Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Protein Sci ; 32(10): e4781, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37703013

RESUMO

The 11 lytic transglycosylases of Pseudomonas aeruginosa have overlapping activities in the turnover of the cell-wall peptidoglycan. Rare lipoprotein A (RlpA) is distinct among the 11 by its use of only peptidoglycan lacking peptide stems. The spatial localization of RlpA and its interactome within P. aeruginosa are unknown. We employed suppression of introduced amber codons at sites in the rlpA gene for the introduction of the unnatural-amino-acids Νζ -[(2-azidoethoxy)carbonyl]-l-lysine (compound 1) and Nζ -[[[3-(3-methyl-3H-diazirin-3-yl)propyl]amino]carbonyl]-l-lysine (compound 2). In live P. aeruginosa, full-length RlpA incorporating compound 1 into its sequence was fluorescently tagged using strained-promoted alkyne-azide cycloaddition and examined by fluorescence microscopy. RlpA is present at low levels along the sidewall length of the bacterium, and at higher levels at the nascent septa of replicating bacteria. In intact P. aeruginosa, UV photolysis of full-length RlpA having compound 2 within its sequence generated a transient reactive carbene, which engaged in photoaffinity capture of neighboring proteins. Thirteen proteins were identified. Three of these proteins-PBP1a, PBP5, and MreB-are members of the bacterial divisome. The use of the complementary methodologies of non-canonical amino-acid incorporation, photoaffinity proximity analysis, and fluorescent microscopy confirm a dominant septal location for the RlpA enzyme of P. aeruginosa, as a divisome-associated activity. This accomplishment adds to the emerging recognition of the value of these methodologies for identification of the intracellular localization of bacterial proteins.


Assuntos
Lipoproteína(a) , Pseudomonas aeruginosa , Lipoproteína(a)/metabolismo , Códon de Terminação/metabolismo , Peptidoglicano/metabolismo , Lisina/metabolismo
2.
J Biol Chem ; 299(10): 105198, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37660917

RESUMO

The bacterial cell envelope is the structure with which the bacterium engages with, and is protected from, its environment. Within this envelop is a conserved peptidoglycan polymer which confers shape and strength to the cell envelop. The enzymatic processes that build, remodel, and recycle the chemical components of this cross-linked polymer are preeminent targets of antibiotics and exploratory targets for emerging antibiotic structures. We report a comprehensive kinetic and structural analysis for one such enzyme, the Pseudomonas aeruginosa anhydro-N-acetylmuramic acid (anhNAM) kinase (AnmK). AnmK is an enzyme in the peptidoglycan-recycling pathway of this pathogen. It catalyzes the pairing of hydrolytic ring opening of anhNAM with concomitant ATP-dependent phosphoryl transfer. AnmK follows a random-sequential kinetic mechanism with respect to its anhNAM and ATP substrates. Crystallographic analyses of four distinct structures (apo AnmK, AnmK:AMPPNP, AnmK:AMPPNP:anhNAM, and AnmK:ATP:anhNAM) demonstrate that both substrates enter the active site independently in an ungated conformation of the substrate subsites, with protein loops acting as gates for anhNAM binding. Catalysis occurs within a closed conformational state for the enzyme. We observe this state crystallographically using ATP-mimetic molecules. A remarkable X-ray structure for dimeric AnmK sheds light on the precatalytic and postcatalytic ternary complexes. Computational simulations in conjunction with the high-resolution X-ray structures reveal the full catalytic cycle. We further report that a P. aeruginosa strain with disrupted anmK gene is more susceptible to the ß-lactam imipenem compared to the WT strain. These observations position AnmK for understanding the nexus among peptidoglycan recycling, susceptibility to antibiotics, and bacterial virulence.


