Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 9(11)2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158165

RESUMO

Peptidoglycan (PG) is made of a polymer of disaccharides organized as a three-dimensional mesh-like network connected together by peptidic cross-links. PG is a dynamic structure that is essential for resistance to environmental stressors. Remodeling of PG occurs throughout the bacterial life cycle, particularly during bacterial division and separation into daughter cells. Numerous autolysins with various substrate specificities participate in PG remodeling. Expression of these enzymes must be tightly regulated, as an excess of hydrolytic activity can be detrimental for the bacteria. In non-tuberculous mycobacteria such as Mycobacterium abscessus, the function of PG-modifying enzymes has been poorly investigated. In this study, we characterized the function of the PG amidase, Ami1 from M. abscessus. An ami1 deletion mutant was generated and the phenotypes of the mutant were evaluated with respect to susceptibility to antibiotics and virulence in human macrophages and zebrafish. The capacity of purified Ami1 to hydrolyze muramyl-dipeptide was demonstrated in vitro. In addition, the screening of a 9200 compounds library led to the selection of three compounds inhibiting Ami1 in vitro. We also report the structural characterization of Ami1 which, combined with in silico docking studies, allows us to propose a mode of action for these inhibitors.


Assuntos
Mycobacterium abscessus/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Animais , Cristalografia por Raios X , Modelos Animais de Doenças , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Deleção de Genes , Humanos , Larva/microbiologia , Macrófagos/microbiologia , Testes de Sensibilidade Microbiana , Simulação de Acoplamento Molecular , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium abscessus/patogenicidade , Mycobacterium abscessus/ultraestrutura , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Fenótipo , Homologia Estrutural de Proteína , Células THP-1 , Virulência , Peixe-Zebra
2.
Nature ; 578(7796): 582-587, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32051588

RESUMO

Addressing the ongoing antibiotic crisis requires the discovery of compounds with novel mechanisms of action that are capable of treating drug-resistant infections1. Many antibiotics are sourced from specialized metabolites produced by bacteria, particularly those of the Actinomycetes family2. Although actinomycete extracts have traditionally been screened using activity-based platforms, this approach has become unfavourable owing to the frequent rediscovery of known compounds. Genome sequencing of actinomycetes reveals an untapped reservoir of biosynthetic gene clusters, but prioritization is required to predict which gene clusters may yield promising new chemical matter2. Here we make use of the phylogeny of biosynthetic genes along with the lack of known resistance determinants to predict divergent members of the glycopeptide family of antibiotics that are likely to possess new biological activities. Using these predictions, we uncovered two members of a new functional class of glycopeptide antibiotics-the known glycopeptide antibiotic complestatin and a newly discovered compound we call corbomycin-that have a novel mode of action. We show that by binding to peptidoglycan, complestatin and corbomycin block the action of autolysins-essential peptidoglycan hydrolases that are required for remodelling of the cell wall during growth. Corbomycin and complestatin have low levels of resistance development and are effective in reducing bacterial burden in a mouse model of skin MRSA infection.


Assuntos
Antibacterianos , Descoberta de Drogas , Peptídeos Cíclicos , Peptidoglicano/efeitos dos fármacos , Peptidoglicano/metabolismo , Actinobacteria/química , Actinobacteria/genética , Actinobacteria/metabolismo , Animais , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Vias Biossintéticas/genética , Parede Celular/metabolismo , Clorofenóis/química , Clorofenóis/metabolismo , Clorofenóis/farmacologia , Modelos Animais de Doenças , Resistência Microbiana a Medicamentos/efeitos dos fármacos , Resistência Microbiana a Medicamentos/genética , Feminino , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Família Multigênica , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Peptídeos Cíclicos/farmacologia , Filogenia , Pele/microbiologia , Infecções Estafilocócicas/microbiologia
3.
J Biol Chem ; 295(10): 3347-3361, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31974163

