Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Cell ; 56(9): 1296-1312.e7, 2021 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-33878346

RESUMO

Mammalian inner ear and fish lateral line sensory hair cells (HCs) detect fluid motion to transduce environmental signals. Actively maintained ionic homeostasis of the mammalian inner ear endolymph is essential for HC function. In contrast, fish lateral line HCs are exposed to the fluctuating ionic composition of the aqueous environment. Using lineage labeling, in vivo time-lapse imaging and scRNA-seq, we discovered highly motile skin-derived cells that invade mature mechanosensory organs of the zebrafish lateral line and differentiate into Neuromast-associated (Nm) ionocytes. This invasion is adaptive as it is triggered by environmental fluctuations. Our discovery of Nm ionocytes challenges the notion of an entirely placodally derived lateral line and identifies Nm ionocytes as likely regulators of HC function possibly by modulating the ionic microenvironment. Nm ionocytes provide an experimentally accessible in vivo system to study cell invasion and migration, as well as the physiological adaptation of vertebrate organs to changing environmental conditions.


Assuntos
Adaptação Fisiológica , Movimento Celular , Meio Ambiente , Homeostase , Sistema da Linha Lateral/citologia , Peixe-Zebra/fisiologia , Animais , Biomarcadores/metabolismo , Contagem de Células , Fatores de Transcrição Forkhead/metabolismo , Brânquias/citologia , Células Ciliadas Auditivas/citologia , Concentração de Íons de Hidrogênio , Imageamento Tridimensional , Receptores Notch/metabolismo , Salinidade , Transdução de Sinais , Pele/citologia , Proteínas de Peixe-Zebra/metabolismo
2.
J Biol Chem ; 291(5): 2535-46, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26620564

RESUMO

Screening of new compounds directed against key protein targets must continually keep pace with emerging antibiotic resistances. Although periplasmic enzymes of bacterial cell wall biosynthesis have been among the first drug targets, compounds directed against the membrane-integrated catalysts are hardly available. A promising future target is the integral membrane protein MraY catalyzing the first membrane associated step within the cytoplasmic pathway of bacterial peptidoglycan biosynthesis. However, the expression of most MraY homologues in cellular expression systems is challenging and limits biochemical analysis. We report the efficient production of MraY homologues from various human pathogens by synthetic cell-free expression approaches and their subsequent characterization. MraY homologues originating from Bordetella pertussis, Helicobacter pylori, Chlamydia pneumoniae, Borrelia burgdorferi, and Escherichia coli as well as Bacillus subtilis were co-translationally solubilized using either detergent micelles or preformed nanodiscs assembled with defined membranes. All MraY enzymes originating from Gram-negative bacteria were sensitive to detergents and required nanodiscs containing negatively charged lipids for obtaining a stable and functionally folded conformation. In contrast, the Gram-positive B. subtilis MraY not only tolerates detergent but is also less specific for its lipid environment. The MraY·nanodisc complexes were able to reconstitute a complete in vitro lipid I and lipid II forming pipeline in combination with the cell-free expressed soluble enzymes MurA-F and with the membrane-associated protein MurG. As a proof of principle for future screening platforms, we demonstrate the inhibition of the in vitro lipid II biosynthesis with the specific inhibitors fosfomycin, feglymycin, and tunicamycin.


Assuntos
Proteínas de Bactérias/química , Monossacarídeos/biossíntese , Oligopeptídeos/biossíntese , Transferases/química , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Bacillus subtilis/enzimologia , Vias Biossintéticas , Bordetella pertussis/enzimologia , Borrelia burgdorferi/enzimologia , Parede Celular/química , Sistema Livre de Células , Chlamydophila pneumoniae/enzimologia , Citoplasma/química , DNA/química , Detergentes/química , Escherichia coli/enzimologia , Fosfomicina/química , Helicobacter pylori/enzimologia , Micelas , Peptídeos/química , Peptidoglicano/química , Proteínas/química , Proteínas Recombinantes/química , Transferases (Outros Grupos de Fosfato Substituídos) , Tunicamicina/química , Uridina Difosfato Ácido N-Acetilmurâmico/biossíntese
3.
Biochim Biophys Acta ; 1848(11 Pt B): 3062-71, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25934055

