Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Pharmacol Ther ; 109(4): 1000-1020, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33576025

RESUMO

Multidrug-resistant bacteria are causing a serious global health crisis. A dramatic decline in antibiotic discovery and development investment by pharmaceutical industry over the last decades has slowed the adoption of new technologies. It is imperative that we create new mechanistic insights based on latest technologies, and use translational strategies to optimize patient therapy. Although drug development has relied on minimal inhibitory concentration testing and established in vitro and mouse infection models, the limited understanding of outer membrane permeability in Gram-negative bacteria presents major challenges. Our team has developed a platform using the latest technologies to characterize target site penetration and receptor binding in intact bacteria that inform translational modeling and guide new discovery. Enhanced assays can quantify the outer membrane permeability of ß-lactam antibiotics and ß-lactamase inhibitors using multiplex liquid chromatography tandem mass spectrometry. While ß-lactam antibiotics are known to bind to multiple different penicillin-binding proteins (PBPs), their binding profiles are almost always studied in lysed bacteria. Novel assays for PBP binding in the periplasm of intact bacteria were developed and proteins identified via proteomics. To characterize bacterial morphology changes in response to PBP binding, high-throughput flow cytometry and time-lapse confocal microscopy with fluorescent probes provide unprecedented mechanistic insights. Moreover, novel assays to quantify cytosolic receptor binding and intracellular drug concentrations inform target site occupancy. These mechanistic data are integrated by quantitative and systems pharmacology modeling to maximize bacterial killing and minimize resistance in in vitro and mouse infection models. This translational approach holds promise to identify antibiotic combination dosing strategies for patients with serious infections.


Assuntos
Técnicas Bacteriológicas/métodos , Descoberta de Drogas/métodos , Farmacorresistência Bacteriana Múltipla/fisiologia , Bactérias Gram-Negativas/efeitos dos fármacos , Bactérias Gram-Negativas/fisiologia , Animais , Membrana Celular/fisiologia , Modelos Animais de Doenças , Humanos , Modelos Teóricos , Proteínas de Ligação às Penicilinas/fisiologia , beta-Lactamas/farmacologia
2.
Sci Rep ; 10(1): 12588, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32724139

RESUMO

Chlamydia trachomatis serovar L2 and Chlamydia muridarum, which do not express FtsZ, undergo polarized cell division. During division, peptidoglycan assembles at the pole of dividing Chlamydia trachomatis cells where daughter cell formation occurs, and peptidoglycan regulates at least two distinct steps in the polarized division of Chlamydia trachomatis and Chlamydia muridarum. Cells treated with inhibitors that prevent peptidoglycan synthesis or peptidoglycan crosslinking by penicillin-binding protein 2 (PBP2) are unable to initiate polarized division, while cells treated with inhibitors that prevent peptidoglycan crosslinking by penicillin-binding protein 3 (PBP3/FtsI) initiate polarized division, but the process arrests at an early stage of daughter cell growth. Consistent with their distinct roles in polarized division, peptidoglycan organization is different in cells treated with PBP2 and PBP3-specific inhibitors. Our analyses indicate that the sequential action of PBP2 and PBP3 drives changes in peptidoglycan organization that are essential for the polarized division of these obligate intracellular bacteria. Furthermore, the roles we have characterized for PBP2 and PBP3 in regulating specific steps in chlamydial cell division have not been described in other bacteria.


Assuntos
Divisão Celular/fisiologia , Chlamydia trachomatis/citologia , Proteínas de Ligação às Penicilinas/fisiologia , Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Peptidoglicano/biossíntese , Peptidoglicano/metabolismo
3.
Int J Mol Sci ; 21(12)2020 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-32630428

RESUMO

Peptidoglycan is generally considered one of the main determinants of cell shape in bacteria. In rod-shaped bacteria, cell elongation requires peptidoglycan synthesis to lengthen the cell wall. In addition, peptidoglycan is synthesized at the division septum during cell division. Sporulation of Bacillus subtilis begins with an asymmetric cell division. Formation of the sporulation septum requires almost the same set of proteins as the vegetative septum; however, these two septa are significantly different. In addition to their differences in localization, the sporulation septum is thinner and it contains SpoIIE, a crucial sporulation specific protein. Here we show that peptidoglycan biosynthesis is linked to the cell division machinery during sporulation septum formation. We detected a direct interaction between SpoIIE and GpsB and found that both proteins co-localize during the early stages of asymmetric septum formation. We propose that SpoIIE is part of a multi-protein complex which includes GpsB, other division proteins and peptidoglycan synthesis proteins, and could provide a link between the peptidoglycan synthesis machinery and the complex morphological changes required for forespore formation during B. subtilis sporulation.