RESUMO

Bacteria account for 1000-fold more biomass than humans. They vary widely in shape and size. The morphological diversity of bacteria is due largely to the different peptidoglycan-based cell wall structures that encase bacterial cells. Although the basic structure of peptidoglycan is highly conserved, consisting of long glycan strands that are cross-linked by short peptide chains, the mature cell wall is chemically diverse. Peptidoglycan hydrolases and cell wall-tailoring enzymes that regulate glycan strand length, the degree of cross-linking, and the addition of other modifications to peptidoglycan are central in determining the final architecture of the bacterial cell wall. Historically, it has been difficult to biochemically characterize these enzymes that act on peptidoglycan because suitable peptidoglycan substrates were inaccessible. In this review, we discuss fundamental aspects of bacterial cell wall synthesis, describe the regulation and diverse biochemical and functional activities of peptidoglycan hydrolases, and highlight recently developed methods to make and label defined peptidoglycan substrates. We also review how access to these substrates has now enabled biochemical studies that deepen our understanding of how bacterial cell wall enzymes cooperate to build a mature cell wall. Such improved understanding is critical to the development of new antibiotics that disrupt cell wall biogenesis, a process essential to the survival of bacteria.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Proteínas de Bactérias/agonistas , Proteínas de Bactérias/antagonistas & inibidores , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , N-Acetil-Muramil-L-Alanina Amidase/química , Peptidoglicano/química , Peptidoglicano/metabolismo , Estrutura Terciária de Proteína , Staphylococcus aureus/enzimologia , Especificidade por Substrato
4.
Infect Immun ; 87(8)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31182616

RESUMO

The peptidoglycan in Gram-negative bacteria is a dynamic structure in constant remodeling. This dynamism, achieved through synthesis and degradation, is essential because the peptidoglycan is necessary to maintain the structure of the cell but has to have enough plasticity to allow the transport and assembly of macromolecular complexes in the periplasm and outer membrane. In addition, this remodeling has to be coordinated with the division process. Among the multiple mechanisms bacteria have to degrade the peptidoglycan are the lytic transglycosidases, enzymes of the lysozyme family that cleave the glycan chains generating gaps in the mesh structure increasing its permeability. Because these enzymes can act as autolysins, their activity has to be tightly regulated, and one of the mechanisms bacteria have evolved is the synthesis of membrane bound or periplasmic inhibitors. In the present study, we identify a periplasmic lytic transglycosidase inhibitor (PhiA) in Brucella abortus and demonstrate that it inhibits the activity of SagA, a lytic transglycosidase we have previously shown is involved in the assembly of the type IV secretion system. A phiA deletion mutant results in a strain with the incapacity to synthesize a complete lipopolysaccharide but with a higher replication rate than the wild-type parental strain, suggesting a link between peptidoglycan remodeling and speed of multiplication.


Assuntos
Brucella abortus/patogenicidade , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Glicosídeo Hidrolases/fisiologia , Lipopolissacarídeos/biossíntese , Complexos Multienzimáticos/fisiologia , Peptidoglicano/metabolismo , Transferases/fisiologia , Sistemas de Secreção Tipo IV/fisiologia , Virulência
5.
Bioorg Med Chem ; 27(5): 721-728, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30711310

RESUMO

New compounds able to counteract staphylococcal biofilm formation are needed. In this study we investigate the mechanism of action of pyrrolomycins, whose potential as antimicrobial agents has been demonstrated. We performed a new efficient and easy method to use microwave organic synthesis suitable for obtaining pyrrolomycins in good yields and in suitable amount for their in vitro in-depth investigation. We evaluate the inhibitory activity towards Sortase A (SrtA), a transpeptidase responsible for covalent anchoring in Gram-positive peptidoglycan of many surface proteins involved in adhesion and in biofilm formation. All compounds show a good inhibitory activity toward SrtA, having IC50 values ranging from 130 to 300 µM comparable to berberine hydrochloride. Of note compound 1d shows a good affinity in docking experiment to SrtA and exhibits the highest capability to interfere with biofilm formation of S. aureus showing an IC50 of 3.4 nM. This compound is also effective in altering S. aureus murein hydrolase activity that is known to be responsible for degradation, turnover, and maturation of bacterial peptidoglycan and involved in the initial stages of S. aureus biofilm formation.