Assuntos
Proteínas de Bactérias , Modelos Moleculares , Fosfotransferases , Pseudomonas aeruginosa , Antibacterianos , Catálise , Cristalografia por Raios X , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Fosfotransferases/genética , Fosfotransferases/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estrutura Terciária de Proteína , Ativação Enzimática/genética , Farmacorresistência Bacteriana/genética
3.
Mar Drugs ; 21(2)2023 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-36827127

RESUMO

The title of this essay is as much a question as it is a statement. The discovery of the ß-lactam antibiotics-including penicillins, cephalosporins, and carbapenems-as largely (if not exclusively) secondary metabolites of terrestrial fungi and bacteria, transformed modern medicine. The antibiotic ß-lactams inactivate essential enzymes of bacterial cell-wall biosynthesis. Moreover, the ability of the ß-lactams to function as enzyme inhibitors is of such great medical value, that inhibitors of the enzymes which degrade hydrolytically the ß-lactams, the ß-lactamases, have equal value. Given this privileged status for the ß-lactam ring, it is therefore a disappointment that the exemplification of this ring in marine secondary metabolites is sparse. It may be that biologically active marine ß-lactams are there, and simply have yet to be encountered. In this report, we posit a second explanation: that the value of the ß-lactam to secure an ecological advantage in the marine environment might be compromised by its close structural similarity to the ß-lactones of quorum sensing. The steric and reactivity similarities between the ß-lactams and the ß-lactones represent an outside-of-the-box opportunity for correlating new structures and new enzyme targets for the discovery of compelling biological activities.


Assuntos
Antibacterianos , beta-Lactamas , beta-Lactamas/metabolismo , beta-Lactamas/farmacologia , Antibacterianos/farmacologia , Penicilinas/metabolismo , Penicilinas/farmacologia , beta-Lactamases , Bactérias/metabolismo , Lactonas , Oceanos e Mares
4.
Commun Biol ; 5(1): 1314, 2022 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-36451021

RESUMO

The protein networks of cell-wall-biosynthesis assemblies are largely unknown. A key class of enzymes in these assemblies is the lytic transglycosylases (LTs), of which eleven exist in P. aeruginosa. We have undertaken a pulldown strategy in conjunction with mass-spectrometry-based proteomics to identify the putative binding partners for the eleven LTs of P. aeruginosa. A total of 71 putative binding partners were identified for the eleven LTs. A systematic assessment of the binding partners of the rare lipoprotein A (RlpA), one of the pseudomonal LTs, was made. This 37-kDa lipoprotein is involved in bacterial daughter-cell separation by an unknown process. RlpA participates in both the multi-protein and multi-enzyme divisome and elongasome assemblies. We reveal an extensive protein-interaction network for RlpA involving at least 19 proteins. Their kinetic parameters for interaction with RlpA were assessed by microscale thermophoresis, surface-plasmon resonance, and isothermal-titration calorimetry. Notable RlpA binding partners include PBP1b, PBP4, and SltB1. Elucidation of the protein-interaction networks for each of the LTs, and specifically for RlpA, opens opportunities for the study of their roles in the complex protein assemblies intimately involved with the cell wall as a structural edifice critical for bacterial survival.


Assuntos
Lipoproteína(a) , Pseudomonas aeruginosa , Glicosiltransferases , Parede Celular , Mapas de Interação de Proteínas
5.
Comput Struct Biotechnol J ; 19: 5392-5405, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34667534

RESUMO

The penicillin-binding proteins are the enzyme catalysts of the critical transpeptidation crosslinking polymerization reaction of bacterial peptidoglycan synthesis and the molecular targets of the penicillin antibiotics. Here, we report a combined crystallographic, small-angle X-ray scattering (SAXS) in-solution structure, computational and biophysical analysis of PBP1 of Staphylococcus aureus (saPBP1), providing mechanistic clues about its function and regulation during cell division. The structure reveals the pedestal domain, the transpeptidase domain, and most of the linker connecting to the "penicillin-binding protein and serine/threonine kinase associated" (PASTA) domains, but not its two PASTA domains, despite their presence in the construct. To address this absence, the structure of the PASTA domains was determined at 1.5 Å resolution. Extensive molecular-dynamics simulations interpret the PASTA domains of saPBP1 as conformationally mobile and separated from the transpeptidase domain. This conclusion was confirmed by SAXS experiments on the full-length protein in solution. A series of crystallographic complexes with ß-lactam antibiotics (as inhibitors) and penta-Gly (as a substrate mimetic) allowed the molecular characterization of both inhibition by antibiotics and binding for the donor and acceptor peptidoglycan strands. Mass-spectrometry experiments with synthetic peptidoglycan fragments revealed binding by PASTA domains in coordination with the remaining domains. The observed mobility of the PASTA domain in saPBP1 could play a crucial role for in vivo interaction with its glycosyltransferase partner in the membrane or with other components of the divisome machinery, as well as for coordination of transpeptidation and polymerization processes in the bacterial divisome.