Assuntos
Bacillus subtilis/metabolismo , Proteínas de Ligação às Penicilinas/metabolismo , Esporos Bacterianos/metabolismo , Divisão Celular Assimétrica/fisiologia , Proteínas de Bactérias/metabolismo , Ciclo Celular , Divisão Celular/fisiologia , Forma Celular , Parede Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação às Penicilinas/fisiologia , Peptidoglicano/metabolismo , Esporos Bacterianos/fisiologia
4.
Artigo em Inglês | MEDLINE | ID: mdl-32041714

RESUMO

Enterococcus faecalis and Enterococcus faecium are commensals of the gastrointestinal tract of most terrestrial organisms, including humans, and are major causes of health care-associated infections. Such infections are difficult or impossible to treat, as the enterococcal strains responsible are often resistant to multiple antibiotics. One intrinsic resistance trait that is conserved among E. faecalis and E. faecium is cephalosporin resistance, and prior exposure to cephalosporins is one of the most well-known risk factors for acquisition of an enterococcal infection. Cephalosporins inhibit peptidoglycan biosynthesis by acylating the active-site serine of penicillin-binding proteins (PBPs) to prevent the PBPs from catalyzing cross-linking during peptidoglycan synthesis. For decades, a specific PBP (known as Pbp4 or Pbp5) that exhibits low reactivity toward cephalosporins has been thought to be the primary PBP required for cephalosporin resistance. We analyzed other PBPs and report that in both E. faecalis and E. faecium, a second PBP, PbpA(2b), is also required for resistance; notably, the cephalosporin ceftriaxone exhibits a lethal effect on the ΔpbpA mutant. Strikingly, PbpA(2b) exhibits low intrinsic reactivity with cephalosporins in vivo and in vitro Unlike the Δpbp5 mutant, the ΔpbpA mutant exhibits a variety of phenotypic defects in growth kinetics, cell wall integrity, and cellular morphology, indicating that PbpA(2b) and Pbp5(4) are not functionally redundant and that PbpA(2b) plays a more central role in peptidoglycan synthesis. Collectively, our results shift the current understanding of enterococcal cephalosporin resistance and suggest a model in which PbpA(2b) and Pbp5(4) cooperate to coordinately mediate peptidoglycan cross-linking in the presence of cephalosporins.


Assuntos
Resistência às Cefalosporinas/fisiologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecium/efeitos dos fármacos , Proteínas de Ligação às Penicilinas/fisiologia , Acilação , Infecção Hospitalar/tratamento farmacológico , Infecção Hospitalar/microbiologia , Eletroforese em Gel de Poliacrilamida , Enterococcus faecalis/metabolismo , Enterococcus faecium/metabolismo , Trato Gastrointestinal/microbiologia , Humanos , Immunoblotting , Concentração Inibidora 50 , Microscopia Eletrônica de Transmissão , Proteínas de Ligação às Penicilinas/química , Proteínas de Ligação às Penicilinas/metabolismo , Peptidoglicano/biossíntese
5.
Infect Immun ; 87(2)2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30510100

RESUMO

Neisseria gonorrhoeae releases peptidoglycan fragments during growth, and these molecules induce an inflammatory response in the human host. The proinflammatory molecules include peptidoglycan monomers, peptidoglycan dimers, and free peptides. These molecules can be released by the actions of lytic transglycosylases or an amidase. However, >40% of the gonococcal cell wall is cross-linked, where the peptide stem on one peptidoglycan strand is linked to the peptide stem on a neighboring strand, suggesting that endopeptidases may be required for the release of many peptidoglycan fragments. Therefore, we characterized mutants with individual or combined mutations in genes for the low-molecular-mass penicillin-binding proteins PBP3 and PBP4. Mutations in either dacB, encoding PBP3, or pbpG, encoding PBP4, did not significantly reduce the release of peptidoglycan monomers or free peptides. A mutation in dacB caused the appearance of a larger-sized peptidoglycan monomer, the pentapeptide monomer, and an increased release of peptidoglycan dimers, suggesting the involvement of this enzyme in both the removal of C-terminal d-Ala residues from stem peptides and the cleavage of cross-linked peptidoglycan. Mutation of both dacB and pbpG eliminated the release of tripeptide-containing peptidoglycan fragments concomitantly with the appearance of pentapeptide and dipeptide peptidoglycan fragments and higher-molecular-weight peptidoglycan dimers. In accord with the loss of tripeptide peptidoglycan fragments, the level of human NOD1 activation by the dacB pbpG mutants was significantly lower than that by the wild type. We conclude that PBP3 and PBP4 overlap in function for cross-link cleavage and that these endopeptidases act in the normal release of peptidoglycan fragments during growth.