Assuntos
Antibacterianos/farmacologia , Pirróis/farmacologia , Aminoaciltransferases/química , Aminoaciltransferases/metabolismo , Antibacterianos/síntese química , Antibacterianos/farmacocinética , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Biofilmes/efeitos dos fármacos , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Ensaios Enzimáticos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Testes de Sensibilidade Microbiana , Micro-Ondas , Simulação de Acoplamento Molecular , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Pseudomonas aeruginosa/efeitos dos fármacos , Pirróis/síntese química , Pirróis/farmacocinética , Staphylococcus aureus/efeitos dos fármacos
6.
J Enzyme Inhib Med Chem ; 33(1): 1239-1247, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30141354

RESUMO

Autolysin E (AtlE) is a cell wall degrading enzyme that catalyzes the hydrolysis of the ß-1,4-glycosidic bond between the N-acetylglucosamine and N-acetylmuramic acid units of the bacterial peptidoglycan. Using our recently determined crystal structure of AtlE from Staphylococcus aureus and a combination of pharmacophore modeling, similarity search, and molecular docking, a series of (Phenylureido)piperidinyl benzamides were identified as potential binders and surface plasmon resonance (SPR) and saturation-transfer difference (STD) NMR experiments revealed that discovered compounds bind to AtlE in a lower micromolar range. (phenylureido)piperidinyl benzamides are the first reported non-substrate-like compounds that interact with this enzyme and enable further study of the interaction of small molecules with bacterial AtlE as potential inhibitors of this target.


Assuntos
Antibacterianos/farmacologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Piperidinas/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/síntese química , Antibacterianos/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Piperidinas/síntese química , Piperidinas/química , Staphylococcus aureus/enzimologia , Relação Estrutura-Atividade
7.
SAR QSAR Environ Res ; 29(9): 647-660, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30160524

RESUMO

A structure-based approach is applied for the development of inhibitors of bacterial N-acetyglucosaminidase (autolysin). Autolysins are enzymes involved in the degradation of peptidoglycan and therefore participate in bacterial cell growth and different lysis phenomena. Several studies indicate that by the inhibition of autolysins, and consequently of bacterial cell division, antibacterial activity can be obtained, thus paving the road to a novel group of therapeutics against human pathogens. As crystal structures of the autolysin E (AtlE)-ligand complexes were obtained in our laboratories, fragment-based virtual screening was the method of choice for the initial studies. Fragment libraries from various databases were merged to increase the number of compounds for the virtual screening. Twenty-four commercially available virtual hits were selected and subjected to quantitative analysis of binding interactions using the surface plasmon resonance technique. Twelve fragments showed fragment-AtlE interactions. For F1, the top hit of the virtual screening, a KD of 228 µM was determined, while other fragments displayed non-stoichiometric binding. Blind docking of potential binders uncovers three possible allosteric sites. Ligands of N-acetyglucosaminidase identified in our study represent valuable information for the further development of AtlE inhibitors, which could in future represent antibacterial agents acting by a novel mode of action.


Assuntos
Acetilglucosaminidase/antagonistas & inibidores , Antibacterianos/química , Proteínas de Bactérias/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Relação Quantitativa Estrutura-Atividade , Modelos Moleculares , Simulação de Acoplamento Molecular , Bibliotecas de Moléculas Pequenas
8.
SAR QSAR Environ Res ; 27(7): 573-87, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27686112