6.
ACS Pharmacol Transl Sci ; 4(3): 1204-1213, 2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-34151210

RESUMO

(R)-ND-336-designated as compound (R)-5-is a highly selective inhibitor of matrix metalloproteinase (MMP)-9 with efficacy in accelerating diabetic wound healing in murine models. (R)-ND-336 belongs to the class of thiirane inhibitors of MMPs and it is currently undergoing Investigation New Drug (IND)-enabling studies. We investigated the in vitro metabolism of (R)-ND-336 using S9 fractions obtained from mice, rats, dogs, minipigs, monkeys, and humans in order to select the rodent and nonrodent species for toxicology studies. Three metabolites were observed. One metabolite, M3, was observed across all species. Metabolite M2 was found in rats, monkeys, and humans. Metabolite M1 was observed only in rats. The identities of the metabolites were suggested by liquid chromatography/tandem mass spectroscopy (LC/MS-MS) analyses, which were authenticated by comparison to synthetic samples. Metabolites M2 and M3 arise from oxidative deamination of (R)-ND-336 by monoamine oxidase to give the arylaldehyde as a transient (and unobserved) intermediate. Reductive metabolism of this aldehyde gives the alcohol metabolite M2, while further oxidative metabolism of the aldehyde produces the carboxylate metabolite M3. A minor route of metabolism, seen only in rats, is N-acetylation of (R)-ND-336 to give the acetamide M1. The metabolism of (R)-ND-336 is distinctly different from that of the prototype member of this thiirane class ((±)-1, lacking the 4-aminomethyl aryl substituent) which is metabolized primarily by oxidation α to the sulfone to lead to a benzenesulfinate metabolite. All three metabolites are poorer MMP-9 inhibitors, compared to (R)-ND-336 (MMP-9, K i = 19 nM): M3, MMP-9 IC50 > 100 µM; M2, K i = 390 nM; and M1, IC50 > 100 µM). The rat and the minipig were selected as the rodent and nonrodent species, respectively, for toxicology studies.

7.
ACS Med Chem Lett ; 12(6): 991-995, 2021 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-34141083

RESUMO

Clostridioides difficile is a leading health threat. This pathogen initiates intestinal infections during gut microbiota dysbiosis caused by oral administration of antibiotics. C. difficile is difficult to eradicate due to its ability to form spores, which are not susceptible to antibiotics. To address the urgent need for treating recurrent C. difficile infection, antibiotics that selectively target C. difficile over common gut microbiota are needed. We herein describe the class of picolinamide antibacterials which show potent and selective activity against C. difficile. The structure-activity relationship of 108 analogues of isonicotinamide 4, a compound that is equally active against methicillin-resistant Staphylococcus aureus and C. difficile, was investigated. Introduction of the picolinamide core as exemplified by analogue 87 resulted in exquisite potency and selectivity against C. difficile. The ability of the picolinamide class to selectively target C. difficile and to prevent gut dysbiosis holds promise for the treatment of recurrent C. difficile infection.

8.
Chem Rev ; 121(6): 3412-3463, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33373523

RESUMO

The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 µm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using ß-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of ß-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the ß-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the ß-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the ß-lactams.