Assuntos
Neisseria gonorrhoeae/patogenicidade , Proteína Adaptadora de Sinalização NOD1/fisiologia , Proteínas de Ligação às Penicilinas/fisiologia , Fragmentos de Peptídeos/metabolismo , Peptidoglicano/metabolismo , Parede Celular/metabolismo , Endopeptidases/metabolismo , Glicosiltransferases/metabolismo , Humanos , Neisseria gonorrhoeae/genética , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteínas de Ligação às Penicilinas/genética , Transdução de Sinais/fisiologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-29021974

RESUMO

Yersinia enterocolitica encodes a chromosomal AmpC ß-lactamase under the regulation of the classical ampR-ampC system. To obtain a further understanding to the role of low-molecular-mass penicillin-binding proteins (LMM PBPs) including PBP4, PBP5, PBP6, and PBP7, as well as NagZ and AmpR in ampC regulation of Y. enterocolitica, series of single/multiple mutant strains were systematically constructed and the ampC expression levels were determined by luxCDABE reporter system, reverse transcription-PCR (RT-PCR) and ß-lactamase activity test. Sequential deletion of PBP5 and other LMM PBPs result in a continuously growing of ampC expression level, the ß-lactamse activity of quadruple deletion strain YEΔ4Δ5Δ6Δ7 (pbp4, pbp5, pbp6, and pbp7 inactivated) is approached to the YEΔD123 (ampD1, ampD2, and ampD3 inactivated). Deletion of nagZ gene caused two completely different results in YEΔD123 and YEΔ4Δ5Δ6Δ7, NagZ is indispensable for YEΔ4Δ5Δ6Δ7 ampC derepression phenotype but dispensable for YEΔD123. AmpR is essential for ampC hyperproduction in these two types of strains, inactivation of AmpR notable reduced the ampC expression level in both YEΔD123 and YEΔ4Δ5Δ6Δ7.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas de Ligação às Penicilinas/fisiologia , Yersinia enterocolitica/metabolismo , beta-Lactamases/fisiologia , Acetilglucosaminidase/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Perfilação da Expressão Gênica , Regulação Bacteriana da Expressão Gênica/genética , Técnicas de Inativação de Genes , Teste de Complementação Genética , Testes de Sensibilidade Microbiana , Mutação , N-Acetil-Muramil-L-Alanina Amidase , Proteínas de Ligação às Penicilinas/genética , Regiões Promotoras Genéticas , Yersinia enterocolitica/enzimologia , Yersinia enterocolitica/genética , beta-Lactamases/genética , beta-Lactamases/metabolismo
7.
BMC Microbiol ; 16(1): 234, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27716106

RESUMO

BACKGROUND: Community and nosocomial infections by Pseudomonas aeruginosa still create a major therapeutic challenge. The resistance of this opportunist pathogen to ß-lactam antibiotics is determined mainly by production of the inactivating enzyme AmpC, a class C cephalosporinase with a regulation system more complex than those found in members of the Enterobacteriaceae family. This regulatory system also participates directly in peptidoglycan turnover and recycling. One of the regulatory mechanisms for AmpC expression, recently identified in clinical isolates, is the inactivation of LMM-PBP4 (Low-Molecular-Mass Penicillin-Binding Protein 4), a protein whose catalytic activity on natural substrates has remained uncharacterized until now. RESULTS: We carried out in vivo activity trials for LMM-PBP4 of Pseudomonas aeruginosa on macromolecular peptidoglycan of Escherichia coli and Pseudomonas aeruginosa. The results showed a decrease in the relative quantity of dimeric, trimeric and anhydrous units, and a smaller reduction in monomer disaccharide pentapeptide (M5) levels, validating the occurrence of D,D-carboxypeptidase and D,D-endopeptidase activities. Under conditions of induction for this protein and cefoxitin treatment, the reduction in M5 is not fully efficient, implying that LMM-PBP4 of Pseudomonas aeruginosa presents better behaviour as a D,D-endopeptidase. Kinetic evaluation of the direct D,D-peptidase activity of this protein on natural muropeptides M5 and D45 confirmed this bifunctionality and the greater affinity of LMM-PBP4 for its dimeric substrate. A three-dimensional model for the monomeric unit of LMM-PBP4 provided structural information which supports its catalytic performance. CONCLUSIONS: LMM-PBP4 of Pseudomonas aeruginosa is a bifunctional enzyme presenting both D,D-carboxypeptidase and D,D-endopeptidase activities; the D,D-endopeptidase function is predominant. Our study provides unprecedented functional and structural information which supports the proposal of this protein as a potential hydrolase-autolysin associated with peptidoglycan maturation and recycling. The fact that mutant PBP4 induces AmpC, may indicate that a putative muropeptide-subunit product of the DD-EPase activity of PBP4 could be a negative regulator of the pathway. This data contributes to understanding of the regulatory aspects of resistance to ß-lactam antibiotics in this bacterial model.