RESUMO

Autolysin E (AtlE) is a bacteriolytic enzyme which plays an important role in division and growth of bacterial cells and therefore represents a promising potential drug target. Its 3D structure has been recently elucidated. We used in silico prediction tools to study substrate or ligand (inhibitor) binding regions of AtlE. We applied several freely available tools and a commercial tool for binding site identification and compared results of the prediction. Calculation time, number of predictions and output data provided by specific software vary according to the different approaches utilized by specific method categories. Despite different approaches, binding sites in similar locations on the protein were predicted. Specific amino acid residues that form these binding sites were predicted as binding residues. The predicted residues, especially those with predicted highest conservation score, could theoretically have catalytic and binding properties. According to our results, we assume that E138, which has the highest conservation score, is the catalytic residue and F161, G162 and Y224, which are also highly conserved, are responsible for substrate binding. Ligands developed with binding specificity towards these residues could inhibit the catalysis and binding of the substrate of AtlE. The molecules with inhibitory potency could therefore represent potential new antibacterial agents.


Assuntos
N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , N-Acetil-Muramil-L-Alanina Amidase/química , Sítios de Ligação , Simulação por Computador , Desenho de Fármacos , Ligantes , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Relação Quantitativa Estrutura-Atividade
9.
Antimicrob Agents Chemother ; 58(9): 5164-80, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24957840

RESUMO

In this study, our objective was to determine whether a synergistic antimicrobial combination in vitro would be beneficial in the downregulation of pneumococcal virulence genes and whether the associated inflammation of the lung tissue induced by multidrug-resistant Streptococcus pneumoniae infection in vivo needs to be elucidated in order to consider this mode of therapy in case of severe pneumococcal infection. We investigated in vivo changes in the expression of these virulence determinants using an efficacious combination determined in previous studies. BALB/c mice were infected with 10(6) CFU of bacteria. Intravenous levofloxacin at 150 mg/kg and/or ceftriaxone at 50 mg/kg were initiated 18 h postinfection; the animals were sacrificed 0 to 24 h after the initiation of treatment. The levels of cytokines, chemokines, and C-reactive protein (CRP) in the serum and lungs, along with the levels of myeloperoxidase and nitric oxide the inflammatory cell count in bronchoalveolar lavage fluid (BALF), changes in pneumolysin and autolysin gene expression and COX-2 and inducible nitric oxide synthase (iNOS) protein expression in the lungs were estimated. Combination therapy downregulated inflammation and promoted bacterial clearance. Pneumolysin and autolysin expression was downregulated, with a concomitant decrease in the expression of COX-2 and iNOS in lung tissue. Thus, the combination of levofloxacin and ceftriaxone can be considered for therapeutic use even in cases of pneumonia caused by drug-resistant isolates.


Assuntos
Ceftriaxona/farmacologia , Levofloxacino/farmacologia , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Infecções Pneumocócicas/tratamento farmacológico , Pneumonia Pneumocócica/tratamento farmacológico , Pneumonia/tratamento farmacológico , Streptococcus pneumoniae/efeitos dos fármacos , Estreptolisinas/antagonistas & inibidores , Animais , Antibacterianos/farmacologia , Bacteriemia/tratamento farmacológico , Bacteriemia/metabolismo , Bacteriemia/microbiologia , Proteínas de Bactérias/antagonistas & inibidores , Líquido da Lavagem Broncoalveolar/microbiologia , Modelos Animais de Doenças , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Quimioterapia Combinada/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Infecções Pneumocócicas/metabolismo , Infecções Pneumocócicas/microbiologia , Pneumonia/metabolismo , Pneumonia/microbiologia , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/microbiologia , Streptococcus pneumoniae/metabolismo , Virulência/efeitos dos fármacos , Fatores de Virulência/metabolismo
10.
PLoS Pathog ; 9(2): e1003197, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23468634