Assuntos
Antibacterianos/farmacologia , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação às Penicilinas/metabolismo , Staphylococcus aureus/química , beta-Lactamas/farmacologia , Animais , Antibacterianos/metabolismo , Proteínas de Bactérias/metabolismo , Biocatálise , Parede Celular/metabolismo , Cefalosporinas/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/metabolismo , Humanos , Penicilinas/farmacologia , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Resistência beta-Lactâmica , beta-Lactamas/metabolismo
9.
ACS Infect Dis ; 6(9): 2362-2368, 2020 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-32786277

RESUMO

A major challenge for chemotherapy of bacterial infections is perturbation of the intestinal microbiota. Clostridioides difficile is a Gram-positive bacterium of the gut that can thrive under this circumstance. Its production of dormant and antibiotic-impervious spores results in chronic disruption of normal gut flora and debilitating diarrhea and intestinal infection. C. difficile is responsible for 12,800 deaths per year in the United States. Here, we report the discovery of 2-(4-(3-(trifluoromethoxy)phenoxy)picolinamido)benzo[d]oxazole-5-carboxylate as an antibacterial with potent and selective activity against C. difficile. Its MIC50 and MIC90 (the concentration required to inhibit the growth of 50% and 90% of all the tested strains, respectively) values, documented across 101 strains of C. difficile, are 0.12 and 0.25 µg/mL, respectively. The compound targets cell wall biosynthesis, as assessed by macromolecular biosynthesis assays and by scanning electron microscopy. Animals infected with a lethal dose of C. difficile and treated with compound 1 had a similar survival compared to treatment with vancomycin, which is the frontline antibiotic used for C. difficile infection.


Assuntos
Clostridioides difficile , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Clostridioides , Ácidos Picolínicos , Vancomicina/farmacologia
10.
ACS Chem Biol ; 15(5): 1184-1194, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31990176

RESUMO

Gram-negative bacteria have evolved an elaborate pathway to sense and respond to exposure to ß-lactam antibiotics. The ß-lactam antibiotics inhibit penicillin-binding proteins, whereby the loss of their activities alters/damages the cell-wall peptidoglycan. Bacteria sense this damage and remove the affected peptidoglycan into complex recycling pathways. As an offshoot of these pathways, muropeptide chemical signals generated from the cell-wall recycling manifest the production of a class C ß-lactamase, which hydrolytically degrades the ß-lactam antibiotic as a resistance mechanism. We disclose the use of a fluorescence probe that detects the activation of the recycling system by the formation of the key muropeptides involved in signaling. This same probe additionally detects natural-product cell-wall-active antibiotics that are produced in situ by cohabitating bacteria.


Assuntos
Antibacterianos/metabolismo , Proteínas de Bactérias/metabolismo , Corantes Fluorescentes/química , Imagem Óptica/métodos , Pseudomonas aeruginosa/efeitos dos fármacos , beta-Lactamas/metabolismo , Produtos Biológicos/metabolismo , Parede Celular/metabolismo , Metaboloma/efeitos dos fármacos , Proteínas de Ligação às Penicilinas/metabolismo , Transdução de Sinais , Resistência beta-Lactâmica/efeitos dos fármacos
11.
Protein Sci ; 29(3): 629-646, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31747090

RESUMO

The history of modern medicine cannot be written apart from the history of the antibiotics. Antibiotics are cytotoxic secondary metabolites that are isolated from Nature. The antibacterial antibiotics disproportionately target bacterial protein structure that is distinct from eukaryotic protein structure, notably within the ribosome and within the pathways for bacterial cell-wall biosynthesis (for which there is not a eukaryotic counterpart). This review focuses on a pre-eminent class of antibiotics-the ß-lactams, exemplified by the penicillins and cephalosporins-from the perspective of the evolving mechanisms for bacterial resistance. The mechanism of action of the ß-lactams is bacterial cell-wall destruction. In the monoderm (single membrane, Gram-positive staining) pathogen Staphylococcus aureus the dominant resistance mechanism is expression of a ß-lactam-unreactive transpeptidase enzyme that functions in cell-wall construction. In the diderm (dual membrane, Gram-negative staining) pathogen Pseudomonas aeruginosa a dominant resistance mechanism (among several) is expression of a hydrolytic enzyme that destroys the critical ß-lactam ring of the antibiotic. The key sensing mechanism used by P. aeruginosa is monitoring the molecular difference between cell-wall construction and cell-wall deconstruction. In both bacteria, the resistance pathways are manifested only when the bacteria detect the presence of ß-lactams. This review summarizes how the ß-lactams are sensed and how the resistance mechanisms are manifested, with the expectation that preventing these processes will be critical to future chemotherapeutic control of multidrug resistant bacteria.


