Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
PLoS Pathog ; 19(7): e1011536, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37486930

RESUMO

Central metabolic pathways control virulence and antibiotic resistance, and constitute potential targets for antibacterial drugs. In Staphylococcus aureus the role of the pentose phosphate pathway (PPP) remains largely unexplored. Mutation of the 6-phosphogluconolactonase gene pgl, which encodes the only non-essential enzyme in the oxidative phase of the PPP, significantly increased MRSA resistance to ß-lactam antibiotics, particularly in chemically defined media with physiologically-relevant concentrations of glucose, and reduced oxacillin (OX)-induced lysis. Expression of the methicillin-resistance penicillin binding protein 2a and peptidoglycan architecture were unaffected. Carbon tracing and metabolomics revealed extensive metabolic reprogramming in the pgl mutant including increased flux to glycolysis, the TCA cycle, and several cell envelope precursors, which was consistent with increased ß-lactam resistance. Morphologically, pgl mutant cells were smaller than wild-type with a thicker cell wall and ruffled surface when grown in OX. The pgl mutation reduced resistance to Congo Red, sulfamethoxazole and oxidative stress, and increased resistance to targocil, fosfomycin and vancomycin. Levels of lipoteichoic acids (LTAs) were significantly reduced in pgl, which may limit cell lysis, while the surface charge of pgl cells was significantly more positive. A vraG mutation in pgl reversed the increased OX resistance phenotype, and partially restored wild-type surface charge, but not LTA levels. Mutations in vraF or graRS from the VraFG/GraRS complex that regulates DltABCD-mediated d-alanylation of teichoic acids (which in turn controls ß-lactam resistance and surface charge), also restored wild-type OX susceptibility. Collectively these data show that reduced levels of LTAs and OX-induced lysis combined with a VraFG/GraRS-dependent increase in cell surface positive charge are accompanied by significantly increased OX resistance in an MRSA pgl mutant.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Staphylococcus aureus Resistente à Meticilina/genética , Staphylococcus aureus Resistente à Meticilina/metabolismo , Via de Pentose Fosfato/genética , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Oxacilina/farmacologia , Parede Celular/metabolismo , Monobactamas/metabolismo , Resistência beta-Lactâmica/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Testes de Sensibilidade Microbiana
2.
J Bacteriol ; 205(3): e0047622, 2023 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-36840595

RESUMO

Antibiotic tolerance, the ability of bacteria to sustain viability in the presence of typically bactericidal antibiotics for extended time periods, is an understudied contributor to treatment failure. The Gram-negative pathogen Vibrio cholerae, the causative agent of cholera, becomes highly tolerant to ß-lactam antibiotics (penicillin and related compounds) in a process requiring the two-component system VxrAB. VxrAB is induced by exposure to cell wall damaging conditions, which results in the differential regulation of >100 genes. While the effectors of VxrAB are relatively well known, VxrAB environment-sensing and activation mechanisms remain a mystery. Here, we used transposon mutagenesis to screen for mutants that spontaneously upregulate VxrAB signaling. This screen was answered by genes known to be required for proper cell envelope homeostasis, validating the approach. Unexpectedly, we also uncovered a new connection between central carbon metabolism and antibiotic tolerance in Vibrio cholerae. Inactivation of pgi (vc0374, coding for glucose-6-phosphate isomerase) resulted in an intracellular accumulation of glucose-6-phosphate and fructose-6-phosphate, concomitant with a marked cell envelope defect, resulting in VxrAB induction. Deletion of pgi also increased sensitivity to ß-lactams and conferred a growth defect on salt-free LB, phenotypes that could be suppressed by deleting sugar uptake systems and by supplementing cell wall precursors in the growth medium. Our data suggest an important connection between central metabolism and cell envelope integrity and highlight a potential new target for developing novel antimicrobial agents. IMPORTANCE Antibiotic tolerance (the ability to survive exposure to antibiotics) is a stepping stone toward antibiotic resistance (the ability to grow in the presence of antibiotics), an increasingly common cause of antibiotic treatment failure. The mechanisms promoting tolerance are poorly understood. Here, we identified central carbon metabolism as a key contributor to antibiotic tolerance and resistance. A strain with a mutation in a sugar utilization pathway accumulates metabolites that likely shut down the synthesis of cell wall precursors, which weakens the cell wall and thus increases susceptibility to cell wall-active drugs. Our results illuminate the connection between central carbon metabolism and cell wall homeostasis in V. cholerae and suggest that interfering with metabolism may be a fruitful future strategy for the development of antibiotic adjuvants.


Assuntos
Vibrio cholerae , Vibrio cholerae/metabolismo , Mutação , Penicilinas/farmacologia , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Monobactamas/metabolismo , beta-Lactamas/farmacologia
3.
Molecules ; 28(20)2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37894491