RESUMO

Peptidoglycan hydrolases are a double-edged sword. They are required for normal cell division, but when dysregulated can become autolysins lethal to bacteria. How bacteria ensure that peptidoglycan hydrolases function only in the correct spatial and temporal context remains largely unknown. Here, we demonstrate that dysregulation converts the essential mycobacterial peptidoglycan hydrolase RipA to an autolysin that compromises cellular structural integrity. We find that mycobacteria control RipA activity through two interconnected levels of regulation in vivo-protein interactions coordinate PG hydrolysis, while proteolysis is necessary for RipA enzymatic activity. Dysregulation of RipA protein complexes by treatment with a peptidoglycan synthase inhibitor leads to excessive RipA activity and impairment of correct morphology. Furthermore, expression of a RipA dominant negative mutant or of differentially processed RipA homologues reveals that RipA is produced as a zymogen, requiring proteolytic processing for activity. The amount of RipA processing differs between fast-growing and slow-growing mycobacteria and correlates with the requirement for peptidoglycan hydrolase activity in these species. Together, the complex picture of RipA regulation is a part of a growing paradigm for careful control of cell wall hydrolysis by bacteria during growth, and may represent a novel target for chemotherapy development.


Assuntos
Parede Celular/enzimologia , Complexos Multienzimáticos/metabolismo , Mycobacterium smegmatis/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Divisão Celular , DNA Bacteriano/análise , Inibidores Enzimáticos/farmacologia , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/ultraestrutura , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Proteólise
11.
Antimicrob Agents Chemother ; 56(11): 5804-10, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22926575

RESUMO

Due to their abilities to form strong biofilms, Staphylococcus aureus and Staphylococcus epidermidis are the most frequently isolated pathogens in persistent and chronic implant-associated infections. As biofilm-embedded bacteria are more resistant to antibiotics and the immune system, they are extremely difficult to treat. Therefore, biofilm-active antibiotics are a major challenge. Here we investigated the effect of the lantibiotic gallidermin on two representative biofilm-forming staphylococcal species. Gallidermin inhibits not only the growth of staphylococci in a dose-dependent manner but also efficiently prevents biofilm formation by both species. The effect on biofilm might be due to repression of biofilm-related targets, such as ica (intercellular adhesin) and atl (major autolysin). However, gallidermin's killing activity on 24-h and 5-day-old biofilms was significantly decreased. A subpopulation of 0.1 to 1.0% of cells survived, comprising "persister" cells of an unknown genetic and physiological state. Like many other antibiotics, gallidermin showed only limited activity on cells within mature biofilms.


Assuntos
Bacteriocinas/farmacologia , Biofilmes/efeitos dos fármacos , Peptídeos/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus epidermidis/efeitos dos fármacos , Adesinas Bacterianas/metabolismo , Biofilmes/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Staphylococcus aureus/crescimento & desenvolvimento , Staphylococcus epidermidis/crescimento & desenvolvimento
12.
Microb Cell Fact ; 10: 62, 2011 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-21813023

RESUMO

BACKGROUND: Through modification of the flagellin type III secretion pathway of Bacillus halodurans heterologous peptides could be secreted into the medium as flagellin fusion monomers. The stability of the secreted monomers was significantly enhanced through gene-targeted inactivation of host cell extracellular proteases. In evaluating the biotechnological potential of this extracellular secretion system an anti-viral therapeutic peptide, Enfuvirtide, was chosen. Currently, Enfuvirtide is synthesised utilizing 106 chemical steps. We used Enfuvirtide as a model system in an effort to develop a more cost-effective biological process for therapeutic peptide production. RESULTS: An attempt was made to increase the levels of the fusion peptide by two strategies, namely strain improvement through gene-targeted knock-outs, as well as vector and cassette optimization. Both approaches proved to be successful. Through chromosomal inactivation of the spo0A, lytC and lytE genes, giving rise to strain B. halodurans BhFDL05S, the secretion of recombinant peptide fusions was increased 10-fold. Cassette optimization, incorporating an expression vector pNW33N and the N- and C-terminal regions of the flagellin monomer as an in-frame peptide fusion, resulted in a further 3.5-fold increase in the secretion of recombinant peptide fusions. CONCLUSIONS: The type III flagellar secretion system of B. halodurans has been shown to successfully secrete a therapeutic peptide as a heterologous flagellin fusion. Improvements to both the strain and expression cassette led to increased levels of recombinant peptide, showing promise for a biotechnological application.