RESUMO

Antimicrobial peptides (AMPs) are natural antibiotics produced by virtually all living organisms. Typically, AMPs are cationic and amphiphilic and first contacts with target microbes involve interactions with negatively charged components of the cell envelope such as lipopolysaccharide (LPS), and wall- or lipoteichoic acids (WTA, LTA). The importance of charge-mediated interactions of AMPs with the cell envelope is reflected by effective microbial resistance mechanisms which are based on reduction of the overall charge of these polymers. The anionic polymers are linked in various ways to the stress-bearing polymer of the cell envelope, the peptidoglycan, which is made of a highly conserved building block, a disaccharide-pentapeptide moiety that also contains charged residues. This structural element, in spite of its conservation throughout the bacterial world, can undergo genus- and species-specific modifications that also impact significantly on the overall charge of the cell envelope and on the binding affinity of AMPs. The modification reactions involved largely occur on the membrane-bound peptidoglycan building block, the so-called lipid II, which is a most prominent target for AMPs. In this review, we focus on modifications of lipid II and peptidoglycan and discuss their consequences for the interactions with various classes of AMPs, such as defensins, lantibiotics and glyco-(lipo)-peptide antibiotics. This article is part of a Special Issue entitled: Bacterial Resistance to Antimicrobial Peptides.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Parede Celular/metabolismo , Farmacorresistência Bacteriana , Bactérias Gram-Positivas/metabolismo , Infecções por Bactérias Gram-Positivas/metabolismo , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Animais , Antibacterianos/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Bactérias Gram-Positivas/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/prevenção & controle , Interações Hospedeiro-Patógeno , Humanos , Viabilidade Microbiana , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Uridina Difosfato Ácido N-Acetilmurâmico/química , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo
4.
ACS Chem Biol ; 10(4): 1034-42, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25574687

RESUMO

Lantibiotics, an abbreviation for "lanthionine-containing antibiotics", interfere with bacterial metabolism by a mechanism not exploited by the antibiotics currently in clinical use. Thus, they have aroused interest as a source for new therapeutic agents because they can overcome existing resistance mechanisms. Starting from fermentation broth extracts preselected from a high-throughput screening program for discovering cell-wall inhibitors, we isolated a series of related class I lantibiotics produced by different genera of actinomycetes. Analytical techniques together with explorative chemistry have been used to establish their structures: the newly described compounds share a common 24 aa sequence with the previously reported lantibiotic planosporicin (aka 97518), differing at positions 4, 6, and 14. All of these compounds maintain an overall -1 charge at physiological pH. While all of these lantibiotics display modest antibacterial activity, their potency can be substantially modulated by progressively eliminating the negative charges, with the most active compounds carrying basic amide derivatives of the two carboxylates originally present in the natural compounds. Interestingly, both natural and chemically modified lantibiotics target the key biosynthetic intermediate lipid II, but the former compounds do not bind as effectively as the latter in vivo. Remarkably, the basic derivatives display an antibacterial potency and a killing effect similar to those of NAI-107, a distantly related actinomycete-produced class I lantibiotic which lacks altogether carboxyl groups and which is a promising clinical candidate for treating Gram-positive infections caused by multi-drug-resistant pathogens.