Assuntos
Antibacterianos/farmacologia , Parede Celular/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , beta-Lactamas/farmacologia , Antibacterianos/química , Farmacorresistência Bacteriana/efeitos dos fármacos , Bactérias Gram-Negativas/citologia , Bactérias Gram-Positivas/citologia , Testes de Sensibilidade Microbiana , beta-Lactamas/química
12.
ACS Chem Biol ; 15(1): 189-196, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31877028

RESUMO

BglX is a heretofore uncharacterized periplasmic glycoside hydrolase (GH) of the human pathogen Pseudomonas aeruginosa. X-ray analysis identifies it as a protein homodimer. The two active sites of the homodimer comprise catalytic residues provided by each monomer. This arrangement is seen in <2% of the hydrolases of known structure. In vitro substrate profiling shows BglX is a catalyst for ß-(1→2) and ß-(1→3) saccharide hydrolysis. Saccharides with ß-(1→4) or ß-(1→6) bonds, and the ß-(1→4) muropeptides from the cell-wall peptidoglycan, are not substrates. Additional structural insights from X-ray analysis (including structures of a mutant enzyme-derived Michaelis complex, two transition-state mimetics, and two enzyme-product complexes) enabled the comprehensive description of BglX catalysis. The half-chair (4H3) conformation of the transition-state oxocarbenium species, the approach of the hydrolytic water molecule to the oxocarbenium species, and the stepwise release of the two reaction products were also visualized. The substrate pattern for BglX aligns with the [ß-(1→2)-Glc]x and [ß-(1→3)-Glc]x periplasmic osmoregulated periplasmic glucans, and possibly with the Psl exopolysaccharides, of P. aeruginosa. Both polysaccharides are implicated in biofilm formation. Accordingly, we show that inactivation of the bglX gene of P. aeruginosa PAO1 attenuates biofilm formation.


Assuntos
Biofilmes , Glicosídeo Hidrolases/metabolismo , Peptidoglicano/metabolismo , Polissacarídeos/química , Pseudomonas aeruginosa/enzimologia , Catálise , Domínio Catalítico , Membrana Celular/metabolismo , Cristalografia por Raios X , Regulação da Expressão Gênica , Glicosídeo Hidrolases/genética , Humanos , Hidrólise , Modelos Moleculares , Mutação , Ligação Proteica , Multimerização Proteica , Pseudomonas aeruginosa/genética , Relação Estrutura-Atividade
13.
ACS Med Chem Lett ; 10(8): 1148-1153, 2019 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-31413798

RESUMO

ß-Lactams are used routinely to treat Staphylococcus aureus infections. However, the emergence of methicillin-resistant S. aureus (MRSA) renders them clinically precarious. We describe a class of cinnamonitrile adjuvants that restore the activity of oxacillin (a penicillin member of the ß-lactams) against MRSA. The lead adjuvants were tested against six important strains of MRSA, one vancomycin-intermediate S. aureus (VISA) strain, and one linezolid-resistant S. aureus strain. Five compounds out of 84 total compounds showed broad potentiation. At 8 µM (E)-3-(5-(3,4-dichlorobenzyl)-2-(trifluoromethoxy)phenyl)-2-(methylsulfonyl)acrylonitrile (26) potentiated oxacillin with a >4000-fold reduction of its MIC (from 256 to 0.06 mg·L-1). This class of adjuvants holds promise for reversal of the resistance phenotype of MRSA.