Assuntos
Bacillus/metabolismo , Flagelina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Enfuvirtida , Flagelina/genética , Técnicas de Inativação de Genes , Proteína gp41 do Envelope de HIV/biossíntese , Proteína gp41 do Envelope de HIV/genética , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , N-Acetil-Muramil-L-Alanina Amidase/genética , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Fragmentos de Peptídeos/biossíntese , Fragmentos de Peptídeos/genética , Proteínas Recombinantes de Fusão/genética , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
FEBS J ; 274(2): 364-76, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17229144

RESUMO

Streptococcus pneumoniae is one of the major pathogens worldwide. The use of currently available antibiotics to treat pneumococcal diseases is hampered by increasing resistance levels; also, capsular polysaccharide-based vaccination is of limited efficacy. Therefore, it is desirable to find targets for the development of new antimicrobial drugs specifically designed to fight pneumococcal infections. Choline-binding proteins are a family of polypeptides, found in all S. pneumoniae strains, that take part in important physiologic processes of this bacterium. Among them are several murein hydrolases whose enzymatic activity is usually inhibited by an excess of choline. Using a simple chromatographic procedure, we have identified several choline analogs able to strongly interact with the choline-binding module (C-LytA) of the major autolysin of S. pneumoniae. Two of these compounds (atropine and ipratropium) display a higher binding affinity to C-LytA than choline, and also increase the stability of the protein. CD and fluorescence spectroscopy analyses revealed that the conformational changes of C-LytA upon binding of these alkaloids are different to those induced by choline, suggesting a different mode of binding. In vitro inhibition assays of three pneumococcal, choline-dependent cell wall lytic enzymes also demonstrated a greater inhibitory efficiency of those molecules. Moreover, atropine and ipratropium strongly inhibited in vitro pneumococcal growth, altering cell morphology and reducing cell viability, a very different response than that observed upon addition of an excess of choline. These results may open up the possibility of the development of bicyclic amines as new antimicrobials for use against pneumococcal pathologies.


Assuntos
Aminas/química , Hidrolases de Éster Carboxílico/química , N-Acetil-Muramil-L-Alanina Amidase/química , Streptococcus pneumoniae/metabolismo , Atropina/química , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Proliferação de Células , Parede Celular/metabolismo , Celulose/química , Colina/química , Dicroísmo Circular , Dimerização , Etanolaminas/química , Ipratrópio/química , Modelos Químicos , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Desnaturação Proteica , Espectrometria de Fluorescência
14.
J Biol Chem ; 280(20): 19948-57, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15769740

RESUMO

The search for new drugs against Streptococcus pneumoniae (pneumococcus) is driven by the 1.5 million deaths it causes annually. Choline-binding proteins attach to the pneumococcal cell wall through domains that recognize choline moieties, and their involvement in pneumococcal virulence makes them potential targets for drug development. We have defined chemical criteria involved in the docking of small molecules from a three-dimensional structural library to the major pneumococcal autolysin (LytA) choline binding domain. These criteria were used to identify compounds that could interfere with the attachment of this protein to the cell wall, and several quinolones that fit this framework were found to inhibit the cell wall-degrading activity of LytA. Furthermore, these compounds produced similar effects on other enzymes with different catalytic activities but that contained a similar choline binding domain; that is, autolysin (LytC) and the phage lytic enzyme (Cpl-1). Finally, we resolved the crystal structure of the complex between the choline binding domain of LytA and ofloxacin at a resolution of 2.6 Angstroms. These data constitute an important launch pad from which effective drugs to combat pneumococcal infections can be developed.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Ofloxacino/química , Ofloxacino/farmacologia , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/patogenicidade , Bacteriólise/efeitos dos fármacos , Sítios de Ligação , Parede Celular/efeitos dos fármacos , Cristalografia por Raios X , Desenho de Fármacos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , Estrutura Terciária de Proteína , Streptococcus pneumoniae/enzimologia , Termodinâmica , Virulência/efeitos dos fármacos
15.
J Infect Dis ; 188(7): 1040-8, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-14513425