Assuntos
Proteínas de Ligação às Penicilinas/fisiologia , Pseudomonas aeruginosa/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Carboxipeptidases/metabolismo , Cefoxitina/farmacologia , Infecção Hospitalar , DNA Bacteriano/genética , Endopeptidases/metabolismo , Ativação Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Modelos Moleculares , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/isolamento & purificação , Proteínas de Ligação às Penicilinas/metabolismo , Peptidoglicano/metabolismo , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Proteínas Recombinantes , Resistência beta-Lactâmica/genética , beta-Lactamas/farmacologia
8.
PLoS One ; 9(6): e99605, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24932751

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) is a major multidrug resistant pathogen responsible for several difficult-to-treat infections in humans. Clinical Hetero-resistant (HeR) MRSA strains, mostly associated with persistent infections, are composed of mixed cell populations that contain organisms with low levels of resistance (hetero-resistant HeR) and those that display high levels of drug resistance (homo-resistant HoR). However, the full understanding of ß-lactam-mediated HeR/HoR selection remains to be completed. In previous studies we demonstrated that acquisition of the HoR phenotype during exposure to ß-lactam antibiotics depended on two key elements: (1) activation of the SOS response, a conserved regulatory network in bacteria that is induced in response to DNA damage, resulting in increased mutation rates, and (2) adaptive metabolic changes redirecting HeR-MRSA metabolism to the tricarboxylic acid (TCA) cycle in order to increase the energy supply for cell-wall synthesis. In the present work, we identified that both main mechanistic components are associated through TCA cycle-mediated reactive oxygen species (ROS) production, which temporally affects DNA integrity and triggers activation of the SOS response resulting in enhanced mutagenesis. The present work brings new insights into a role of ROS generation on the development of resistance to ß-lactam antibiotics in a model of natural occurrence, emphasizing the cytoprotective role in HeR-MRSA survival mechanism.


Assuntos
Antibacterianos/farmacologia , Ciclo do Ácido Cítrico/fisiologia , Farmacorresistência Bacteriana Múltipla/fisiologia , Staphylococcus aureus Resistente à Meticilina/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Resistência beta-Lactâmica/fisiologia , beta-Lactamas/farmacologia , 2,2'-Dipiridil/farmacologia , Adaptação Fisiológica , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Dano ao DNA , DNA Bacteriano/análise , Perfilação da Expressão Gênica , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/genética , Taxa de Mutação , Oxacilina/farmacologia , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Resposta SOS em Genética , Tioureia/farmacologia
9.
Mol Microbiol ; 90(5): 939-55, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24118410

RESUMO

Bacterial cell shapes are manifestations of programs carried out by multi-protein machines that synthesize and remodel the resilient peptidoglycan (PG) mesh and other polymers surrounding cells. GpsB protein is conserved in low-GC Gram-positive bacteria and is not essential in rod-shaped Bacillus subtilis, where it plays a role in shuttling penicillin-binding proteins (PBPs) between septal and side-wall sites of PG synthesis. In contrast, we report here that GpsB is essential in ellipsoid-shaped, ovococcal Streptococcus pneumoniae (pneumococcus), and depletion of GpsB leads to formation of elongated, enlarged cells containing unsegregated nucleoids and multiple, unconstricted rings of fluorescent-vancomycin staining, and eventual lysis. These phenotypes are similar to those caused by selective inhibition of Pbp2x by methicillin that prevents septal PG synthesis. Dual-protein 2D and 3D-SIM (structured illumination) immunofluorescence microscopy (IFM) showed that GpsB and FtsZ have overlapping, but not identical, patterns of localization during cell division and that multiple, unconstricted rings of division proteins FtsZ, Pbp2x, Pbp1a and MreC are in elongated cells depleted of GpsB. These patterns suggest that GpsB, like Pbp2x, mediates septal ring closure. This first dual-protein 3D-SIM IFM analysis also revealed separate positioning of Pbp2x and Pbp1a in constricting septa, consistent with two separable PG synthesis machines.


Assuntos
Proteínas de Bactérias/fisiologia , Peptidoglicano/metabolismo , Streptococcus pneumoniae/citologia , Streptococcus pneumoniae/metabolismo , Fatores de Virulência/fisiologia , Proteínas de Bactérias/metabolismo , Divisão Celular , Proteínas do Citoesqueleto/metabolismo , Deleção de Genes , Imageamento Tridimensional , Meticilina/farmacologia , Microscopia de Fluorescência , Proteínas de Ligação às Penicilinas/fisiologia , Peptidil Transferases/fisiologia , Fenótipo , Transporte Proteico , Streptococcus pneumoniae/genética , Fatores de Virulência/metabolismo
10.
Mol Microbiol ; 85(1): 164-78, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22624979