14.
ACS Chem Biol ; 14(2): 296-303, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30620575

RESUMO

The interplay between the activities of lytic transglycosylases (LTs) and penicillin-binding proteins (PBPs) is critical for the health of the bacterial cell wall. Bulgecin A (a natural-product inhibitor of LTs) potentiates the activity of ß-lactam antibiotics (inhibitors of PBPs), underscoring this intimate mechanistic interdependence. Bulgecin A in the presence of an appropriate ß-lactam causes bulge deformation due to the formation of aberrant peptidoglycan at the division site of the bacterium. As Pseudomonas aeruginosa, a nefarious human pathogen, has 11 LT paralogs, the answer as to which LT activity correlates with ß-lactam potentiation is important and is currently unknown. Growth of P. aeruginosa PAO1 strains harboring individual transposon-insertion mutants at each of the 11 genes for LTs, in the presence of the ß-lactam antibiotic ceftazidime or meropenem, implicated the gene products of slt, mltD, and mltG (of the 11), in bulge formation and potentiation. Hence, the respective enzymes would be the targets of inhibition by bulgecin A, which was indeed documented. We further demonstrated by imaging in real time and by SEM that cell lysis occurs by the structural failure of this bulge. Upon removal of the ß-lactam antibiotic prior to lysis, P. aeruginosa experiences delayed recovery from the elongation and bulge phenotype in the presence of bulgecin A. These observations argue for a collaborative role for the target LTs in the repair of the aberrant cell wall, the absence of activities of which in the presence of bulgecin A results in potentiation of the ß-lactam antibiotic.


Assuntos
Acetilglucosamina/análogos & derivados , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Prolina/análogos & derivados , Pseudomonas aeruginosa/efeitos dos fármacos , beta-Lactamas/farmacologia , Acetilglucosamina/farmacologia , Testes de Sensibilidade Microbiana , Prolina/farmacologia , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/fisiologia
16.
Biochemistry ; 57(42): 6090-6098, 2018 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-30256085

RESUMO

Lytic transglycosylases (LTs) are bacterial enzymes that catalyze the cleavage of the glycan strands of the bacterial cell wall. The mechanism of this cleavage is a remarkable intramolecular transacetalization reaction, accomplished by an ensemble of active-site residues. Because the LT reaction occurs in parallel with the cell wall bond-forming reactions catalyzed by the penicillin-binding proteins, simultaneous inhibition of both enzymes can be particularly bactericidal to Gram-negative bacteria. The MltE lytic transglycosylase is the smallest of the eight LTs encoded by the Escherichia coli genome. Prior crystallographic and computational studies identified four active-site residues-E64, S73, S75, and Y192-as playing roles in catalysis. Each of these four residues was individually altered by mutation to give four variant enzymes (E64Q, S73A, S75A, and Y192F). All four variants showed reduced catalytic activity [soluble wild type (100%) > soluble Y192F and S75A (both 40%) > S73A (4%) > E64Q (≤1%)]. The crystal structure of each variant protein was determined at the resolution of 2.12 Å for E64Q, 2.33 Å for Y192F, 1.38 Å for S73A, and 1.35 Å for S75A. These variants show alteration of the hydrogen-bond interactions of the active site. Within the framework of a prior computational study of the LT mechanism, we suggest the mechanistic role of these four active-site residues in MltE catalysis.


Assuntos
Escherichia coli K12/enzimologia , Proteínas de Escherichia coli/química , Glicosiltransferases/química , Substituição de Aminoácidos , Catálise , Domínio Catalítico , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Glicosiltransferases/genética , Mutação de Sentido Incorreto
17.
ACS Infect Dis ; 4(6): 860-867, 2018 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-29716193

RESUMO

The bulgecins are iminosaccharide secondary metabolites of the Gram-negative bacterium Paraburkholderia acidophila and inhibitors of lytic transglycosylases of bacterial cell-wall biosynthesis and remodeling. The activities of the bulgecins are intimately intertwined with the mechanism of a cobiosynthesized ß-lactam antibiotic. ß-Lactams inhibit the penicillin-binding proteins, enzymes also critical to cell-wall biosynthesis. The simultaneous loss of the lytic transglycosylase (by bulgecin) and penicillin-binding protein (by ß-lactams) activities results in deformation of the septal cell wall, observed microscopically as a bulge preceding bacterial cell lysis. We describe a practical synthesis of the three naturally occurring bulgecin iminosaccharides and their mechanistic evaluation in a series of microbiological studies. These studies identify potentiation by the bulgecin at subminimum inhibitory concentrations of the ß-lactam against three pathogenic Gram-negative bacteria and establish for the first time that this potentiation results in a significant increase in the bactericidal efficacy of a clinical ß-lactam.