Assuntos
Actinobacteria/metabolismo , Antibacterianos/química , Antibacterianos/farmacologia , Bacteriocinas/farmacologia , Actinobacteria/química , Actinobacteria/classificação , Actinobacteria/crescimento & desenvolvimento , Bacteriocinas/química , Bacteriocinas/isolamento & purificação , Avaliação Pré-Clínica de Medicamentos/métodos , Concentração de Íons de Hidrogênio , Espectroscopia de Ressonância Magnética , Testes de Sensibilidade Microbiana , Estrutura Molecular , Streptomyces/classificação , Streptomyces/metabolismo , Relação Estrutura-Atividade
5.
Antimicrob Agents Chemother ; 59(2): 772-81, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25403671

RESUMO

Oritavancin is a semisynthetic derivative of the glycopeptide antibiotic chloroeremomycin with activity against Gram-positive pathogens, including vancomycin-resistant staphylococci and enterococci. Compared to vancomycin, oritavancin is characterized by the presence of two additional residues, a hydrophobic 4'-chlorobiphenyl methyl moiety and a 4-epi-vancosamine substituent, which is also present in chloroeremomycin. Here, we show that oritavancin and its des-N-methylleucyl variant (des-oritavancin) effectively inhibit lipid I- and lipid II-consuming peptidoglycan biosynthesis reactions in vitro. In contrast to that for vancomycin, the binding affinity of oritavancin to the cell wall precursor lipid II appears to involve, in addition to the D-Ala-D-Ala terminus, other species-specific binding sites of the lipid II molecule, i.e., the crossbridge and D-isoglutamine in position 2 of the lipid II stem peptide, both characteristic for a number of Gram-positive pathogens, including staphylococci and enterococci. Using purified lipid II and modified lipid II variants, we studied the impact of these modifications on the binding of oritavancin and compared it to those of vancomycin, chloroeremomycin, and des-oritavancin. Analysis of the binding parameters revealed that additional intramolecular interactions of oritavancin with the peptidoglycan precursor appear to compensate for the loss of a crucial hydrogen bond in vancomycin-resistant strains, resulting in enhanced binding affinity. Augmenting previous findings, we show that amidation of the lipid II stem peptide predominantly accounts for the increased binding of oritavancin to the modified intermediates ending in D-Ala-D-Lac. Corroborating our conclusions, we further provide biochemical evidence for the phenomenon of the antagonistic effects of mecA and vanA resistance determinants in Staphylococcus aureus, thus partially explaining the low frequency of methicillin-resistant S. aureus (MRSA) acquiring high-level vancomycin resistance.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Glicopeptídeos/química , Glicopeptídeos/farmacologia , Enterococcus faecium/química , Lipoglicopeptídeos , Testes de Sensibilidade Microbiana , Staphylococcus aureus/efeitos dos fármacos
6.
J Biol Chem ; 289(50): 34953-64, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25342741

RESUMO

Fungi and bacteria compete with an arsenal of secreted molecules for their ecological niche. This repertoire represents a rich and inexhaustible source for antibiotics and fungicides. Antimicrobial peptides are an emerging class of fungal defense molecules that are promising candidates for pharmaceutical applications. Based on a co-cultivation system, we studied the interaction of the coprophilous basidiomycete Coprinopsis cinerea with different bacterial species and identified a novel defensin, copsin. The polypeptide was recombinantly produced in Pichia pastoris, and the three-dimensional structure was solved by NMR. The cysteine stabilized α/ß-fold with a unique disulfide connectivity, and an N-terminal pyroglutamate rendered copsin extremely stable against high temperatures and protease digestion. Copsin was bactericidal against a diversity of Gram-positive bacteria, including human pathogens such as Enterococcus faecium and Listeria monocytogenes. Characterization of the antibacterial activity revealed that copsin bound specifically to the peptidoglycan precursor lipid II and therefore interfered with the cell wall biosynthesis. In particular, and unlike lantibiotics and other defensins, the third position of the lipid II pentapeptide is essential for effective copsin binding. The unique structural properties of copsin make it a possible scaffold for new antibiotics.