RESUMO

The Streptococcus pneumoniae cell-wall components peptidoglycan and lipoteichoic acid activate Toll-like receptor 2 (TLR2), which transduces an inflammatory response. After exposure to penicillin, type 2 S. pneumoniae strain D39, but not the isogenic autolysin-deficient mutant AL2, induced significantly enhanced interleukin-8 promoter activity in TLR2-transfected HeLa cells. Lag-phase D39 exhibited enhanced TLR2 activation after exposure to penicillin at levels below the minimum inhibitory concentration (MIC); in contrast, early log-phase S. pneumoniae were most active when exposed to the MIC. This enhancement was not ablated by heat treatment but was attenuated by autolysin inhibitors. The antimicrobial activity of moxifloxacin and erythromycin was not associated with TLR2 activation by S. pneumoniae. These data show that penicillin treatment of S. pneumoniae releases proinflammatory cell-wall components that activate TLR2 and that this activity is dependent on autolysin, the growth phase of the organism, and the antibiotic concentration.


Assuntos
Compostos Aza , Fluoroquinolonas , Glicoproteínas de Membrana/imunologia , Penicilinas/farmacologia , Infecções Pneumocócicas/imunologia , Quinolinas , Receptores de Superfície Celular/imunologia , Streptococcus pneumoniae/imunologia , Anti-Infecciosos/farmacologia , Colina/farmacologia , Contagem de Colônia Microbiana , Inibidores Enzimáticos/farmacologia , Eritromicina/farmacologia , Etanolamina/farmacologia , Células HeLa , Humanos , Interleucina-8/imunologia , Interleucina-8/metabolismo , Moxifloxacina , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , N-Acetil-Muramil-L-Alanina Amidase/imunologia , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Infecções Pneumocócicas/tratamento farmacológico , Streptococcus pneumoniae/efeitos dos fármacos , Receptor 2 Toll-Like , Receptores Toll-Like , Transfecção
16.
J Bacteriol ; 179(9): 2958-62, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9139914

RESUMO

We investigated the cell surface localization of the atl gene products of Staphylococcus aureus exposed to a lytic concentration (4 MIC) of penicillin G (PCG) by means of immunoelectron microscopy using anti-62-kDa N-acetylmuramyl-L-alanine amidase or anti-51-kDa endo-beta-N-acetylglucosaminidase immunoglobulin G. Protein A-gold conjugates reacting with antigen-antibody complex localized at sites of defects of the cell wall at the nascent cross wall. Anti-62-kDa N-acetylmuramyl-L-alanine amidase or anti-51-kDa endo-beta-N-acetylglucosaminidase immunoglobulin G inhibited the decreased turbidity caused by PCG-induced lysis and the formation of defects in the wall. The autolysis-defective mutant, S. aureus RUSAL2 (atl::Tn551), exposed to 4 MIC of PCG resisted autolysis and formation of the wall defect. These results suggest that activation or deregulation of the atl gene products at localized sites where formation of new cross wall was disturbed by PCG causes small defects in the cell wall in situ, eventually leading to general autolysis.