Assuntos
Glicopeptídeos/síntese química , Glicopeptídeos/farmacologia , Inibidores de beta-Lactamases/síntese química , Inibidores de beta-Lactamases/farmacologia , beta-Lactamas/síntese química , beta-Lactamas/farmacologia , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Técnicas de Química Sintética , Relação Dose-Resposta a Droga , Glicopeptídeos/química , Testes de Sensibilidade Microbiana , Modelos Biológicos , Estrutura Molecular , Inibidores de beta-Lactamases/química , beta-Lactamas/química
18.
Chem Rev ; 118(12): 5952-5984, 2018 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-29847102

RESUMO

The importance of the cell wall to the viability of the bacterium is underscored by the breadth of antibiotic structures that act by blocking key enzymes that are tasked with cell-wall creation, preservation, and regulation. The interplay between cell-wall integrity, and the summoning forth of resistance mechanisms to deactivate cell-wall-targeting antibiotics, involves exquisite orchestration among cell-wall synthesis and remodeling and the detection of and response to the antibiotics through modulation of gene regulation by specific effectors. Given the profound importance of antibiotics to the practice of medicine, the assertion that understanding this interplay is among the most fundamentally important questions in bacterial physiology is credible. The enigmatic regulation of the expression of the AmpC ß-lactamase, a clinically significant and highly regulated resistance response of certain Gram-negative bacteria to the ß-lactam antibiotics, is the exemplar of this challenge. This review gives a current perspective to this compelling, and still not fully solved, 35-year enigma.


Assuntos
Parede Celular/metabolismo , Bactérias Gram-Negativas/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Parede Celular/química , Farmacorresistência Bacteriana/efeitos dos fármacos , Glicosiltransferases/química , Glicosiltransferases/metabolismo , Hexosaminidases/química , Hexosaminidases/metabolismo , Proteínas de Ligação às Penicilinas/química , Proteínas de Ligação às Penicilinas/metabolismo , beta-Lactamases/química , beta-Lactamases/metabolismo
19.
Sci Rep ; 8(1): 4110, 2018 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-29515200

RESUMO

Lytic transglycosylases (LTs) catalyze the non-hydrolytic cleavage of the bacterial cell wall by an intramolecular transacetalization reaction. This reaction is critically and broadly important in modifications of the bacterial cell wall in the course of its biosynthesis, recycling, manifestation of virulence, insertion of structural entities such as the flagellum and the pili, among others. The first QM/MM analysis of the mechanism of reaction of an LT, that for the Escherichia coli MltE, is undertaken. The study reveals a conformational itinerary consistent with an oxocarbenium-like transition state, characterized by a pivotal role for the active-site glutamic acid in proton transfer. Notably, an oxazolinium intermediate, as a potential intermediate, is absent. Rather, substrate-assisted catalysis is observed through a favorable dipole provided by the N-acetyl carbonyl group of MurNAc saccharide. This interaction stabilizes the incipient positive charge development in the transition state. This mechanism coincides with near-synchronous acetal cleavage and acetal formation.


Assuntos
Parede Celular/enzimologia , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Glicosiltransferases/química , Modelos Moleculares , Sistemas de Secreção Tipo VI/química , Domínio Catalítico , Proteínas de Escherichia coli/metabolismo , Glicosiltransferases/metabolismo , Sistemas de Secreção Tipo VI/metabolismo
20.
Chem Commun (Camb) ; 53(95): 12774-12777, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-29139490

RESUMO

The repeating isoprene unit is a fundamental biosynthetic motif. The repetitive structure presents challenges both for synthesis and for structural characterization. In this synthesis of the (Z8,E2,ω)-undecaprenol of prokaryotic glycobiology, we exemplify solutions to these challenges. Allylation of sulfone-derived carbanions controlled the stereochemistry, and its proof-of-structure was secured by Eu(hfc)3 complexation to disperse the overlaid resonances of its 1H NMR spectrum.


Assuntos
Espectroscopia de Prótons por Ressonância Magnética/métodos , Terpenos/química , Terpenos/síntese química , Estrutura Molecular , Padrões de Referência , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...