Assuntos
Agaricales/metabolismo , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Bactérias/metabolismo , Defensinas/farmacologia , Proteínas Fúngicas/farmacologia , Peptidoglicano/biossíntese , Agaricales/crescimento & desenvolvimento , Sequência de Aminoácidos , Antibacterianos/química , Antibacterianos/metabolismo , Bactérias/crescimento & desenvolvimento , Técnicas de Cocultura , Defensinas/química , Defensinas/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica
7.
Nat Commun ; 5: 4201, 2014 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-24953137

RESUMO

Intracellular Chlamydiaceae do not need to resist osmotic challenges and a functional cell wall was not detected in these pathogens. Nevertheless, a recent study revealed evidence for circular peptidoglycan-like structures in Chlamydiaceae and penicillin inhibits cytokinesis, a phenomenon known as the chlamydial anomaly. Here, by characterizing a cell wall precursor-processing enzyme, we provide insights into the mechanisms underlying this mystery. We show that AmiA from Chlamydia pneumoniae separates daughter cells in an Escherichia coli amidase mutant. Contrary to homologues from free-living bacteria, chlamydial AmiA uses lipid II as a substrate and has dual activity, acting as an amidase and a carboxypeptidase. The latter function is penicillin sensitive and assigned to a penicillin-binding protein motif. Consistent with the lack of a regulatory domain in AmiA, chlamydial CPn0902, annotated as NlpD, is a carboxypeptidase, rather than an amidase activator, which is the case for E. coli NlpD. Functional conservation of AmiA implicates a role in cytokinesis and host response modulation.


Assuntos
Amidoidrolases/metabolismo , Proteínas de Bactérias/metabolismo , Chlamydophila pneumoniae/enzimologia , Penicilinas/farmacologia , Amidoidrolases/antagonistas & inibidores , Amidoidrolases/química , Amidoidrolases/genética , Sequência de Aminoácidos , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Carboxipeptidases/química , Carboxipeptidases/genética , Carboxipeptidases/metabolismo , Parede Celular/enzimologia , Parede Celular/genética , Parede Celular/metabolismo , Chlamydophila pneumoniae/citologia , Chlamydophila pneumoniae/efeitos dos fármacos , Chlamydophila pneumoniae/genética , Citocinese , Dados de Sequência Molecular , Alinhamento de Sequência
8.
J Biol Chem ; 289(17): 12063-12076, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24627484

RESUMO

The lantibiotic NAI-107 is active against Gram-positive bacteria including vancomycin-resistant enterococci and methicillin-resistant Staphylococcus aureus. To identify the molecular basis of its potency, we studied the mode of action in a series of whole cell and in vitro assays and analyzed structural features by nuclear magnetic resonance (NMR). The lantibiotic efficiently interfered with late stages of cell wall biosynthesis and induced accumulation of the soluble peptidoglycan precursor UDP-N-acetylmuramic acid-pentapeptide (UDP-MurNAc-pentapeptide) in the cytoplasm. Using membrane preparations and a complete cascade of purified, recombinant late stage peptidoglycan biosynthetic enzymes (MraY, MurG, FemX, PBP2) and their respective purified substrates, we showed that NAI-107 forms complexes with bactoprenol-pyrophosphate-coupled precursors of the bacterial cell wall. Titration experiments indicate that first a 1:1 stoichiometric complex occurs, which then transforms into a 2:1 (peptide: lipid II) complex, when excess peptide is added. Furthermore, lipid II and related molecules obviously could not serve as anchor molecules for the formation of defined and stable nisin-like pores, however, slow membrane depolarization was observed after NAI-107 treatment, which could contribute to killing of the bacterial cell.


Assuntos
Bacteriocinas/metabolismo , Parede Celular/metabolismo , Terpenos/metabolismo , Sequência de Aminoácidos , Bacteriocinas/química , Bacteriocinas/farmacologia , Parede Celular/efeitos dos fármacos , Parede Celular/fisiologia , Eletroforese em Gel de Poliacrilamida , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Proteômica , Staphylococcus aureus/efeitos dos fármacos
9.
PLoS One ; 7(11): e48981, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166602