Assuntos
N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Penicilina G/farmacologia , Staphylococcus aureus/fisiologia , Anticorpos/farmacologia , Parede Celular/fisiologia , Parede Celular/ultraestrutura , Cinética , Microscopia Imunoeletrônica , Modelos Biológicos , Mutação , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , N-Acetil-Muramil-L-Alanina Amidase/genética , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética
17.
Microb Drug Resist ; 2(1): 131-4, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-9158735

RESUMO

The effects of energy uncouplers on in vivo and in vitro peptidoglycan hydrolase activities in Escherichia coli were determined. Sodium azide, potassium cyanide, and carbonyl cyanide m-chlorophenylhydrazone all inhibited ampicillin-induced lysis of exponential phase cultures, even when they were added to lysis-committed cultures. These energy uncouplers also inhibited the solubilization of radiolabeled peptidoglycan by bacterial suspensions that had been treated with 5% trichloroacetic acid by the method of Hartmann et al.3 to activate the peptidoglycan hydrolases. Therefore, the in vivo and in vitro activities of peptidoglycan hydrolases in E. coli are dependent on membrane energization.


Assuntos
Metabolismo Energético/fisiologia , Escherichia coli/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Desacopladores/farmacologia , Ampicilina/antagonistas & inibidores , Ampicilina/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Metabolismo Energético/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Escherichia coli/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Testes de Sensibilidade Microbiana , Penicilinas/antagonistas & inibidores , Penicilinas/farmacologia
18.
Eur J Biochem ; 235(3): 601-5, 1996 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8654407

RESUMO

A chimeric trifunctional pneumococcal peptidoglycan hydrolase (CHL) has been constructed by fusing a choline-binding domain with two catalytic modules that provide lysozyme and amidase activity. The chimeric enzymes behaves as a choline-dependent enzyme and its activity is comparable to that of the parent enzymes. Site-directed mutagenesis of CHL produced a mutated enzyme [D9A,36A]CHL) that only exhibited an amidase activity. Comparative biochemical analyses of CHL and [D9A, E36A]CHL strongly suggest that the lysozyme catalytic module confers 88% of the total activity of CHL, whereas 12% of the activity can be ascribed to the amidase module. Both enzymatic activities are affected by the process of activation or conversion induced by choline suggesting that the conversion process is produced by a conformational change in the choline-binding domain. Our findings demonstrate that the three modules can acquire the proper folding conformation in the-three domain chimeric CHL enzyme. This experimental evidence supports the modular theory of protein evolution, and demonstrates that modular assembly of functional domains can be a rational approach to construct fully active chimeric enzymes with novel biological or biotechnological properties.


Assuntos
N-Acetil-Muramil-L-Alanina Amidase/genética , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Streptococcus pneumoniae/genética , Amidoidrolases/metabolismo , Vetores Genéticos , Muramidase/antagonistas & inibidores , Muramidase/metabolismo , Mutagênese Sítio-Dirigida , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Streptococcus pneumoniae/enzimologia
19.
J Bacteriol ; 176(15): 4784-9, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7913931

RESUMO

Two extracellular autolysins have been detected in the spent culture supernatants of Pseudomonas aeruginosa PAO1 by using renaturing polyacrylamide gel electrophoresis. The two autolysins were isolated from the culture supernatant by trichloroacetic acid precipitation and were shown to have apparent molecular masses of 26 and 29 kDa. The 26-kDa autolysin first appears during the early exponential phase of growth and then declines sharply, while the 29-kDa autolysin first appears in the late exponential phase of growth and continues well into the stationary phase. Fractionation of whole cells indicated that the 26-kDa enzyme was also localized within the periplasm, with a lesser amount of activity associated with the cytoplasmic membrane. The 29-kDa autolytic activity was distributed within the cell equally between the periplasm and the cytoplasmic membrane. The pH optima of the isolated 26- and 29-kDa autolysins are 6.0 and 5.0, respectively. Further evidence from both protease susceptibility and inhibition studies confirms that these two extracellular autolysins isolated from P. aeruginosa PAO1 are separate and distinct.


Assuntos
N-Acetil-Muramil-L-Alanina Amidase/isolamento & purificação , Pseudomonas aeruginosa/enzimologia , Concentração de Íons de Hidrogênio , N-Acetil-Muramil-L-Alanina Amidase/antagonistas & inibidores , N-Acetil-Muramil-L-Alanina Amidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...