RESUMO

Chlamydiae are obligate intracellular bacterial pathogens that have extensively reduced their genome in adapting to the intracellular environment. The chlamydial genome contains only three annotated cell division genes and lacks ftsZ. How this obligate intracellular pathogen divides is uncharacterized. Chlamydiae contain two high-molecular-weight (HMW) penicillin binding proteins (Pbp) implicated in peptidoglycan synthesis, Pbp2 and Pbp3/FtsI. We show here, using HMW Pbp-specific penicillin derivatives, that both Pbp2 and Pbp3 are essential for chlamydial cell division. Ultrastructural analyses of antibiotic-treated cultures revealed distinct phenotypes: Pbp2 inhibition induced internal cell bodies within a single outer membrane whereas Pbp3 inhibition induced elongated phenotypes with little internal division. Each HMW Pbp interacts with the Chlamydia cell division protein FtsK. Chlamydiae are coccoid yet contain MreB, a rod shape-determining protein linked to Pbp2 in bacilli. Using MreB-specific antibiotics, we show that MreB is essential for chlamydial growth and division. Importantly, co-treatment with MreB-specific and Pbp-specific antibiotics resulted in the MreB-inhibited phenotype, placing MreB upstream of Pbp function in chlamydial cell division. Finally, we showed that MreB also interacts with FtsK. We propose that, in Chlamydia, MreB acts as a central co-ordinator at the division site to substitute for the lack of FtsZ in this bacterium.


Assuntos
Divisão Celular , Chlamydia trachomatis/citologia , Proteínas de Ligação às Penicilinas/fisiologia , Chlamydia trachomatis/genética , Proteínas do Citoesqueleto/metabolismo , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Ligação às Penicilinas/genética
11.
J Bacteriol ; 191(16): 5123-33, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19542290

RESUMO

Because very little is known about cell division in noncylindrical bacteria and cyanobacteria, we investigated 10 putative cytokinetic proteins in the unicellular spherical cyanobacterium Synechocystis strain PCC 6803. Concerning the eight penicillin-binding proteins (PBPs), which define three classes, we found that Synechocystis can survive in the absence of one but not two PBPs of either class A or class C, whereas the unique class B PBP (also termed FtsI) is indispensable. Furthermore, we showed that all three classes of PBPs are required for normal cell size. Similarly, the putative FtsQ and FtsW proteins appeared to be required for viability and normal cell size. We also used a suitable bacterial two-hybrid system to characterize the interaction web among the eight PBPs, FtsQ, and FtsW, as well as ZipN, the crucial FtsZ partner that occurs only in cyanobacteria and plant chloroplasts. We showed that FtsI, FtsQ, and ZipN are self-interacting proteins and that both FtsI and FtsQ interact with class A PBPs, as well as with ZipN. Collectively, these findings indicate that ZipN, in interacting with FtsZ and both FtsI and FtQ, plays a similar role to the Escherichia coli FtsA protein, which is missing in cyanobacteria and chloroplasts.


Assuntos
Proteínas de Bactérias/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Ligação às Penicilinas/fisiologia , Synechocystis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Citometria de Fluxo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Biológicos , Mutação , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/metabolismo , Ligação Proteica/genética , Ligação Proteica/fisiologia , Synechocystis/genética , Synechocystis/crescimento & desenvolvimento , Técnicas do Sistema de Duplo-Híbrido
13.
J Bacteriol ; 191(11): 3526-33, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19346310

RESUMO

In Escherichia coli, the cytoplasmic proteins MreB and FtsZ play crucial roles in ensuring that new muropeptide subunits are inserted into the cell wall in a spatially correct way during elongation and division. In particular, to retain a constant diameter and overall shape, new material must be inserted into the wall uniformly around the cell's perimeter. Current thinking is that MreB accomplishes this feat through intermediary proteins that tether peptidoglycan synthases to the outer face of the inner membrane. We tested this idea in E. coli by using a DD-carboxypeptidase mutant that accumulates pentapeptides in its peptidoglycan, allowing us to visualize new muropeptide incorporation. Surprisingly, inhibiting MreB with the antibiotic A22 did not result in uneven insertion of new wall, although the cells bulged and lost their rod shapes. Instead, uneven (clustered) incorporation occurred only if MreB and FtsZ were inactivated simultaneously, providing the first evidence in E. coli that FtsZ can direct murein incorporation into the lateral cell wall independently of MreB. Inhibiting penicillin binding protein 2 (PBP 2) alone produced the same clustered phenotype, implying that MreB and FtsZ tether peptidoglycan synthases via a common mechanism that includes PBP 2. However, cell shape was determined only by the presence or absence of MreB and not by the even distribution of new wall material as directed by FtsZ.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas de Ligação às Penicilinas/fisiologia , Arabinose/farmacologia , Aztreonam/farmacologia , Parede Celular/efeitos dos fármacos , Escherichia coli/citologia , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Microscopia Confocal , Modelos Biológicos , Proteínas de Ligação às Penicilinas/antagonistas & inibidores , Peptidoglicano/metabolismo , Tioureia/análogos & derivados , Tioureia/farmacologia
14.
J Infect Dis ; 199(4): 513-21, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19143563