RESUMO

The nonantibiotic small molecule cyslabdan, a labdan-type diterpene produced by Streptomyces sp. K04-0144, markedly potentiated the activity of the ß-lactam drug imipenem against methicillin-resistant Staphylococcus aureus (MRSA). To study the mechanism of action of cyslabdan, the proteins that bind to cyslabdan were investigated in an MRSA lysate, which led to the identification of FemA, which is involved in the synthesis of the pentaglycine interpeptide bridge of the peptidoglycan of MRSA. Furthermore, binding assay of cyslabdan to FemB and FemX with the function similar to FemA revealed that cyslabdan had an affinity for FemB but not FemX. In an enzyme-based assay, cyslabdan inhibited FemA activity, where as did not affected FemX and FemB activities. Nonglycyl and monoglycyl murein monomers were accumulated by cyslabdan in the peptidoglycan of MRSA cell walls. These findings indicated that cyslabdan primarily inhibits FemA, thereby suppressing pentaglycine interpeptide bridge synthesis. This protein is a key factor in the determination of ß-lactam resistance in MRSA, and our findings provide a new strategy for combating MRSA.


Assuntos
Acetilcisteína/análogos & derivados , Proteínas de Bactérias/antagonistas & inibidores , Diterpenos/farmacologia , Farmacorresistência Bacteriana/genética , Imipenem/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Acetilcisteína/farmacologia , Compostos de Boro , Cromatografia Líquida de Alta Pressão , Primers do DNA/genética , Sinergismo Farmacológico , Japão , Staphylococcus aureus Resistente à Meticilina/metabolismo , Penicilinas , Peptídeos/metabolismo , Peptidoglicano/metabolismo , Espectrofotometria Ultravioleta , Espectrometria de Massas em Tandem
10.
Biochim Biophys Acta ; 1818(12): 3098-106, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22960287

RESUMO

Nanodiscs (NDs) enable the analysis of membrane proteins (MP) in natural lipid bilayer environments. In combination with cell-free (CF) expression, they could be used for the co-translational insertion of MPs into defined membranes. This new approach allows the characterization of MPs without detergent contact and it could help to identify effects of particular lipids on catalytic activities. Association of MPs with different ND types, quality of the resulting MP/ND complexes as well as optimization parameters are still poorly analyzed. This study describes procedures to systematically improve CF expression protocols for the production of high quality MP/ND complexes. In order to reveal target dependent variations, the co-translational ND complex formation with the bacterial proton pump proteorhodopsin (PR), with the small multidrug resistance transporters SugE and EmrE, as well as with the Escherichia coli MraY translocase was studied. Parameters which modulate the efficiency of MP/ND complex formation have been identified and in particular effects of different lipid compositions of the ND membranes have been analyzed. Recorded force distance pattern as well as characteristic photocycle dynamics indicated the integration of functionally folded PR into NDs. Efficient complex formation of the E. coli MraY translocase was dependent on the ND size and on the lipid composition of the ND membranes. Active MraY protein could only be obtained with ND containing anionic lipids, thus providing new details for the in vitro analysis of this pharmaceutically important protein.


Assuntos
Proteínas de Bactérias/química , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Rodopsina/química , Transferases/química , Antiporters/química , Antiporters/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Escherichia coli/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Rodopsina/metabolismo , Rodopsinas Microbianas , Transferases/metabolismo , Transferases (Outros Grupos de Fosfato Substituídos)
11.
Antimicrob Agents Chemother ; 56(11): 5749-57, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22926563