RESUMO

Staphylococcus aureus is a common human pathogen. Methicillin-resistant Staphylococcus aureus (MRSA) infections pose significant and challenging therapeutic difficulties. MRSA often acquires the non-native gene PBP2a, which results in reduced susceptibility to ß-lactam antibiotics, thus conferring resistance. PBP2a has a lower affinity for methicillin, allowing bacteria to maintain peptidoglycan biosynthesis, a core component of the bacterial cell wall. Consequently, even in the presence of methicillin or other antibiotics, bacteria can develop resistance. Due to genes responsible for resistance, S. aureus becomes MRSA. The fundamental premise of this resistance mechanism is well-understood. Given the therapeutic concerns posed by resistant microorganisms, there is a legitimate demand for novel antibiotics. This review primarily focuses on PBP2a scaffolds and the various screening approaches used to identify PBP2a inhibitors. The following classes of compounds and their biological activities are discussed: Penicillin, Cephalosporins, Pyrazole-Benzimidazole-based derivatives, Oxadiazole-containing derivatives, non-ß-lactam allosteric inhibitors, 4-(3H)-Quinazolinones, Pyrrolylated chalcone, Bis-2-Oxoazetidinyl macrocycles (ß-lactam antibiotics with 1,3-Bridges), Macrocycle-embedded ß-lactams as novel inhibitors, Pyridine-Coupled Pyrimidinones, novel Naphthalimide corbelled aminothiazoximes, non-covalent inhibitors, Investigational-ß-lactam antibiotics, Carbapenem, novel Benzoxazole derivatives, Pyrazolylpyridine analogues, and other miscellaneous classes of scaffolds for PBP2a. Additionally, we discuss the penicillin-binding protein, a crucial target in the MRSA cell wall. Various aspects of PBP2a, bacterial cell walls, peptidoglycans, different crystal structures of PBP2a, synthetic routes for PBP2a inhibitors, and future perspectives on MRSA inhibitors are also explored.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Humanos , Proteínas de Ligação às Penicilinas/química , Staphylococcus aureus Resistente à Meticilina/metabolismo , Meticilina/metabolismo , Meticilina/farmacologia , Staphylococcus aureus/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Monobactamas/metabolismo , Proteínas de Bactérias/química , Testes de Sensibilidade Microbiana
4.
Antimicrob Agents Chemother ; 66(7): e0059022, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35762796

RESUMO

Tebipenem pivoxil hydrobromide (TBP-PI-HBr) is an oral carbapenem prodrug being developed for the treatment of serious bacterial infections. The active moiety, tebipenem, has broad-spectrum activity against common Enterobacterales pathogens, including extended-spectrum-ß-lactamase (ESBL)-producing multidrug-resistant strains. This study evaluated the intrapulmonary pharmacokinetics (PK) and epithelial lining fluid (ELF) and alveolar macrophage (AM) concentrations of tebipenem relative to plasma levels in nonsmoking, healthy adult subjects. Thirty subjects received oral TBP-PI-HBr at 600 mg every 8 h for five doses. Serial blood samples were collected following the last dose. Each subject underwent one standardized bronchoscopy with bronchoalveolar lavage (BAL) 1, 2, 4, 6, or 8 h after the fifth dose of TBP-PI-HBr. The tebipenem area under the concentration-time curve for the 8-h dosing interval (AUC0-8) values in plasma, ELF, and AMs were calculated using the mean concentration at each BAL sampling time. Ratios of AUC0-8 values for total ELF and AMs to those for unbound plasma were determined, using a plasma protein binding value of 42%. Mean values ± standard deviations (SD) of tebipenem maximum (Cmax) and minimum (Cmin) total plasma concentrations were 11.37 ± 3.87 mg/L and 0.043 ± 0.039 mg/L, respectively. Peak tebipenem concentrations in plasma, ELF, and AMs occurred at 1 h and then decreased over 8 h. Ratios of tebipenem AUC0-8 values for ELF and AMs to those for unbound plasma were 0.191 and 0.047, respectively. Four (13.3%) subjects experienced adverse events (diarrhea, fatigue, papule, and coronavirus disease 2019 [COVID-19]); all resolved, and none were severe or serious. Tebipenem is distributed into the lungs of healthy adults, which supports the further evaluation of TBP-PI-HBr for the treatment of lower respiratory tract bacterial infections caused by susceptible pathogens. (This study has been registered at ClinicalTrials.gov under identifier NCT04710407.).


Assuntos
Antibacterianos , COVID-19 , Administração Oral , Adulto , Antibacterianos/farmacocinética , Líquido da Lavagem Broncoalveolar , Carbapenêmicos/metabolismo , Humanos , Pulmão/metabolismo , Monobactamas/metabolismo
5.
Antimicrob Agents Chemother ; 66(10): e0087922, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36102663