RESUMO

BACKGROUND: Acinetobacter baumannii is a bacterial pathogen of increasing medical importance. Little is known about genes important for its survival in vivo. METHODS AND RESULTS: Screening of random transposon mutants of the model pathogen AB307-0294 identified the mutant AB307.27. AB307.27 contained its transposon insertion in pbpG, which encodes the putative low-molecular-mass penicillin-binding protein 7/8 (PBP-7/8). AB307.27 was significantly killed in ascites (P<.001), but its growth in Luria-Bertani broth was similar to that of its parent, AB307-0294 (P=.13). The survival of AB307.27 was significantly decreased in a rat soft-tissue infection model (P<.001) and a rat pneumonia model (P=.002), compared with AB307-0294. AB307.27 was significantly killed in 90% human serum in vitro, compared with AB307-0294 (P<.001). Electron microscopy demonstrated more coccobacillary forms of AB307.27, compared with AB307-0294, suggesting a possible modulation in the peptidoglycan, which may affect susceptibility to host defense factors. CONCLUSIONS: These findings demonstrate that PBP-7/8 contributes to the pathogenesis of A. baumannii. PBP-7/8 either directly or indirectly contributes to the resistance of AB307-0294 to complement-mediated bactericidal activity. An understanding of how PBP-7/8 contributes to serum resistance will lend insight into the role of this low-molecular-mass PBP whose function is poorly understood.


Assuntos
Infecções por Acinetobacter/imunologia , Infecções por Acinetobacter/microbiologia , Acinetobacter baumannii/crescimento & desenvolvimento , Proteínas de Ligação às Penicilinas/fisiologia , Acinetobacter baumannii/genética , Acinetobacter baumannii/patogenicidade , Sequência de Aminoácidos , Animais , Atividade Bactericida do Sangue , Proteínas do Sistema Complemento/imunologia , Simulação por Computador , Elementos de DNA Transponíveis , Interpretação Estatística de Dados , Modelos Animais de Doenças , Humanos , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Mutação , Proteínas de Ligação às Penicilinas/química , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/metabolismo , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Ratos , Infecções dos Tecidos Moles/imunologia , Infecções dos Tecidos Moles/microbiologia , Urina/microbiologia
15.
Res Microbiol ; 160(2): 117-24, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19063962

RESUMO

The study was focused on the role of the penicillin binding protein PBP4* of Bacillus subtilis during growth in high salinity rich media. Using pbpE-lacZ fusion, we found that transcription of the pbpE gene is induced in stationary phase and by increased salinity. This increase was also corroborated at the translation level for PBP4* by western blot. Furthermore, we showed that a strain harboring gene disruption in the structural gene (pbpE) for the PBP4* endopeptidase resulted in a salt-sensitive phenotype and increased sensitivity to cell envelope active antibiotics (vancomycin, penicillin and bacitracin). Since the pbpE gene seems to be part of a two-gene operon with racX, a racX::pRV300 mutant was obtained. This mutant behaved like the wild-type strain with respect to high salt. Electron microscopy showed that high salt and mutation of pbpE resulted in cell wall defects. Whole cells or purified peptidoglycan from WT cultures grown in high salt medium showed increased autolysis and susceptibility to mutanolysin. We demonstrate through zymogram analysis that PBP4* has murein hydrolyze activity. All these results support the hypothesis that peptidoglycan is modified in response to high salt and that PBP4* contributes to this modification.


Assuntos
Bacillus subtilis/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/fisiologia , Proteínas de Ligação às Penicilinas/fisiologia , Salinidade , D-Ala-D-Ala Carboxipeptidase Tipo Serina/fisiologia , Antibacterianos/farmacologia , Bacillus subtilis/crescimento & desenvolvimento , Bacillus subtilis/ultraestrutura , Bacitracina/farmacologia , Bacteriólise , Parede Celular/efeitos dos fármacos , Parede Celular/ultraestrutura , Microscopia Eletrônica de Transmissão , N-Acetil-Muramil-L-Alanina Amidase/deficiência , Penicilina G/farmacologia , Proteínas de Ligação às Penicilinas/deficiência , Peptidoglicano/metabolismo , D-Ala-D-Ala Carboxipeptidase Tipo Serina/deficiência , Transcrição Gênica , Vancomicina/farmacologia
16.
Am J Obstet Gynecol ; 199(5): 548.e1-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18486087

RESUMO

OBJECTIVE: We sought to determine the role lipid rafts and phosphoinositide 3-kinase (PI3K) in invasiveness of group B streptococci (GBS) to endometrial cells. STUDY DESIGN: Antibiotic protection assay and electron microscopy were used to evaluate the invasion of GBS to human endometrial Ishikawa cells cholesterol-depleted by using methyl-beta-cyclodextrin or treated with PI3K inhibitors: wortmannin or LY294002. Immunoblotting analysis of Akt phosphorylation and cellular imaging of GFP-Akt-PH probe were used to assess PI3Ks activation in infected cells. RESULTS: Infected Ishikawa cells streptococci are associated to membrane ruffles with morphological features of undergoing internalization. GBS remained attached but completely failed to invade to cholesterol-depleted human endometrial cells or displayed decreased invasiveness in the presence of PI3K inhibitors. Cholesterol depletion resulted in loss of membrane ruffling and dispersion of raft-associated molecules: monosialoganglioside GM1 and PI3K. CONCLUSION: This work provides the evidence that lipid rafts and raft-associated PI3K are implicated in GBS invasion to human endometrial cells.


Assuntos
1-Fosfatidilinositol 4-Quinase/fisiologia , Endométrio/microbiologia , Microdomínios da Membrana/microbiologia , Streptococcus agalactiae/crescimento & desenvolvimento , 1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , Androstadienos/farmacologia , Aderência Bacteriana , Cápsulas Bacterianas/fisiologia , Células Cultivadas , Colesterol/fisiologia , Cromonas/farmacologia , Feminino , Humanos , Immunoblotting , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Morfolinas/farmacologia , Proteínas de Ligação às Penicilinas/fisiologia , Infecções Estreptocócicas/metabolismo , Streptococcus agalactiae/patogenicidade , Streptococcus agalactiae/ultraestrutura , Transfecção , Wortmanina , beta-Ciclodextrinas/farmacologia
17.
J Immunol ; 179(5): 3196-202, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17709535

RESUMO

Group B streptococcus (GBS) is a major cause of neonatal pneumonia. The early interactions between innate airway defenses and this pathogen are likely to be a critical factor in determining the outcome for the host. The surface-localized penicillin-binding protein (PBP)1a, encoded by ponA, is known to be an important virulence trait in a sepsis model of GBS infection that promotes resistance to neutrophil killing and more specifically to neutrophil antimicrobial peptides (AMPs). In this study, we used an aerosolization model to explore the role of PBP1a in evasion of innate immune defenses in the neonatal lung. The ponA mutant strain was cleared more rapidly from the lungs of neonatal rat pups compared with the wild-type strain, which could be linked to a survival defect in the presence of alveolar macrophages (AM). Rat AM were found to secrete beta-defensin and cathelicidin AMP homologues, and the GBS ponA mutant was more susceptible than the wild-type strain to killing by these peptides in vitro. Collectively, our observations suggest that PBP1a-mediated resistance to AM AMPs promotes the survival of GBS in the neonatal lung. Additionally, AM are traditionally thought to clear bacteria through phagocytic uptake; our data indicate that secretion of AMPs may also participate in limiting bacterial replication in the airway.


Assuntos
Proteínas de Bactérias/fisiologia , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Proteínas de Ligação às Penicilinas/fisiologia , Pneumonia Bacteriana/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus agalactiae/patogenicidade , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas de Bactérias/genética , Líquido da Lavagem Broncoalveolar/imunologia , Líquido da Lavagem Broncoalveolar/microbiologia , Feminino , Imunidade Inata , Pulmão/crescimento & desenvolvimento , Pulmão/microbiologia , Macrófagos Alveolares/microbiologia , Proteínas de Ligação às Penicilinas/genética , Pneumonia Bacteriana/microbiologia , Ratos , Ratos Sprague-Dawley , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/enzimologia , Streptococcus agalactiae/genética , beta-Defensinas/metabolismo , Catelicidinas
18.
Biochemistry ; 46(27): 8050-7, 2007 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-17567045

RESUMO

The genome of Staphylococcus aureus is constantly in a state of flux, acquiring genes that enable the bacterium to maintain resistance in the face of antibiotic pressure. The acquisition of the mecA gene from an unknown origin imparted S. aureus with broad resistance to beta-lactam antibiotics, with the resultant strain designated as methicillin-resistant S. aureus (MRSA). Epidemiological and genetic evidence suggests that the gene encoding PBP 2a of MRSA might have originated from Staphylococcus sciuri, an animal pathogen, where it exists as a silent gene of unknown function. We synthesized, cloned, and expressed the mecA gene of S. sciuri in Escherichia coli, and the protein product was purified to homogeneity. Biochemical characterization and comparison of the protein to PBP 2a of S. aureus revealed them to be highly similar. These characteristics start with sequence similarity but extend to biochemical behavior in inhibition by beta-lactam antibiotics, to the existence of an allosteric site for binding of bacterial peptidoglycan, to the issues of the sheltered active site, and to the need for conformational change in making the active site accessible to the substrate and the inhibitors. Altogether, the evidence strongly argues that the kinship between the two proteins is deep-rooted on the basis of many biochemical attributes quantified in this study.


Assuntos
Proteínas de Bactérias/fisiologia , Resistência a Meticilina , Proteínas de Ligação às Penicilinas/fisiologia , Staphylococcus aureus/fisiologia , Staphylococcus/fisiologia , Proteínas de Bactérias/genética , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Modelos Moleculares , Proteínas de Ligação às Penicilinas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrofotometria Ultravioleta , Staphylococcus/genética , Staphylococcus aureus/genética
19.
J Bacteriol ; 189(14): 4975-83, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17513478

RESUMO

Penicillin-binding protein 2 (PBP 2) has long been known to be essential for rod-shaped morphology in gram-negative bacteria, including Escherichia coli and Pseudomonas aeruginosa. In the course of earlier studies with P. aeruginosa PBP 2, we observed that E. coli was sensitive to the overexpression of its gene, pbpA. In this study, we examined E. coli overproducing both P. aeruginosa and E. coli PBP 2. Growth of cells entered a stationary phase soon after induction of gene expression, and cells began to lyse upon prolonged incubation. Concomitant with the growth retardation, cells were observed to have changed morphologically from typical rods into enlarged spheres. Inactive derivatives of the PBP 2s were engineered, involving site-specific replacement of their catalytic Ser residues with Ala in their transpeptidase module. Overproduction of these inactive PBPs resulted in identical effects. Likewise, overproduction of PBP 2 derivatives possessing only their N-terminal non-penicillin-binding module (i.e., lacking their C-terminal transpeptidase module) produced similar effects. However, E. coli overproducing engineered derivatives of PBP 2 lacking their noncleavable, N-terminal signal sequence and membrane anchor were found to grow and divide at the same rate as control cells. The morphological effects and lysis were also eliminated entirely when overproduction of PBP 2 and variants was conducted with E. coli MHD79, a strain lacking six lytic transglycosylases. A possible interaction between the N-terminal domain of PBP 2 and lytic transglycosylases in vivo through the formation of multienzyme complexes is discussed.


Assuntos
Bacteriólise/genética , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Proteínas de Ligação às Penicilinas/genética , Bacteriólise/fisiologia , Western Blotting , Eletroforese em Gel de Poliacrilamida , Escherichia coli/fisiologia , Escherichia coli/ultraestrutura , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Microscopia Eletrônica de Varredura , Modelos Genéticos , Mutação , Proteínas de Ligação às Penicilinas/metabolismo , Proteínas de Ligação às Penicilinas/fisiologia , Peptidoglicano Glicosiltransferase/genética , Peptidoglicano Glicosiltransferase/metabolismo , Pseudomonas aeruginosa/genética
20.
J Bacteriol ; 189(9): 3525-31, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17307860

RESUMO

We constructed a conditional mutant of pbpA in which transcription of the gene was placed under the control of an IPTG (isopropyl-beta-D-thiogalactopyranoside)-inducible promoter in order to explore the role of PBP1 in growth, cell wall structure, and cell division. A methicillin-resistant strain and an isogenic methicillin-susceptible strain, each carrying the pbpA mutation, were unable to grow in the absence of the inducer. Conditional mutants of pbpA transferred into IPTG-free medium underwent a four- to fivefold increase in cell mass, which was not accompanied by a proportional increase in viable titer. Examination of thin sections of such cells by transmission electron microscopy or fluorescence microscopy of intact cells with Nile red-stained membranes showed a morphologically heterogeneous population of bacteria with abnormally increased sizes, distorted axial ratios, and a deficit in the number of cells with completed septa. Immunofluorescence with an antibody specific for PBP1 localized the protein to sites of cell division. No alteration in the composition of peptidoglycan was detectable in pbpA conditional mutants grown in the presence of a suboptimal concentration of IPTG, which severely restricted the rate of growth, and the essential function of PBP1 could not be replaced by PBP2A present in methicillin-resistant cells. These observations suggest that PBP1 is not a major contributor to the cross-linking of peptidoglycan and that its essential function must be intimately integrated into the mechanism of cell division.


Assuntos
Proteínas de Bactérias/fisiologia , Divisão Celular , Proteínas de Ligação às Penicilinas/fisiologia , Staphylococcus aureus/fisiologia , Antibacterianos/farmacologia , Biomassa , Parede Celular/ultraestrutura , Contagem de Colônia Microbiana , Expressão Gênica , Genes Bacterianos , Genes Essenciais , Teste de Complementação Genética , Meticilina/farmacologia , Resistência a Meticilina , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Mutação , Proteínas de Ligação às Penicilinas/genética , Peptidoglicano/análise , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...