RESUMO

Mersacidin, gallidermin, and nisin are lantibiotics, antimicrobial peptides containing lanthionine. They show potent antibacterial activity. All three interfere with cell wall biosynthesis by binding lipid II, but they display different levels of interaction with the cytoplasmic membrane. On one end of the spectrum, mersacidin interferes with cell wall biosynthesis by binding lipid II without integrating into bacterial membranes. On the other end of the spectrum, nisin readily integrates into membranes, where it forms large pores. It destroys the membrane potential and causes leakage of nutrients and ions. Gallidermin, in an intermediate position, also readily integrates into membranes. However, pore formation occurs only in some bacteria and depends on membrane composition. In this study, we investigated the impact of nisin, gallidermin, and mersacidin on cell wall integrity, membrane pore formation, and membrane depolarization in Bacillus subtilis. The impact of the lantibiotics on the cell envelope was correlated to the proteomic response they elicit in B. subtilis. By drawing on a proteomic response library, including other envelope-targeting antibiotics such as bacitracin, vancomycin, gramicidin S, or valinomycin, YtrE could be identified as the most reliable marker protein for interfering with membrane-bound steps of cell wall biosynthesis. NadE and PspA were identified as markers for antibiotics interacting with the cytoplasmic membrane.


Assuntos
Bacillus subtilis/efeitos dos fármacos , Proteínas de Bactérias/biossíntese , Bacteriocinas/farmacologia , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Nisina/farmacologia , Peptídeos/farmacologia , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/genética , Transporte Biológico/efeitos dos fármacos , Biomarcadores/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Parede Celular/genética , Parede Celular/metabolismo , Eletroforese em Gel Bidimensional , Potenciais da Membrana/efeitos dos fármacos , Potássio/metabolismo , Proteoma/antagonistas & inibidores , Proteoma/genética , Proteoma/metabolismo , Relação Estrutura-Atividade , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo
12.
J Biol Chem ; 287(24): 20270-80, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22514280

RESUMO

Empedopeptin is a natural lipodepsipeptide antibiotic with potent antibacterial activity against multiresistant Gram-positive bacteria including methicillin-resistant Staphylococcus aureus and penicillin-resistant Streptococcus pneumoniae in vitro and in animal models of bacterial infection. Here, we describe its so far elusive mechanism of antibacterial action. Empedopeptin selectively interferes with late stages of cell wall biosynthesis in intact bacterial cells as demonstrated by inhibition of N-acetylglucosamine incorporation into polymeric cell wall and the accumulation of the ultimate soluble peptidoglycan precursor UDP-N-acetylmuramic acid-pentapeptide in the cytoplasm. Using membrane preparations and the complete cascade of purified, recombinant late stage peptidoglycan biosynthetic enzymes and their respective purified substrates, we show that empedopeptin forms complexes with undecaprenyl pyrophosphate containing peptidoglycan precursors. The primary physiological target of empedopeptin is undecaprenyl pyrophosphate-N-acetylmuramic acid(pentapeptide)-N-acetylglucosamine (lipid II), which is readily accessible at the outside of the cell and which forms a complex with the antibiotic in a 1:2 molar stoichiometry. Lipid II is bound in a region that involves at least the pyrophosphate group, the first sugar, and the proximal parts of stem peptide and undecaprenyl chain. Undecaprenyl pyrophosphate and also teichoic acid precursors are bound with lower affinity and constitute additional targets. Calcium ions are crucial for the antibacterial activity of empedopeptin as they promote stronger interaction with its targets and with negatively charged phospholipids in the membrane. Based on the high structural similarity of empedopeptin to the tripropeptins and plusbacins, we propose this mechanism of action for the whole compound class.


Assuntos
Cálcio/metabolismo , Parede Celular/metabolismo , Farmacorresistência Bacteriana Múltipla , Peptidoglicano/metabolismo , Staphylococcus aureus/metabolismo , Streptococcus pneumoniae/metabolismo , Acetilglucosamina/metabolismo , Membrana Celular/metabolismo , Depsipeptídeos/metabolismo , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Staphylococcus aureus/crescimento & desenvolvimento , Streptococcus pneumoniae/crescimento & desenvolvimento , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Uridina Difosfato Ácido N-Acetilmurâmico/biossíntese , Açúcares de Uridina Difosfato/metabolismo
13.
PLoS Pathog ; 8(1): e1002509, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22291598

RESUMO

The peptidoglycan of Staphylococcus aureus is characterized by a high degree of crosslinking and almost completely lacks free carboxyl groups, due to amidation of the D-glutamic acid in the stem peptide. Amidation of peptidoglycan has been proposed to play a decisive role in polymerization of cell wall building blocks, correlating with the crosslinking of neighboring peptidoglycan stem peptides. Mutants with a reduced degree of amidation are less viable and show increased susceptibility to methicillin. We identified the enzymes catalyzing the formation of D-glutamine in position 2 of the stem peptide. We provide biochemical evidence that the reaction is catalyzed by a glutamine amidotransferase-like protein and a Mur ligase homologue, encoded by SA1707 and SA1708, respectively. Both proteins, for which we propose the designation GatD and MurT, are required for amidation and appear to form a physically stable bi-enzyme complex. To investigate the reaction in vitro we purified recombinant GatD and MurT His-tag fusion proteins and their potential substrates, i.e. UDP-MurNAc-pentapeptide, as well as the membrane-bound cell wall precursors lipid I, lipid II and lipid II-Gly5. In vitro amidation occurred with all bactoprenol-bound intermediates, suggesting that in vivo lipid II and/or lipid II-Gly5 may be substrates for GatD/MurT. Inactivation of the GatD active site abolished lipid II amidation. Both, murT and gatD are organized in an operon and are essential genes of S. aureus. BLAST analysis revealed the presence of homologous transcriptional units in a number of gram-positive pathogens, e.g. Mycobacterium tuberculosis, Streptococcus pneumonia and Clostridium perfringens, all known to have a D-iso-glutamine containing PG. A less negatively charged PG reduces susceptibility towards defensins and may play a general role in innate immune signaling.


Assuntos
Proteínas de Bactérias/metabolismo , Parede Celular/enzimologia , Complexos Multienzimáticos/metabolismo , Infecções Estafilocócicas/enzimologia , Staphylococcus aureus/enzimologia , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Catálise , Parede Celular/genética , Humanos , Imunidade Inata/fisiologia , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/imunologia , Mutação , Óperon/fisiologia , Transdução de Sinais/fisiologia , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Uridina Difosfato Ácido N-Acetilmurâmico/genética , Uridina Difosfato Ácido N-Acetilmurâmico/imunologia , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo
14.
J Biol Chem ; 286(45): 38844-53, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21937437

RESUMO

MraY translocase catalyzes the first committed membrane-bound step of bacterial peptidoglycan synthesis leading to the formation of lipid I. The essential membrane protein therefore has a high potential as target for drug screening approaches to develop antibiotics against gram-positive as well as gram-negative bacteria. However, the production of large integral membrane proteins in conventional cellular expression systems is still very challenging. Cell-free expression technologies have been optimized in recent times for the production of membrane proteins in the presence of detergents (D-CF), lipids (L-CF), or as precipitates (P-CF). We report the development of preparative scale production protocols for the MraY homologues of Escherichia coli and Bacillus subtilis in all three cell-free expression modes followed by their subsequent quality evaluation. Although both proteins can be cell-free produced at comparable high levels, their requirements for optimal expression conditions differ markedly. B. subtilus MraY was stably folded in all three expression modes and showed highest translocase activities after P-CF production followed by defined treatment with detergents. In contrast, the E. coli MraY appears to be unstable after post- or cotranslational solubilization in detergent micelles. Expression kinetics and reducing conditions were identified as optimization parameters for the quality improvement of E. coli MraY. Most remarkably, in contrast to B. subtilis MraY the E. coli MraY has to be stabilized by lipids and only the production in the L-CF mode in the presence of preformed liposomes resulted in stable and translocase active protein samples.


Assuntos
Bacillus subtilis , Proteínas de Bactérias/biossíntese , Escherichia coli , Expressão Gênica , Dobramento de Proteína , Proteínas Recombinantes/biossíntese , Transferases/biossíntese , Proteínas de Bactérias/genética , Sistema Livre de Células/química , Cinética , Proteínas Recombinantes/genética , Transferases/genética , Transferases (Outros Grupos de Fosfato Substituídos)
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...