RESUMO

Xeruborbactam (formerly QPX7728) is a cyclic boronate inhibitor of numerous serine and metallo-beta-lactamases. At concentrations generally higher than those required for beta-lactamase inhibition, xeruborbactam has direct antibacterial activity against some Gram-negative bacteria, with MIC50/MIC90 values of 16/32 µg/mL and 16/64 µg/mL against carbapenem-resistant Enterobacterales and carbapenem-resistant Acinetobacter baumannii, respectively (the MIC50/MIC90 values against Pseudomonas aeruginosa are >64 µg/mL). In Klebsiella pneumoniae, inactivation of OmpK36 alone or in combination with OmpK35 resulted in 2- to 4-fold increases in the xeruborbactam MIC. In A. baumannii and P. aeruginosa, AdeIJK and MexAB-OprM, respectively, affected xeruborbactam's antibacterial potency (the MICs were 4- to 16-fold higher in efflux-proficient strains). In Escherichia coli and K. pneumoniae, the 50% inhibitory concentrations (IC50s) of xeruborbactam's binding to penicillin-binding proteins (PBPs) PBP1a/PBP1b, PBP2, and PBP3 were in the 40 to 70 µM range; in A. baumannii, xeruborbactam bound to PBP1a, PBP2, and PBP3 with IC50s of 1.4 µM, 23 µM, and 140 µM, respectively. Treating K. pneumoniae and P. aeruginosa with xeruborbactam at 1× and 2× MIC resulted in changes of cellular morphology similar to those observed with meropenem; the morphological changes observed after treatment of A. baumannii were consistent with inhibition of multiple PBPs but were unique to xeruborbactam compared to the results for control beta-lactams. No single-step xeruborbactam resistance mutants were obtained after selection at 4× MIC of xeruborbactam using wild-type strains of E. coli, K. pneumoniae, and A. baumannii; mutations selected at 2× MIC in K. pneumoniae did not affect antibiotic potentiation by xeruborbactam through beta-lactamase inhibition. Consistent with inhibition of PBPs, xeruborbactam enhanced the potencies of beta-lactam antibiotics even against strains that lacked beta-lactamase. In a large panel of KPC-producing clinical isolates, the MIC90 values of meropenem tested with xeruborbactam (8 µg/mL) were at least 4-fold lower than those in combination with vaborbactam at 64 µg/mL, the concentration of vaborbactam that is associated with complete inhibition of KPC. The additional enhancement of the potency of beta-lactam antibiotics beyond beta-lactamase inhibition may contribute to the potentiation of beta-lactam antibiotics by xeruborbactam.


Assuntos
Antibacterianos , Escherichia coli , Meropeném/farmacologia , Meropeném/metabolismo , Proteínas de Ligação às Penicilinas/genética , Escherichia coli/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo , beta-Lactamases/genética , beta-Lactamas/farmacologia , Testes de Sensibilidade Microbiana , Klebsiella pneumoniae/genética , Carbapenêmicos/farmacologia , Carbapenêmicos/metabolismo , Monobactamas/metabolismo , Pseudomonas aeruginosa/metabolismo , Serina/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-29555629

RESUMO

The conjugation of siderophores to antimicrobial molecules is an attractive strategy to overcome the low outer membrane permeability of Gram-negative bacteria. In this Trojan horse approach, the transport of drug conjugates is redirected via TonB-dependent receptors (TBDR), which are involved in the uptake of essential nutrients, including iron. Previous reports have demonstrated the involvement of the TBDRs PiuA and PirA from Pseudomonas aeruginosa and their orthologues in Acinetobacter baumannii in the uptake of siderophore-beta-lactam drug conjugates. By in silico screening, we further identified a PiuA orthologue, termed PiuD, present in clinical isolates, including strain LESB58. The piuD gene in LESB58 is located at the same genetic locus as piuA in strain PAO1. PiuD has a similar crystal structure as PiuA and is involved in the transport of the siderophore-drug conjugates BAL30072, MC-1, and cefiderocol in strain LESB58. To screen for additional siderophore-drug uptake systems, we overexpressed 28 of the 34 TBDRs of strain PAO1 and identified PfuA, OptE, OptJ, and the pyochelin receptor FptA as novel TBDRs conferring increased susceptibility to siderophore-drug conjugates. The existence of a TBDR repertoire in P. aeruginosa able to transport siderophore-drug molecules potentially decreases the likelihood of resistance emergence during therapy.


Assuntos
Antibacterianos/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Membrana/metabolismo , Pseudomonas aeruginosa/metabolismo , Sideróforos/metabolismo , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/genética , Transporte Biológico/efeitos dos fármacos , Proteínas de Transporte/genética , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cefalosporinas/metabolismo , Proteínas de Membrana/genética , Monobactamas/metabolismo , Pseudomonas aeruginosa/genética , Tiazóis/metabolismo , beta-Lactamas/metabolismo , Cefiderocol
7.
Bioorg Med Chem Lett ; 28(4): 748-755, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29336873

RESUMO

Metallo-ß-lactamases (MBLs), such as New Delhi metallo-ß-lactamase (NDM-1) have spread world-wide and present a serious threat. Expression of MBLs confers resistance in Gram-negative bacteria to all classes of ß-lactam antibiotics, with the exception of monobactams, which are intrinsically stable to MBLs. However, existing first generation monobactam drugs like aztreonam have limited clinical utility against MBL-expressing strains because they are impacted by serine ß-lactamases (SBLs), which are often co-expressed in clinical isolates. Here, we optimized novel monobactams for stability against SBLs, which led to the identification of LYS228 (compound 31). LYS228 is potent in the presence of all classes of ß-lactamases and shows potent activity against carbapenem-resistant isolates of Enterobacteriaceae (CRE).


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Enterobacteriáceas Resistentes a Carbapenêmicos/efeitos dos fármacos , Monobactamas/farmacologia , Resistência beta-Lactâmica/efeitos dos fármacos , beta-Lactamases/metabolismo , Animais , Antibacterianos/efeitos adversos , Antibacterianos/química , Antibacterianos/metabolismo , Aztreonam/farmacologia , Células CHO , Cricetulus , Estabilidade de Medicamentos , Escherichia coli/efeitos dos fármacos , Feminino , Humanos , Meropeném , Camundongos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Monobactamas/efeitos adversos , Monobactamas/química , Monobactamas/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Convulsões/induzido quimicamente , Relação Estrutura-Atividade , Tienamicinas/farmacologia
8.
Antimicrob Agents Chemother ; 59(9): 5159-64, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26055384

RESUMO

Three Klebsiella pneumoniae clinical isolates demonstrating carbapenem resistance were recovered from different patients hospitalized at two medical centers in São Paulo, Brazil. Resistance to all ß-lactams, quinolones, and some aminoglycosides was observed for these isolates that were susceptible to polymyxin B. Carbapenem hydrolysis, which was inhibited by clavulanic acid, was observed for all K. pneumoniae isolates that belonged to the same pulsed-field gel electrophoresis (PFGE) type and a novel sequence type (ST), ST1781 (clonal complex 442 [CC442]). A 10-kb nonconjugative incompatibility group Q (IncQ) plasmid, denominated p60136, was transferred to Escherichia coli strain TOP10 cells by electroporation. The full sequencing of p60136 showed that it was composed of a mobilization system, ISKpn23, the phosphotransferase aph3A-VI, and a 941-bp open reading frame (ORF) that codified a 313-amino acid protein. This ORF was named bla BKC-1. Brazilian Klebsiella carbapenemase-1 (BKC-1) showed a pI of 6.0 and possessed the highest identity (63%) with a ß-lactamase of Sinorhizobium meliloti, an environmental bacterium. Hydrolysis studies demonstrated that purified BKC-1 not only hydrolyzed carbapenems but also penicillins, cephalosporins, and monobactams. However, the carbapenems were less efficiently hydrolyzed due to their very low kcat values (0.0016 to 0.031 s(-1)). In fact, oxacillin was the best substrate for BKC-1 (kcat /Km , 53,522.6 mM(-1) s(-1)). Here, we report a new class A carbapenemase, confirming the diversity and rapid evolution of ß-lactamases in K. pneumoniae clinical isolates.


Assuntos
Proteínas de Bactérias/metabolismo , Klebsiella pneumoniae/enzimologia , beta-Lactamases/metabolismo , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Brasil , Carbapenêmicos/metabolismo , Carbapenêmicos/farmacologia , Cefalosporinas/metabolismo , Cefalosporinas/farmacologia , Farmacorresistência Bacteriana , Eletroforese em Gel de Campo Pulsado , Testes de Sensibilidade Microbiana , Monobactamas/metabolismo , Monobactamas/farmacologia , Penicilinas/metabolismo , Penicilinas/farmacologia , Sinorhizobium meliloti/efeitos dos fármacos , Sinorhizobium meliloti/metabolismo
9.
Nat Commun ; 14(1): 8198, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-38081813

RESUMO

Antibiotic resistance of bacteria is considered one of the most alarming developments in modern medicine. While varied pathways for bacteria acquiring antibiotic resistance have been identified, there still are open questions concerning the mechanisms underlying resistance. Here, we show that alpha phenol-soluble modulins (PSMαs), functional bacterial amyloids secreted by Staphylococcus aureus, catalyze hydrolysis of ß-lactams, a prominent class of antibiotic compounds. Specifically, we show that PSMα2 and, particularly, PSMα3 catalyze hydrolysis of the amide-like bond of the four membered ß-lactam ring of nitrocefin, an antibiotic ß-lactam surrogate. Examination of the catalytic activities of several PSMα3 variants allowed mapping of the active sites on the amyloid fibrils' surface, specifically underscoring the key roles of the cross-α fibril organization, and the combined electrostatic and nucleophilic functions of the lysine arrays. Molecular dynamics simulations further illuminate the structural features of ß-lactam association upon the fibril surface. Complementary experimental data underscore the generality of the functional amyloid-mediated catalytic phenomenon, demonstrating hydrolysis of clinically employed ß-lactams by PSMα3 fibrils, and illustrating antibiotic degradation in actual S. aureus biofilms and live bacteria environments. Overall, this study unveils functional amyloids as catalytic agents inducing degradation of ß-lactam antibiotics, underlying possible antibiotic resistance mechanisms associated with bacterial biofilms.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Antibióticos beta Lactam , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Monobactamas/metabolismo , beta-Lactamas/farmacologia , beta-Lactamas/metabolismo , Infecções Estafilocócicas/microbiologia , Bactérias
10.
Transplant Cell Ther ; 29(1): 54.e1-54.e6, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36208727

RESUMO

The mechanism(s) of acquisition of extended-spectrum cephalosporin-resistant Enterobacteriaceae (ESCRE) on inpatient hospital units dedicated to hematopoietic stem cell transplantation (HSCT) is unclear. The objectives of this study were to determine whether ESCRE organisms are transmitted among patients housed on a HSCT unit, clarify the mechanisms involved, and determine whether routine surveillance for ESCRE carriage and contact isolation for ESCRE carriers is beneficial. The study was conducted on a 30-bed inpatient unit dedicated to the care of patients with hematologic malignancies and HSCT recipients. To investigate whether ESCRE organisms may be transmitted vertically to subsequent room occupants, presumably through contamination of room surfaces, we (1) cultured 6 high touch areas in 10 rooms before and 9 rooms after terminal cleaning that had been occupied by patients with ESCRE carriage, (2) determined the in vitro survivals of our most common clinical ESCRE species, and (3) followed the subsequent room occupants of 54 consecutive ESCRE colonized patients for the development of inpatient acquired ESCRE carriage. To investigate whether ESCRE organisms are transmitted horizontally among inpatients we (1) sequenced 60 available ESCRE Escherichia coli isolates obtained from unit inpatients and searched for identities using complete-genome multisequence locus typing (cgMLST) and (2) retrospectively tabulated the cumulative rates of acquired ESCRE carriage in 356 patients admitted for a first HSCT before (200 patients) or after (156 patients) institution of universal ESCRE stool surveillance and contact isolation for carriers. No ESCRE organisms were cultured from patient rooms before or after terminal cleaning. In vitro, few, if any, ESCRE organisms survived longer than 2 hours. Nine of the subsequent occupants of a room in which a patient with ESCRE carriage had resided were detected with ESCRE carriage, only 2 of whom carried the same species as that of the prior occupant. DNA sequencing and cgMLST determination of the 60 E. coli isolates showed 53 cgMLST strains. Seven of the 53 strains were shared by 2 patients. After institution of universal ESCRE surveillance/isolation there was a significant decline in acquired ESCRE carriage among HSCT recipients. We conclude that vertical transmission of ESCRE organisms through room contamination appears to be uncommon on modern HSCT units. Conversely, our results are consistent with the horizontal spread of ESCRE organisms, probably mediated by intermediate vectors such as personnel or shared equipment. Further studies are needed to better define the magnitude of and risk factors for ESCRE horizontal transfers and the benefits of ESCRE surveillance/isolation.


Assuntos
Infecção Hospitalar , Transplante de Células-Tronco Hematopoéticas , Humanos , Enterobacteriaceae/genética , Enterobacteriaceae/metabolismo , Cefalosporinas/uso terapêutico , Cefalosporinas/metabolismo , Escherichia coli/metabolismo , Estudos Retrospectivos , Infecção Hospitalar/prevenção & controle , beta-Lactamases/genética , beta-Lactamases/metabolismo , Monobactamas/metabolismo , Transplante de Células-Tronco Hematopoéticas/efeitos adversos
11.
Microbiol Spectr ; 11(1): e0297822, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36602386

RESUMO

The frequent resistance associated with ß-lactam antibiotics and the high frequency of mutations in ß-lactamases constitute a major clinical challenge that can no longer be ignored. Andrographolide (AP), a natural active compound, has been shown to restore susceptibility to ß-lactam antibiotics. Fluorescence quenching and molecular simulation showed that AP quenched the intrinsic fluorescence of ß-lactamase BlaZ and stably bound to the residues in the catalytic cavity of BlaZ. Of note, AP was found to reduce the stability of the cell wall (CW) in methicillin-resistant Staphylococcus aureus (MRSA), and in combination with penicillin G (PEN), it significantly induced CW roughness and dispersion and even caused its disintegration, while the same concentration of PEN did not. In addition, transcriptome sequencing revealed that AP induced a significant stress response and increased peptidoglycan (PG) synthesis but disrupted its cross-linking, and it repressed the expression of critical genes such as mecA, blaZ, and sarA. We also validated these findings by quantitative reverse transcription-PCR (qRT-PCR). Association analysis using the GEO database showed that the alterations caused by AP were similar to those caused by mutations in the sarA gene. In summary, AP was able to restore the susceptibility of MRSA to ß-lactam antibiotics, mainly by inhibiting the ß-lactamase BlaZ, by downregulating the expression of critical resistance genes such as mecA and blaZ, and by disrupting CW homeostasis. In addition, restoration of susceptibility to antibiotics could be achieved by inhibiting the global regulator SarA, providing an effective solution to alleviate the problem of bacterial resistance. IMPORTANCE Increasingly, alternatives to antibiotics are being used to mitigate the rapid onset and development of bacterial resistance, and the combination of natural compounds with traditional antibiotics has become an effective therapeutic strategy. Therefore, we attempted to discover more mechanisms to restore susceptibility and effective dosing strategies. Andrographolide (AP), as a natural active ingredient, can mediate recovery of susceptibility of MRSA to ß-lactam antibiotics. AP bound stably to the ß-lactamase BlaZ and impaired its hydrolytic activity. Notably, AP was able to downregulate the expression of critical resistance genes such as mecA, blaZ, and sarA. Meanwhile, it disrupted the CW cross-linking and homeostasis, while the same concentration of penicillin could not. The multiple inhibitory effect of AP resensitizes intrinsically resistant bacteria to ß-lactam antibiotics, effectively prolonging the use cycle of these antibiotics and providing an effective solution to reduce the dosage of antibiotics and providing a theoretical reference for the prevention and control of MRSA.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Testes de Sensibilidade Microbiana , Antibacterianos/farmacologia , Antibacterianos/metabolismo , beta-Lactamases/genética , beta-Lactamases/metabolismo , Penicilinas , Monobactamas/metabolismo , Monobactamas/farmacologia
12.
Front Cell Infect Microbiol ; 13: 1139796, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37234778

RESUMO

Introduction: Biofilm formation is the major pathogenicity of Staphylococcus epidermidis (S. epidermidis), which enhances bacterial resistance to antibiotics. Isookanin has potential inhibitory activity on biofilm. Method: The inhibiting mechanisms of isookanin against biofilm formation through surface hydrophobicity assay, exopolysaccharides, eDNA, gene expression analysis, microscopic visualization, and molecular docking were explored. Additionally, the combination of isookanin and ß-lactam antibiotics were evaluated by the broth micro-checkerboard assay. Results: The results showed that isookanin could decrease the biofilm formation of S. epidermidis by ≥85% at 250 µg/mL. The exopolysaccharides, eDNA and surface hydrophobicity were reduced after treatment with isookanin. Microscopic visualization analysis showed that there were fewer bacteria on the surface of the microscopic coverslip and the bacterial cell membrane was damaged after treatment with isookanin. The down-regulation of icaB and up-regulation of icaR were observed after treatment with isookanin. Additionally, the RNAIII gene was significantly up-regulated (p < 0.0001) at the mRNA level. Molecular docking showed that isookanin could bind to biofilm-related proteins. This indicated that isookanin can affect biofilm formation at the initial attachment phase and the aggregation phase. The FICI index showed that the combination of isookanin and ß-lactam antibiotics were synergistic and could reduce doses of antibiotics by inhibiting biofilm formation. Discussion: This study improved the antibiotic susceptibility of S. epidermidis through inhibition of the biofilm formation, and provided a guidance for the treatment of antibiotic resistance caused by biofilm.


Assuntos
Antibacterianos , Staphylococcus epidermidis , Staphylococcus epidermidis/genética , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Regulação para Baixo , Simulação de Acoplamento Molecular , Biofilmes , Monobactamas/metabolismo , Testes de Sensibilidade Microbiana
13.
mBio ; 14(1): e0247822, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36507833

RESUMO

The purine-derived signaling molecules c-di-AMP and (p)ppGpp control mecA/PBP2a-mediated ß-lactam resistance in methicillin-resistant Staphylococcus aureus (MRSA) raise the possibility that purine availability can control antibiotic susceptibility. Consistent with this, exogenous guanosine and xanthosine, which are fluxed through the GTP branch of purine biosynthesis, were shown to significantly reduce MRSA ß-lactam resistance. In contrast, adenosine (fluxed to ATP) significantly increased oxacillin resistance, whereas inosine (which can be fluxed to ATP and GTP via hypoxanthine) only marginally increased oxacillin susceptibility. Furthermore, mutations that interfere with de novo purine synthesis (pur operon), transport (NupG, PbuG, PbuX) and the salvage pathway (DeoD2, Hpt) increased ß-lactam resistance in MRSA strain JE2. Increased resistance of a nupG mutant was not significantly reversed by guanosine, indicating that NupG is required for guanosine transport, which is required to reduce ß-lactam resistance. Suppressor mutants resistant to oxacillin/guanosine combinations contained several purine salvage pathway mutations, including nupG and hpt. Guanosine significantly increased cell size and reduced levels of c-di-AMP, while inactivation of GdpP, the c-di-AMP phosphodiesterase negated the impact of guanosine on ß-lactam susceptibility. PBP2a expression was unaffected in nupG or deoD2 mutants, suggesting that guanosine-induced ß-lactam susceptibility may result from dysfunctional c-di-AMP-dependent osmoregulation. These data reveal the therapeutic potential of purine nucleosides, as ß-lactam adjuvants that interfere with the normal activation of c-di-AMP are required for high-level ß-lactam resistance in MRSA. IMPORTANCE The clinical burden of infections caused by antimicrobial resistant (AMR) pathogens is a leading threat to public health. Maintaining the effectiveness of existing antimicrobial drugs or finding ways to reintroduce drugs to which resistance is widespread is an important part of efforts to address the AMR crisis. Predominantly, the safest and most effective class of antibiotics are the ß-lactams, which are no longer effective against methicillin-resistant Staphylococcus aureus (MRSA). Here, we report that the purine nucleosides guanosine and xanthosine have potent activity as adjuvants that can resensitize MRSA to oxacillin and other ß-lactam antibiotics. Mechanistically, exposure of MRSA to these nucleosides significantly reduced the levels of the cyclic dinucleotide c-di-AMP, which is required for ß-lactam resistance. Drugs derived from nucleotides are widely used in the treatment of cancer and viral infections highlighting the clinical potential of using purine nucleosides to restore or enhance the therapeutic effectiveness of ß-lactams against MRSA and potentially other AMR pathogens.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Nucleosídeos de Purina/metabolismo , Nucleosídeos de Purina/farmacologia , Proteínas de Bactérias/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo , Oxacilina/farmacologia , beta-Lactamas/farmacologia , Monobactamas/metabolismo , Monobactamas/farmacologia , Guanosina/metabolismo , Guanosina/farmacologia , Trifosfato de Adenosina/metabolismo , Guanosina Trifosfato/metabolismo , Testes de Sensibilidade Microbiana , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/metabolismo , Resistência beta-Lactâmica/genética
14.
Commun Biol ; 5(1): 1059, 2022 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-36198902

RESUMO

Gram-negative porins are the main entry for small hydrophilic molecules. We studied translocation of structurally related cephalosporins, ceftazidime (CAZ), cefotaxime (CTX) and cefepime (FEP). CAZ is highly active on E. coli producing OmpF (Outer membrane protein F) but less efficient on cells expressing OmpC (Outer membrane protein C), whereas FEP and CTX kill bacteria regardless of the porin expressed. This matches with the different capacity of CAZ and FEP to accumulate into bacterial cells as quantified by LC-MS/MS (Liquid Chromatography Tandem Mass Spectrometry). Furthermore, porin reconstitution into planar lipid bilayer and zero current assays suggest permeation of ≈1,000 molecules of CAZ per sec and per channel through OmpF versus ≈500 through OmpC. Here, the instant killing is directly correlated to internal drug concentration. We propose that the net negative charge of CAZ represents a key advantage for permeation through OmpF porins that are less cation-selective than OmpC. These data could explain the decreased susceptibility to some cephalosporins of enterobacteria that exclusively express OmpC porins.


Assuntos
Cefalosporinas , Enterobacteriaceae , Cefepima/metabolismo , Cefotaxima/metabolismo , Ceftazidima , Cefalosporinas/farmacologia , Cromatografia Líquida , Escherichia coli/metabolismo , Bicamadas Lipídicas/metabolismo , Monobactamas/metabolismo , Porinas/química , Porinas/metabolismo , Espectrometria de Massas em Tandem
15.
Biomolecules ; 11(7)2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34356681

RESUMO

Novel antimicrobial strategies are urgently required because of the rising threat of multi drug resistant bacterial strains and the infections caused by them. Among the available target structures, the so-called penicillin binding proteins are of particular interest, owing to their good accessibility in the periplasmic space, and the lack of homologous proteins in humans, reducing the risk of side effects of potential drugs. In this report, we focus on the interaction of the innovative ß-lactam antibiotic AIC499 with penicillin binding protein 3 (PBP3) from Escherichia coli and Pseudomonas aeruginosa. This recently developed monobactam displays broad antimicrobial activity, against Gram-negative strains, and improved resistance to most classes of ß-lactamases. By analyzing crystal structures of the respective complexes, we were able to explore the binding mode of AIC499 to its target proteins. In addition, the apo structures determined for PBP3, from P. aeruginosa and the catalytic transpeptidase domain of the E. coli orthologue, provide new insights into the dynamics of these proteins and the impact of drug binding.


Assuntos
Monobactamas/metabolismo , Proteínas de Ligação às Penicilinas/química , Proteínas de Ligação às Penicilinas/metabolismo , Cristalografia por Raios X , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Monobactamas/química , Proteínas de Ligação às Penicilinas/genética , Conformação Proteica , Pseudomonas aeruginosa
16.
J Med Microbiol ; 57(Pt 8): 974-979, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18628498

RESUMO

Limited beta-lactams show antipseudomonal activity. The rapid spread of IMP-type metallo-beta-lactamases (MBLs), which have a broad spectrum of substrates and a poor susceptibility to clinically available inhibitors, further restricts beta-lactam use. In the present study, we evaluated the potency of IMP-10 MBL in hydrolysing antipseudomonal beta-lactams currently available in the clinic. Crude IMP-10 MBL was prepared from two clinical isolates of Pseudomonas aeruginosa harbouring the bla(IMP-10) gene. The sensitivity of beta-lactams to hydrolysis by IMP-10 MBL was determined by comparing the MICs of 14 antipseudomonal beta-lactams against a susceptible strain of P. aeruginosa in the presence and absence of IMP-10 MBL. Carbapenems (imipenem, meropenem and panipenem) and extended-spectrum cephems (ceftazidime, cefoperazone, cefsulodin and cefepime) were sensitive to the hydrolysing activity of IMP-10 MBL. By comparison, the fourth-generation cephem (cefpirome), the extended-spectrum penicillins (carbenicillin, ticarcillin, piperacillin and mezlocillin) and monobactams (aztreonam and carumonam) were relatively resistant to IMP-10 MBL. The sensitivity profile of antipseudomonal beta-lactams to IMP-10 MBL generated in the present study provides a valuable reference for antibiotic selection by medical professionals.


Assuntos
Monobactamas/metabolismo , beta-Lactamases/metabolismo , beta-Lactamas/metabolismo , Cefalosporinas/metabolismo , Hidrólise , Testes de Sensibilidade Microbiana , Monobactamas/farmacologia , Penicilinas/metabolismo , Sensibilidade e Especificidade , Especificidade por Substrato , beta-Lactamas/farmacologia
17.
J Med Chem ; 41(21): 3961-71, 1998 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-9767633

RESUMO

Bridged monobactams are novel, potent, mechanism-based inhibitors of class C beta-lactamases, designed using X-ray crystal structures of the enzymes. They stabilize the acyl-enzyme intermediate by blocking access of water to the enzyme-inhibitor ester bond. Bridged monobactams are selective class C beta-lactamase inhibitors, with half-inhibition constants as low as 10 nM, and are less effective against class A and class B enzymes (half-inhibition constants > 100 microM) because of the different hydrolysis mechanisms in these classes of beta-lactamases. The stability of the acyl-enzyme complexes formed with class C beta-lactamases (half-lives up to 2 days were observed) enabled determination of their crystal structures. The conformation of the inhibitor moiety was close to that predicted by molecular modeling, confirming a simple reaction mechanism, unlike those of known beta-lactamase inhibitors such as clavulanic acid and penam sulfones, which involve secondary rearrangements. Synergy between the bridged monobactams and beta-lactamase-labile antibiotics could be observed when such combinations were tested against strains of Enterobacteriaceae that produce large amounts of class C beta-lactamases. The minimal inhibitory concentration of the antibiotic of more than 64 mg/L could be decreased to 0.25 mg/L in a 1:4 combination with the inhibitor.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Monobactamas/síntese química , Inibidores de beta-Lactamases , Acilação , Sítios de Ligação , Ceftriaxona/farmacologia , Cefalosporinas/farmacologia , Citrobacter freundii/efeitos dos fármacos , Citrobacter freundii/enzimologia , Sinergismo Farmacológico , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/enzimologia , Cinética , Modelos Moleculares , Conformação Molecular , Monobactamas/metabolismo , Monobactamas/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/enzimologia , Resistência beta-Lactâmica , beta-Lactamases/metabolismo
18.
Diagn Microbiol Infect Dis ; 12(1): 25-31, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2653714

RESUMO

The historical background information concerning the metabolism of cephalosporins and selected other antiinfectives was reviewed as a preface to discussion concerning desacetyl-cefotaxime (dCTX), a metabolite of cefotaxime (CTX) sodium. Cephalothin and cephapirin were metabolized at the 3-position to less active desacetyl forms. However, the parent drugs and their metabolites interact in a favorable manner resulting in dominant additive or synergist inhibition of susceptible bacterial pathogens. Similarly, CTX plus dCTX have been described as being synergist in their activity against greater than 70% of Enterobacteriaceae or staphylococci and greater than 80% of anaerobic bacteria. Antagonism was rare with only two species and of no clinical significance. More recently, reported studies showed enhanced activity of CTX/dCTX against pathogens producing meningitis compared to ceftriaxone and other contemporary therapeutic agents. The dCTX compound was classified as a drug with a potency superior to a "second-generation" cephalosporin and possessed greater beta-lactamase stability against some enzymes compared to CTX. These features may explain low reported rates of superinfections and adverse side-effects, including resistance arising on chemotherapy. Physicians are cautioned to take into account the potential favorable effects of drug metabolites on antimicrobial spectrum, potency and applied pharmacokinetics. In the case of CTX/dCTX, the laboratory results for CTX will always underestimate its value or potency because of the contribution of the metabolite.


Assuntos
Cefotaxima/metabolismo , Cefalosporinas/metabolismo , 4-Quinolonas , Animais , Anti-Infecciosos/metabolismo , Bactérias Anaeróbias/efeitos dos fármacos , Cefotaxima/análogos & derivados , Cefotaxima/farmacologia , Cefalosporinas/farmacologia , Cefalotina/análogos & derivados , Cefalotina/metabolismo , Cefapirina/análogos & derivados , Cefapirina/metabolismo , Fenômenos Químicos , Química , Humanos , Monobactamas/metabolismo , Staphylococcus/efeitos dos fármacos
19.
FEMS Microbiol Lett ; 174(1): 185-90, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10234838

RESUMO

A clinical strain of Escherichia coli (strain Ec 41553) that was resistant to ceftazidime produced a TEM-type beta-lactamase with a pI of 5.4. Clavulanic acid restored the ceftazidime activity, thus suggesting an extended spectrum beta-lactamase (ESBL). The gene encoding ESBL was located in a plasmid of 57 kb. After cloning and sequencing, the ESBL (TEM-29B) showed one amino acid replacement with respect to the TEM-1 sequence, Arg-164 to His. This change increased mainly the rate of hydrolysis of ceftazidime but not of cefotaxime and aztreonam. The relevance of this substitution in the increase of ceftazidime MIC is thus stressed.


Assuntos
Ceftazidima/farmacologia , Resistência às Cefalosporinas , Proteínas de Escherichia coli , Escherichia coli/efeitos dos fármacos , Escherichia coli/enzimologia , beta-Lactamases/metabolismo , Aztreonam/metabolismo , Cefotaxima/metabolismo , Ceftazidima/metabolismo , Clonagem Molecular , Genes Bacterianos , Testes de Sensibilidade Microbiana , Monobactamas/metabolismo , Análise de Sequência de DNA , Especificidade por Substrato , beta-Lactamases/genética
20.
J Chemother ; 1 Suppl 2: 13-21, 1989 May.
Artigo em Inglês | MEDLINE | ID: mdl-2809698

RESUMO

SQ 30836 is an orally absorbed salt of tigemonam, a new monobactam similar to aztreonam in structure and microbiologic properties. When assayed against 400 clinical isolates, tigemonam's activity was similar to that of aztreonam and carumonam. It was highly effective against Enterobacteriaceae but showed poor activity against gram-positive organisms. It inhibited 90% of Escherichia coli, Klebsiella, Shigella, Yersinia, Proteus, Providencia, and Morganella strains at 0.5 micrograms/mL or less, and all Salmonella and Hafnia strains at 1 micrograms/mL or less. Citrobacter, Enterobacter, and Serratia strains were less susceptible (minimum inhibitory concentrations [MIC30] of 2, 32, and 8 micrograms/mL respectively). The activity of the new compound against Enterobacteriaceae is comparable with and often higher than that of third-generation cephalosporins and oral comparison compounds. In contrast to aztreonam, tigemonam had minimal activity against Pseudomonas sp and glucose nonfermenting gram-negative bacteria. Data suggest that poor penetration through the outer membranes of Pseudomonas sp may be responsible for this failure. Tigemonam was stable to hydrolysis by plasmid-mediated and chromosomal beta-lactamases. It was more stable than aztreonam to hydrolysis by the Kl enzyme of Klebsiella and by the Proteus vulgaris beta-lactamase. Also, measurement of the IC50 (concentration of inhibitor able to reduce the activity of the enzyme by 59%) showed that tigemonam has less affinity than aztreonam for class I cephalosporinases. However, only levels of beta-lactamase, not hydrolysis rates or affinity, correlated to MICs of the two monobactams for the resistant Enterobacter and Citrobacter strains.


Assuntos
Bactérias Gram-Negativas/efeitos dos fármacos , Monobactamas/farmacologia , beta-Lactamases/farmacologia , Interações Medicamentosas , Estabilidade de Medicamentos , Bactérias Gram-Negativas/metabolismo , Hidrólise , Testes de Sensibilidade Microbiana , Monobactamas/metabolismo , beta-Lactamases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA