 KPC-KP isolates. The hydrolysis activities of nitrocefin and ceftazidime in both of the ceftazidime/avibactam MIC 4-8 and 1-2 mg/L groups were significantly higher than those of the MIC ≤0.5 mg/L group, while the hydrolysis activities were 4-4.6-fold higher in the MIC 4-8 mg/L group than in the other two groups when 4 mg/L avibactam was added. The relative expression and copy number of the bla KPC gene in the MIC 4-8 mg/L group were 4.2-4.8-fold higher than in the other two groups. Meanwhile, SDS-PAGE showed that all isolates in the two groups with MIC ≥1 mg/L lacked OmpK35, which had either an early frameshift with a premature stop codon ( n = 15, ST11) or overexpression of the negative regulation genes, micF and ompR ( n = 1, ST15), whereas OmpK35 and OmpK36 could both be observed in all isolates with MIC ≤0.5 mg/L. Conclusions: Decreased ceftazidime/avibactam susceptibility in KPC-KP clinical isolates is caused by high ceftazidime hydrolysis activity and OmpK35 porin deficiency and the majority of isolates belong to ST11.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Ceftazidima/metabolismo , Klebsiella pneumoniae/efectos de los fármacos , Porinas/deficiencia , Inhibidores de beta-Lactamasas/farmacología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ceftazidima/farmacología , Combinación de Medicamentos , Electroforesis en Gel de Poliacrilamida , Humanos , Hidrólisis , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/enzimología , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Pruebas de Sensibilidad Microbiana , Porinas/genética , Porinas/metabolismo , beta-Lactamasas/biosíntesis , beta-Lactamasas/genética
13.
PLoS Pathog ; 11(6): e1004949, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26030609

RESUMEN

The spread of ß-lactamases that hydrolyze penicillins, cephalosporins and carbapenems among Gram-negative bacteria has limited options for treating bacterial infections. Initially, Klebsiella pneumoniae carbapenemase-2 (KPC-2) emerged as a widespread carbapenem hydrolyzing ß-lactamase that also hydrolyzes penicillins and cephalosporins but not cephamycins and ceftazidime. In recent years, single and double amino acid substitution variants of KPC-2 have emerged among clinical isolates that show increased resistance to ceftazidime. Because it confers multi-drug resistance, KPC ß-lactamase is a threat to public health. In this study, the evolution of KPC-2 function was determined in nine clinically isolated variants by examining the effects of the substitutions on enzyme kinetic parameters, protein stability and antibiotic resistance profile. The results indicate that the amino acid substitutions associated with KPC-2 natural variants lead to increased catalytic efficiency for ceftazidime hydrolysis and a consequent increase in ceftazidime resistance. Single substitutions lead to modest increases in catalytic activity while the double mutants exhibit significantly increased ceftazidime hydrolysis and resistance levels. The P104R, V240G and H274Y substitutions in single and double mutant combinations lead to the largest increases in ceftazidime hydrolysis and resistance. Molecular modeling suggests that the P104R and H274Y mutations could facilitate ceftazidime hydrolysis through increased hydrogen bonding interactions with the substrate while the V240G substitution may enhance backbone flexibility so that larger substrates might be accommodated in the active site. Additionally, we observed a strong correlation between gain of catalytic function for ceftazidime hydrolysis and loss of enzyme stability, which is in agreement with the 'stability-function tradeoff' phenomenon. The high Tm of KPC-2 (66.5°C) provides an evolutionary advantage as compared to other class A enzymes such as TEM (51.5°C) and CTX-M (51°C) in that it can acquire multiple destabilizing substitutions without losing the ability to fold into a functional enzyme.


Asunto(s)
Evolución Biológica , Modelos Moleculares , beta-Lactamasas/química , beta-Lactamasas/metabolismo , Antibacterianos/metabolismo , Ceftazidima/metabolismo , Estabilidad de Enzimas/fisiología , Hidrólisis , Pruebas de Sensibilidad Microbiana , Mutagénesis Sitio-Dirigida , Mutación , Estructura Cuaternaria de Proteína
14.
PLoS Genet ; 10(9): e1004640, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25233343

RESUMEN

Expansion or shrinkage of existing tandem repeats (TRs) associated with various biological processes has been actively studied in both prokaryotic and eukaryotic genomes, while their origin and biological implications remain mostly unknown. Here we describe various duplications (de novo TRs) that occurred in the coding region of a ß-lactamase gene, where a conserved structure called the omega loop is encoded. These duplications that occurred under selection using ceftazidime conferred substrate spectrum extension to include the antibiotic. Under selective pressure with one of the original substrates (amoxicillin), a high level of reversion occurred in the mutant ß-lactamase genes completing a cycle back to the original substrate spectrum. The de novo TRs coupled with reversion makes a genetic toggling mechanism enabling reversible switching between the two phases of the substrate spectrum of ß-lactamases. This toggle exemplifies the effective adaptation of de novo TRs for enhanced bacterial survival. We found pairs of direct repeats that mediated the DNA duplication (TR formation). In addition, we found different duos of sequences that mediated the DNA duplication. These novel elements-that we named SCSs (same-strand complementary sequences)-were also found associated with ß-lactamase TR mutations from clinical isolates. Both direct repeats and SCSs had a high correlation with TRs in diverse bacterial genomes throughout the major phylogenetic lineages, suggesting that they comprise a fundamental mechanism shaping the bacterial evolution.


Asunto(s)
Secuencias Repetidas en Tándem/genética , beta-Lactamasas/genética , beta-Lactamasas/metabolismo , Alelos , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Bacterias/genética , Secuencia de Bases , Evolución Biológica , Ceftazidima/metabolismo , Ceftazidima/farmacología , Duplicación de Gen , Genoma Bacteriano , Pruebas de Sensibilidad Microbiana , Modelos Biológicos , Datos de Secuencia Molecular , Filogenia , Mutación Puntual , Especificidad por Sustrato/genética
15.
J Biol Chem ; 290(16): 10382-94, 2015 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-25713062

RESUMEN

ß-Lactamases are bacterial enzymes that hydrolyze ß-lactam antibiotics. TEM-1 is a prevalent plasmid-encoded ß-lactamase in Gram-negative bacteria that efficiently catalyzes the hydrolysis of penicillins and early cephalosporins but not oxyimino-cephalosporins. A previous random mutagenesis study identified a W165Y/E166Y/P167G triple mutant that displays greatly altered substrate specificity with increased activity for the oxyimino-cephalosporin, ceftazidime, and decreased activity toward all other ß-lactams tested. Surprisingly, this mutant lacks the conserved Glu-166 residue critical for enzyme function. Ceftazidime contains a large, bulky side chain that does not fit optimally in the wild-type TEM-1 active site. Therefore, it was hypothesized that the substitutions in the mutant expand the binding site in the enzyme. To investigate structural changes and address whether there is an enlargement in the active site, the crystal structure of the triple mutant was solved to 1.44 Å. The structure reveals a large conformational change of the active site Ω-loop structure to create additional space for the ceftazidime side chain. The position of the hydroxyl group of Tyr-166 and an observed shift in the pH profile of the triple mutant suggests that Tyr-166 participates in the hydrolytic mechanism of the enzyme. These findings indicate that the highly conserved Glu-166 residue can be substituted in the mechanism of serine ß-lactamases. The results reveal that the robustness of the overall ß-lactamase fold coupled with the plasticity of an active site loop facilitates the evolution of enzyme specificity and mechanism.


Asunto(s)
Ceftazidima/química , Escherichia coli/enzimología , Mutación , beta-Lactamasas/química , Biocatálisis , Ceftazidima/metabolismo , Cristalografía por Rayos X , Escherichia coli/genética , Expresión Génica , Ácido Glutámico/química , Ácido Glutámico/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Modelos Moleculares , Penicilinas/química , Penicilinas/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Tirosina/química , Tirosina/metabolismo , Resistencia betalactámica/genética , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
16.
Antimicrob Agents Chemother ; 60(2): 990-1002, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26643332

RESUMEN

Metallo-ß-lactamases (MBLs) hydrolyze virtually all ß-lactam antibiotics, including penicillins, cephalosporins, and carbapenems. The worldwide emergence of antibiotic-resistant bacteria harboring MBLs poses an increasing clinical threat. The MBL German imipenemase-1 (GIM-1) possesses an active site that is narrower and more hydrophobic than the active sites of other MBLs. The GIM-1 active-site groove is shaped by the presence of the aromatic side chains of tryptophan at residue 228 and tyrosine at residue 233, positions where other MBLs harbor hydrophilic residues. To investigate the importance of these two residues, eight site-directed mutants of GIM-1, W228R/A/Y/S and Y233N/A/I/S, were generated and characterized using enzyme kinetics, thermostability assays, and determination of the MICs of representative ß-lactams. The structures of selected mutants were obtained by X-ray crystallography, and their interactions with ß-lactam substrates were modeled in silico. Steady-state kinetics revealed that both positions are important to GIM-1 activity but that the effects of individual mutations vary depending on the ß-lactam substrate. Activity against type 1 substrates bearing electron-donating C-3/C-4 substituents (cefoxitin, meropenem) could be enhanced by mutations at position 228, whereas hydrolysis of type 2 substrates (benzylpenicillin, ampicillin, ceftazidime, imipenem) with methyl or positively charged substituents was favored by mutations at position 233. The crystal structures showed that mutations at position 228 or the Y233A variant alters the conformation of GIM-1 loop L1 rather than that of loop L3, on which the mutations are located. Taken together, these data show that point mutations at both positions 228 and 233 can influence the catalytic properties and the structure of GIM-1.


Asunto(s)
beta-Lactamasas/química , beta-Lactamasas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Ampicilina/metabolismo , Dominio Catalítico , Ceftazidima/metabolismo , Cristalografía por Rayos X , Estabilidad de Enzimas , Hidrólisis , Imipenem/metabolismo , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , beta-Lactamasas/genética
17.
Antimicrob Agents Chemother ; 60(4): 2281-91, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26833150

RESUMEN

Deciphering the mode of action (MOA) of new antibiotics discovered through phenotypic screening is of increasing importance. Metabolomics offers a potentially rapid and cost-effective means of identifying modes of action of drugs whose effects are mediated through changes in metabolism. Metabolomics techniques also collect data on off-target effects and drug modifications. Here, we present data from an untargeted liquid chromatography-mass spectrometry approach to identify the modes of action of eight compounds: 1-[3-fluoro-4-(5-methyl-2,4-dioxo-pyrimidin-1-yl)phenyl]-3-[2-(trifluoromethyl)phenyl]urea (AZ1), 2-(cyclobutylmethoxy)-5'-deoxyadenosine, triclosan, fosmidomycin, CHIR-090, carbonyl cyanidem-chlorophenylhydrazone (CCCP), 5-chloro-2-(methylsulfonyl)-N-(1,3-thiazol-2-yl)-4-pyrimidinecarboxamide (AZ7), and ceftazidime. Data analysts were blind to the compound identities but managed to identify the target as thymidylate kinase for AZ1, isoprenoid biosynthesis for fosmidomycin, acyl-transferase for CHIR-090, and DNA metabolism for 2-(cyclobutylmethoxy)-5'-deoxyadenosine. Changes to cell wall metabolites were seen in ceftazidime treatments, although other changes, presumably relating to off-target effects, dominated spectral outputs in the untargeted approach. Drugs which do not work through metabolic pathways, such as the proton carrier CCCP, have no discernible impact on the metabolome. The untargeted metabolomics approach also revealed modifications to two compounds, namely, fosmidomycin and AZ7. An untreated control was also analyzed, and changes to the metabolome were seen over 4 h, highlighting the necessity for careful controls in these types of studies. Metabolomics is a useful tool in the analysis of drug modes of action and can complement other technologies already in use.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Metaboloma/efectos de los fármacos , Metabolómica , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/genética , Aciltransferasas/metabolismo , Adenosina/metabolismo , Adenosina/farmacología , Antibacterianos/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Ceftazidima/metabolismo , Ceftazidima/farmacología , Pared Celular/química , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Cromatografía Liquida , ADN Bacteriano/antagonistas & inhibidores , ADN Bacteriano/biosíntesis , Escherichia coli/genética , Escherichia coli/metabolismo , Fosfomicina/análogos & derivados , Fosfomicina/metabolismo , Fosfomicina/farmacología , Expresión Génica , Células HEK293 , Humanos , Ácidos Hidroxámicos/metabolismo , Ácidos Hidroxámicos/farmacología , Espectrometría de Masas , Nucleósido-Fosfato Quinasa/antagonistas & inhibidores , Nucleósido-Fosfato Quinasa/genética , Nucleósido-Fosfato Quinasa/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Terpenos/antagonistas & inhibidores , Terpenos/metabolismo , Treonina/análogos & derivados , Treonina/metabolismo , Treonina/farmacología , Triclosán/metabolismo , Triclosán/farmacología
18.
Xenobiotica ; 46(9): 784-92, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26711252

RESUMEN

1. Interspecies allometry scaling for prediction of human excretory amounts in urine or feces was performed for numerous antibacterials. Antibacterials used for urinary scaling were: rifapentine, pefloxacin, trovafloxacin (Gr1/low; <10%); miloxacin, linezolid, PNU-142300 (Gr2/medium; 10-40%); aztreonam, carumonam, cefozopran, doripenem, imipenem, and ceftazidime (Gr3/high; >50%). Rifapentine, cabotegravir, and dolutegravir was used for fecal scaling (high; >50%). 2. The employment of allometry equation: Y = aW(b) enabled scaling of urine/fecal amounts from animal species. Corresponding predicted amounts were converted into % recovery by considering the respective human dose. Comparison of predicted/observed values enabled fold difference and error calculations (mean absolute error [MAE] and root mean square error [RMSE]). Comparisons were made for urinary/fecal data; and qualitative assessment was made amongst Gr1/Gr2/Gr3 for urine. 3. Average correlation coefficient for the allometry scaling was >0.995. Excretory amount predictions were largely within 0.75- to 1.5-fold differences. Average MAE and RMSE were within ±22% and 23%, respectively. Although robust predictions were achieved for higher urinary/fecal excretion (>50%), interspecies scaling was applicable for low/medium excretory drugs. 4. Based on the data, interspecies scaling of urine or fecal excretory amounts may be potentially used as a tool to understand the significance of either urinary or fecal routes of elimination in humans in early development.


Asunto(s)
Antibacterianos/metabolismo , Animales , Antibacterianos/orina , Aztreonam/análogos & derivados , Aztreonam/metabolismo , Carbapenémicos/metabolismo , Ceftazidima/metabolismo , Cefalosporinas/metabolismo , Doripenem , Heces/química , Fluoroquinolonas/metabolismo , Compuestos Heterocíclicos con 3 Anillos/metabolismo , Humanos , Imipenem/metabolismo , Linezolid/metabolismo , Naftiridinas/metabolismo , Oxazinas , Ácido Oxolínico/análogos & derivados , Ácido Oxolínico/metabolismo , Pefloxacina/metabolismo , Piperazinas , Piridonas , Estudios Retrospectivos , Rifampin/análogos & derivados , Rifampin/metabolismo , Cefozoprán
19.
Biochemistry ; 54(21): 3370-80, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25938261

RESUMEN

OXA-163 and OXA-48 are closely related class D ß-lactamases that exhibit different substrate profiles. OXA-163 hydrolyzes oxyimino-cephalosporins, particularly ceftazidime, while OXA-48 prefers carbapenem substrates. OXA-163 differs from OXA-48 by one substitution (S212D) in the active-site ß5 strand and a four-amino acid deletion (214-RIEP-217) in the loop connecting the ß5 and ß6 strands. Although the structure of OXA-48 has been determined, the structure of OXA-163 is unknown. To further understand the basis for their different substrate specificities, we performed enzyme kinetic analysis, inhibition assays, X-ray crystallography, and molecular modeling. The results confirm the carbapenemase nature of OXA-48 and the ability of OXA-163 to hydrolyze the oxyimino-cephalosporin ceftazidime. The crystal structure of OXA-163 determined at 1.72 Å resolution reveals an expanded active site compared to that of OXA-48, which allows the bulky substrate ceftazidime to be accommodated. The structural differences with OXA-48, which cannot hydrolyze ceftazidime, provide a rationale for the change in substrate specificity between the enzymes. OXA-163 also crystallized under another condition that included iodide. The crystal structure determined at 2.87 Å resolution revealed iodide in the active site accompanied by several significant conformational changes, including a distortion of the ß5 strand, decarboxylation of Lys73, and distortion of the substrate-binding site. Further studies showed that both OXA-163 and OXA-48 are inhibited in the presence of iodide. In addition, OXA-10, which is not a member of the OXA-48-like family, is also inhibited by iodide. These findings provide a molecular basis for the hydrolysis of ceftazidime by OXA-163 and, more broadly, show how minor sequence changes can profoundly alter the active-site configuration and thereby affect the substrate profile of an enzyme.


Asunto(s)
Enterobacter cloacae/enzimología , Inhibidores Enzimáticos/farmacología , Yoduros/farmacología , Klebsiella pneumoniae/enzimología , beta-Lactamasas/química , Antibacterianos/metabolismo , Carbapenémicos/metabolismo , Dominio Catalítico , Ceftazidima/metabolismo , Cefalosporinas/metabolismo , Enterobacter cloacae/química , Infecciones por Enterobacteriaceae/microbiología , Humanos , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/química , Simulación del Acoplamiento Molecular , Conformación Proteica
20.
Biochemistry ; 54(32): 5072-82, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26228623

RESUMEN

Diversification of the CTX-M ß-lactamases led to the emergence of variants responsible for decreased susceptibility to ceftazidime, like the Asp240Gly-harboring "ceftazidimases". We solved the crystallographic structure of the Asp240Gly variant CTX-M-96 at 1.2 Å and evaluated the role of Asp240 in the activity toward oxyimino-cephalosporins through simulated models and kinetics. There seem to be subtle changes in the conformation of the active site cavity of CTX-M-96, compared to enzyme variants harboring the Asp240, and these small rearrangements could be due to localized shifts in the environment of the ß3 strand. According to the crystallographic evidence, CTX-M-96 presents a "compact" active site, which in spite of its reduced cavity seems to allow the proper interaction with oxyimino-cephalosporins, as suggested by simulated models. The term "ceftazidimases" that is currently applied for the Asp240Gly-harboring CTX-M variants should be used carefully. Structural differences between CTX-M harboring the Asp240Gly mutation (and also probably others like those at Pro167) do not seem to be conclusive to determine the "ceftazidimase" behavior observed in vivo, which is in turn partially supported by the mild improvement in the catalytic efficiency toward ceftazidime by CTX-M-96 and similar enzymes, compared to "parental" Asp240-harboring variants. In addition, it is observed that alterations in OmpF expression could act synergistically with CTX-M-96 for yielding clinical resistance toward ceftazidime. We therefore propose that the observed resistance in vivo is due to the sum of synergic mechanisms, and the term "cefotaximases associated with ceftazidime resistance" could be conveniently used to describe CTX-M harboring the Asp240Gly substitution.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Ceftazidima/metabolismo , Klebsiella pneumoniae/enzimología , beta-Lactamasas/química , beta-Lactamasas/metabolismo , Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Dominio Catalítico , Ceftazidima/farmacología , Resistencia a las Cefalosporinas/genética , Cristalografía por Rayos X , Genes Bacterianos , Variación Genética , Cinética , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Modelos Moleculares , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , beta-Lactamasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA