Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Am Chem Soc ; 146(33): 23449-23456, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39133525

RESUMO

Natural products are important precursors for antibiotic drug design. These chemical scaffolds serve as synthetic inspiration for chemists who leverage their structures to develop novel antibacterials and chemical probes. We have previously studied carolacton, a natural product macrolactone fromSorangium cellulosum, and discovered a simplified derivative, A2, that maintained apparent biofilm inhibitory activity, although the biological target was unknown. Herein, we utilize affinity-based protein profiling (AfBPP) in situ during biofilm formation to identify the protein target using a photoexcitable cross-linking derivative of A2. From these studies, we identified glucan binding protein B (GbpB), a peptidoglycan hydrolase, as the primary target of A2. Further characterization of the interaction between A2 and GbpB, as well as PcsB, a closely related homologue from the more pathogenic S. pneumoniae, revealed binding to the catalytic CHAP (cysteine, histidine, aminopeptidase) domain. To the best of our knowledge, this is the first report of a small-molecule binder of a conserved and essential bacterial CHAP hydrolase, revealing its potential as an antibiotic target. This work also highlights A2 as a useful tool compound for streptococci and as an initial scaffold for the design of more potent CHAP binders.


Assuntos
Biofilmes , Biofilmes/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Antibacterianos/farmacologia , Antibacterianos/química , Sondas Moleculares/química , Sondas Moleculares/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , Lactonas/química , Lactonas/metabolismo , Lactonas/farmacologia , Hidrolases/metabolismo , Hidrolases/química , Hidrolases/antagonistas & inibidores
2.
Mol Microbiol ; 115(4): 684-698, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33140473

RESUMO

Autolysin is a lytic enzyme that hydrolyzes peptidoglycans of the bacterial cell wall, with a catalytic domain and cell wall-binding (CWB) domains, to be involved in different physiological functions that require bacterial cell wall remodeling. We identified a novel autolysin, Acd24020, from Clostridioides (Clostridium) difficile (C. difficile), with an endopeptidase catalytic domain belonging to the NlpC/P60 family and three bacterial Src-homology 3 domains as CWB domains. The catalytic domain of Acd24020 (Acd24020-CD) exhibited C. difficile-specific lytic activity equivalent to Acd24020, indicating that Acd24020-CD has full-function as a lytic enzyme by itself. To elucidate the specific peptidoglycan-recognition and catalytic reaction mechanisms of Acd24020-CD, biochemical characterization, X-ray structure determination, a modeling study of the enzyme/substrate complex, and mutagenesis analysis were performed. Acd24020-CD has an hourglass-shaped substrate-binding groove across the molecule, which is responsible for recognizing the direct 3-4 cross-linking structure unique to C. difficile peptidoglycan. Based on the X-ray structure and modeling study, we propose a dynamic Cys/His catalyzing mechanism, in which the catalytic Cys299 and His354 residues dynamically change their conformations to complement each step of the enzyme catalytic reaction.


Assuntos
Clostridioides difficile/química , Clostridioides difficile/fisiologia , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/fisiologia , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/fisiologia , Domínio Catalítico , Parede Celular/metabolismo , Clostridioides difficile/enzimologia , Cristalografia por Raios X , Hidrólise , Modelos Moleculares , Mutagênese , N-Acetil-Muramil-L-Alanina Amidase/isolamento & purificação , Peptidoglicano/metabolismo , Conformação Proteica , Domínios Proteicos
3.
Biomolecules ; 10(2)2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32028649

RESUMO

D-amino acid oxidase (DAAO) catalyzes the oxidation of D-amino acids generating hydrogen peroxide, a potential producer of reactive oxygen species. In this study, we used a CLytA-DAAO chimera, both free and bound to magnetic nanoparticles, against colon carcinoma, pancreatic adenocarcinoma, and glioblastoma cell lines. We found that the enzyme induces cell death in most of the cell lines tested and its efficiency increases significantly when it is immobilized in nanoparticles. We also tested this enzyme therapy in non-tumor cells, and we found that there is not cell death induction, or it is significantly lower than in tumor cells. The mechanism triggering cell death is apparently a classical apoptosis pathway in the glioblastoma cell lines, while in colon and pancreatic carcinoma cell lines, CLytA-DAAO-induced cell death is a necrosis. Our results constitute a proof of concept that an enzymatic therapy, based on magnetic nanoparticles-delivering CLytA-DAAO, could constitute a useful therapy against cancer and besides it could be used as an enhancer of other treatments such as epigenetic therapy, radiotherapy, and treatments based on DNA repair.


Assuntos
Apoptose , Colina/química , D-Aminoácido Oxidase/química , Nanopartículas de Magnetita/química , N-Acetil-Muramil-L-Alanina Amidase/química , Necrose , Células 3T3-L1 , Adenocarcinoma/patologia , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Neoplasias do Colo/patologia , Dano ao DNA , Reparo do DNA , Glioblastoma/patologia , Humanos , Concentração Inibidora 50 , Camundongos , Neoplasias Pancreáticas/patologia , Espécies Reativas de Oxigênio/química
4.
Microbiology (Reading) ; 165(9): 1013-1023, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31264955

RESUMO

Mycobacteriophages are viruses that infect and kill mycobacteria. The peptidoglycan hydrolase, lysin A (LysA), coded by one of the most potent mycobacteriophages, D29, carries two catalytic domains at its N-terminus and a cell wall-binding domain at its C-terminus. Here, we have explored the importance of the centrally located lysozyme-like catalytic domain (LD) of LysA in phage physiology. We had previously identified an R198A substitution that causes inactivation of the LD when it is present alone on a polypeptide. Here, we show that upon incorporation of the same mutation (i.e. R350A) in full-length LysA, the protein demonstrates substantially reduced activity in vitro, even in the presence of the N-terminal catalytic domain, and has less efficient mycobacterial cell lysis ability when it is expressed in Mycobacterium smegmatis. These data suggest that an active LD is required for the full-length protein to function optimally. Moreover, a mutant D29 phage harbouring this substitution (D29R350A) in its LysA protein shows significantly delayed host M. smegmatis lysis. However, the mutant phage demonstrates an increase in burst size and plaque diameter. Taken together, our data show the importance of an intact LD region in D29 LysA PG hydrolase, and indicate an evolutionary advantage over other phages that lack such a domain in their endolysins.


Assuntos
Endopeptidases/genética , Micobacteriófagos , Mycobacterium smegmatis/virologia , N-Acetil-Muramil-L-Alanina Amidase/genética , Domínio Catalítico/genética , Parede Celular/metabolismo , Endopeptidases/química , Endopeptidases/metabolismo , Mutação , Micobacteriófagos/genética , Micobacteriófagos/crescimento & desenvolvimento , Micobacteriófagos/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
5.
Mol Microbiol ; 110(1): 114-127, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30039535

RESUMO

Chronic infection with Helicobacter pylori can lead to the development of gastric ulcers and stomach cancers. The helical cell shape of H. pylori promotes stomach colonization. Screens for loss of helical shape have identified several periplasmic peptidoglycan (PG) hydrolases and non-enzymatic putative scaffolding proteins, including Csd5. Both over and under expression of the PG hydrolases perturb helical shape, but the mechanism used to coordinate and localize their enzymatic activities is not known. Using immunoprecipitation and mass spectrometry we identified Csd5 interactions with cytosolic proteins CcmA, a bactofilin required for helical shape, and MurF, a PG precursor synthase, as well as the inner membrane spanning ATP synthase. A combination of Csd5 domain deletions, point mutations, and transmembrane domain chimeras revealed that the N-terminal transmembrane domain promotes MurF, CcmA, and ATP synthase interactions, while the C-terminal SH3 domain mediates PG binding. We conclude that Csd5 promotes helical shape as part of a membrane associated, multi-protein shape complex that includes interactions with the periplasmic cell wall, a PG precursor synthesis enzyme, the bacterial cytoskeleton, and ATP synthase.


Assuntos
Parede Celular/metabolismo , Citoesqueleto/metabolismo , Helicobacter pylori/citologia , Helicobacter pylori/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptídeo Sintases/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Membrana Celular/metabolismo , Deleção de Genes , Helicobacter pylori/genética , ATPases Mitocondriais Próton-Translocadoras/química , ATPases Mitocondriais Próton-Translocadoras/genética , ATPases Mitocondriais Próton-Translocadoras/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/genética , Peptídeo Sintases/química , Peptídeo Sintases/genética , Periplasma/metabolismo , Análise de Sequência de Proteína
6.
Appl Environ Microbiol ; 83(23)2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28970228

RESUMO

To provide food security, innovative approaches to preventing plant disease are currently being explored. Here, we demonstrate that lytic bacteriophages and phage lysis proteins are effective at triggering lysis of the phytopathogen Agrobacterium tumefaciens Phages Atu_ph02 and Atu_ph03 were isolated from wastewater and induced lysis of C58-derived strains of A. tumefaciens The coinoculation of A. tumefaciens with phages on potato discs limited tumor formation. The genomes of Atu_ph02 and Atu_ph03 are nearly identical and are ∼42% identical to those of T7 supercluster phages. In silico attempts to find a canonical lysis cassette were unsuccessful; however, we found a putative phage peptidoglycan hydrolase (PPH), which contains a C-terminal transmembrane domain. Remarkably, the endogenous expression of pph in the absence of additional phage genes causes a block in cell division and subsequent lysis of A. tumefaciens cells. When the presumed active site of the N-acetylmuramidase domain carries an inactivating mutation, PPH expression causes extensive cell branching due to a block in cell division but does not trigger rapid cell lysis. In contrast, the mutation of positively charged residues at the extreme C terminus of PPH causes more rapid cell lysis. Together, these results suggest that PPH causes a block in cell division and triggers cell lysis through two distinct activities. Finally, the potent killing activity of this single lysis protein can be modulated, suggesting that it could be engineered to be an effective enzybiotic.IMPORTANCE The characterization of bacteriophages such as Atu_ph02 and Atu_ph03, which infect plant pathogens such as Agrobacterium tumefaciens, may be the basis of new biocontrol strategies. First, cocktails of diverse bacteriophages could be used as a preventative measure to limit plant diseases caused by bacteria; a bacterial pathogen is unlikely to simultaneously develop resistances to multiple bacteriophage species. The specificity of bacteriophage treatment for the host is an asset in complex communities, such as in orchards where it would be detrimental to harm the symbiotic bacteria in the environment. Second, bacteriophages are potential sources of enzymes that efficiently lyse bacterial cells. These phage proteins may have a broad specificity, but since proteins do not replicate as phages do, their effect is highly localized, providing an alternative to traditional antibiotic treatments. Thus, studies of lytic bacteriophages that infect A. tumefaciens may provide insights for designing preventative strategies against bacterial pathogens.


Assuntos
Agrobacterium tumefaciens/fisiologia , Bacteriólise , Bacteriófagos/enzimologia , Expressão Gênica , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Proteínas Virais/metabolismo , Agrobacterium tumefaciens/genética , Bacteriófagos/classificação , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/genética , Filogenia , Domínios Proteicos , Proteínas Virais/química , Proteínas Virais/genética , Águas Residuárias/virologia
7.
Protein Sci ; 25(4): 840-51, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26799947

RESUMO

A subset of proteins containing NlpC/P60 domains are bacterial peptidoglycan hydrolases that cleave noncanonical peptide linkages and contribute to cell wall remodeling as well as cell separation during late stages of division. Some of these proteins have been shown to cleave peptidoglycan in Mycobacterium tuberculosis and play a role in Mycobacterium marinum virulence of zebra fish; however, there are still significant knowledge gaps concerning the molecular function of these proteins in Mycobacterium avium subspecies paratuberculosis (MAP). The MAP genome sequence encodes five NlpC/P60 domain-containing proteins. We describe atomic resolution crystal structures of two such MAP proteins, MAP_1272c and MAP_1204. These crystal structures, combined with functional assays to measure peptidoglycan cleavage activity, led to the observation that MAP_1272c does not have a functional catalytic core for peptidoglycan hydrolysis. Furthermore, the structure and sequence of MAP_1272c demonstrate that the catalytic residues normally required for hydrolysis are absent, and the protein does not bind peptidoglycan as efficiently as MAP_1204. While the NlpC/P60 catalytic triad is present in MAP_1204, changing the catalytic cysteine-155 residue to a serine significantly diminished catalytic activity, but did not affect binding to peptidoglycan. Collectively, these findings suggest a broader functional repertoire for NlpC/P60 domain-containing proteins than simply hydrolases.


Assuntos
Mycobacterium avium subsp. paratuberculosis/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptidoglicano/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Domínio Catalítico , Parede Celular , Cristalografia por Raios X , Hidrólise , Modelos Moleculares , Mycobacterium avium subsp. paratuberculosis/química , Domínios Proteicos
8.
Sci Rep ; 5: 11886, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26156658

RESUMO

We have previously designed a series of antimicrobial peptides (AMPs) and in the current study, the in vivo therapeutic efficacy and toxicity were investigated. Among all the peptides, DM3 conferred protection to a substantial proportion of the lethally infected mice caused by a strain of penicillin-resistant Streptococcus pneumoniae. Synergism was reported and therapeutic efficacy was significantly enhanced when DM3 was formulated in combination with penicillin (PEN). No toxicity was observed in mice receiving these treatments. The in silico molecular docking study results showed that, DM3 has a strong affinity towards three protein targets; autolysin and pneumococcal surface protein A (pspA). Thus AMPs could serve as supporting therapeutics in combination with conventional antibiotics to enhance treatment outcome.


Assuntos
Antibacterianos/farmacologia , Penicilina G/farmacologia , Peptídeos/farmacologia , Infecções Pneumocócicas/microbiologia , Streptococcus pneumoniae/efeitos dos fármacos , Animais , Antibacterianos/administração & dosagem , Antibacterianos/química , Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Modelos Animais de Doenças , Sinergismo Farmacológico , Humanos , Camundongos , Modelos Moleculares , Simulação de Acoplamento Molecular , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptídeos/química , Infecções Pneumocócicas/tratamento farmacológico , Infecções Pneumocócicas/mortalidade , Infecções Pneumocócicas/patologia , Ligação Proteica , Conformação Proteica , Streptococcus pneumoniae/metabolismo , Estreptolisinas/química , Estreptolisinas/metabolismo
9.
Chemistry ; 21(22): 8076-89, 2015 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-25917218

RESUMO

Choline-binding modules (CBMs) have a ßß-solenoid structure composed of choline-binding repeats (CBR), which consist of a ß-hairpin followed by a short linker. To find minimal peptides that are able to maintain the CBR native structure and to evaluate their remaining choline-binding ability, we have analysed the third ß-hairpin of the CBM from the pneumococcal LytA autolysin. Circular dichroism and NMR data reveal that this peptide forms a highly stable native-like ß-hairpin both in aqueous solution and in the presence of trifluoroethanol, but, strikingly, the peptide structure is a stable amphipathic α-helix in both zwitterionic (dodecylphosphocholine) and anionic (sodium dodecylsulfate) detergent micelles, as well as in small unilamellar vesicles. This ß-hairpin to α-helix conversion is reversible. Given that the ß-hairpin and α-helix differ greatly in the distribution of hydrophobic and hydrophilic side chains, we propose that the amphipathicity is a requirement for a peptide structure to interact and to be stable in micelles or lipid vesicles. To our knowledge, this "chameleonic" behaviour is the only described case of a micelle-induced structural transition between two ordered peptide structures.


Assuntos
Proteínas de Bactérias/química , Colina/metabolismo , Micelas , N-Acetil-Muramil-L-Alanina Amidase/química , Peptídeos/química , Streptococcus pneumoniae/química , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Dicroísmo Circular , Humanos , Modelos Moleculares , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Ressonância Magnética Nuclear Biomolecular , Peptídeos/metabolismo , Fosforilcolina/análogos & derivados , Fosforilcolina/química , Infecções Pneumocócicas/microbiologia , Estrutura Secundária de Proteína , Dodecilsulfato de Sódio/química , Streptococcus pneumoniae/metabolismo , Tensoativos/química , Trifluoretanol/química
10.
Virology ; 477: 125-132, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25432575

RESUMO

Endolysins are lytic enzymes encoded by bacteriophage that represent an emerging class of protein therapeutics. Considering macromolecular thermoresistance correlates with shelf life, PlyG, a Bacillus anthracis endolysin, was thermally characterized to further evaluate its therapeutic potential. Results from a biophysical thermal analysis revealed full-length PlyG and its isolated domains comprised thermal denaturation temperatures exceeding 63°C. In the absence of reducing agent, PlyG was determined to be kinetically unstable, a finding hypothesized to be attributable to the chemical oxidation of cysteine and/or methionine residues. The presence of reducing agent kinetically stabilized the endolysin, with PlyG retaining at least ~50% residual lytic activity after being heated at temperatures up to 80°C and remaining enzymatically functional after being boiled. Furthermore, the endolysin had a kinetic half-life at 50°C and 55°C of 35 and 5.5h, respectively. PlyG represents a thermostable proteinaceous antibacterial with subsequent prolonged therapeutic shelf life expectancy.


Assuntos
Bacillus anthracis/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/efeitos da radiação , Proteínas Virais/química , Proteínas Virais/efeitos da radiação , Antibacterianos/química , Antibacterianos/efeitos da radiação , Estabilidade Enzimática , Cinética , Desnaturação Proteica/efeitos da radiação , Estabilidade Proteica , Temperatura
11.
Cell Commun Adhes ; 21(5): 257-65, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24978154

RESUMO

Bacillus anthracis is a pathogenic, Gram-positive bacterium which chiefly affects the livestock of animals and humans through acute disease anthrax. All around the globe this bio-threat organism damages millions of lives in every year and also most of the drugs were not responding properly in inhibition against this diseased pathogen. In recent development, phage therapy is considered as alternative solution to treat this serious infectious disease. In this study, we elucidated the binding of γ phage lysin plyG enzymes toward the SrtA along with its activator peptide LPXTG. Through protein-protein docking and molecular dynamics simulation studies, we showed the distinguished structure complementarity of SrtA and plyG complex. Especially, MD simulation relates strong and stable interaction occurs between the protein complex structures. These results suggest that additional experimental studies on our approach will lead to availability of better inhibitor against the SrtA.


Assuntos
Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Simulação de Dinâmica Molecular , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Aminoaciltransferases/química , Bacillus anthracis/metabolismo , Proteínas de Bactérias/química , Bacteriófagos/metabolismo , Sítios de Ligação , Cisteína Endopeptidases/química , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , N-Acetil-Muramil-L-Alanina Amidase/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Virais/química
12.
Chembiochem ; 15(3): 425-34, 2014 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-24436194

RESUMO

Glycan microarrays have emerged as novel tools to study carbohydrate-protein interactions. Here we describe the preparation of a covalent microarray with lipochitin oligosaccharides and its use in studying proteins containing LysM domains. The glycan microarray was assembled from glycoconjugates that were synthesized by using recently developed bifunctional chemoselective aminooxy reagents without the need for transient carbohydrate protecting groups. We describe for the first time the preparation of a covalent microarray with lipochitin oligosaccharides and its use for studying proteins containing LysM domains. Lipochitin oligosaccharides (also referred to as Nod factors) were isolated from bacterial strains or chemoenzymatically synthesized. The glycan microarray also included peptidoglycan-related compounds, as well as chitin oligosaccharides of different lengths. In total, 30 ligands were treated with the aminooxy linker molecule. The identity of the glycoconjugates was verified by mass spectrometry, and they were then immobilized on the array. The presence of the glycoconjugates on the array surface was confirmed by use of lectins and human sera (IgG binding). The functionality of our array was tested with a bacterial LysM domain-containing protein, autolysin p60, which is known to act on the bacterial cell wall peptidoglycan. P60 showed specific binding to Nod factors and to chitin oligosaccharides. Increasing affinity was observed with increasing chitin oligomer length.


Assuntos
Proteínas de Bactérias/metabolismo , Glicoconjugados/química , Lipopolissacarídeos/química , Análise em Microsséries/métodos , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Oximas/química , Proteínas de Bactérias/química , Glicoconjugados/metabolismo , Humanos , Imunoglobulina G/imunologia , Lectinas/química , Lectinas/metabolismo , Ligantes , Lipopolissacarídeos/isolamento & purificação , Listeria monocytogenes/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , Peptídeos/síntese química , Peptídeos/química , Peptidoglicano/química , Peptidoglicano/metabolismo , Ligação Proteica
13.
Appl Biochem Biotechnol ; 172(4): 1790-806, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24264995

RESUMO

Bacillus anthracis is a well-known bioweapon pathogen, which coordinates the expression of its virulence factors in response to a specific environmental signal by its protein architecture. Absences of sortase signal functioning may fail to assemble the surface linked proteins and so B. anthracis cannot sustain an infection with host cells. Targeting the signaling mechanism of B. anthracis can be achieved by inhibition of SrtA enzyme through λphage-derived plyG. The lysin enzyme plyG is experimentally proven as bacteriolytic agent, specifically kill's B. anthracis by inhibiting the SrtA. Here, we have screened the peptides from λphage lysin, and these peptides are having the ability as LPXTG competitive inhibitors. In comparison to the activator peptide LPXTG binding motif, λphage lysin based inhibitor peptides are having much supremacy towards binding of SrtA. Finally, peptide structures extracted from PlyG are free from toxic, allergic abilities and also have the ability to terminate the signal transduction mechanism in B. anthracis.


Assuntos
Aminoaciltransferases/metabolismo , Bacillus anthracis/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , Peptídeos/farmacologia , Proteínas Virais/química , Antibacterianos/química , Antibacterianos/farmacologia , Peptídeos/química
14.
J Biol Chem ; 288(52): 36915-25, 2013 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-24235140

RESUMO

Enterococcus faecalis strains are commensal bacteria in humans and other animals, and they are also the causative agent of opportunistic infectious diseases. Bacteriocin 41 (Bac41) is produced by certain E. faecalis clinical isolates, and it is active against other E. faecalis strains. Our genetic analyses demonstrated that the extracellular products of the bacL1 and bacA genes, which are encoded in the Bac41 operon, coordinately express the bacteriocin activity against E. faecalis. In this study, we investigated the molecular functions of the BacL1 and BacA proteins. Immunoblotting and N-terminal amino acid sequence analysis revealed that BacL1 and BacA are secreted without any processing. The coincidental treatment with the recombinant BacL1 and BacA showed complete bacteriocin activity against E. faecalis, but neither BacL1 nor BacA protein alone showed the bacteriocin activity. Interestingly, BacL1 alone demonstrated substantial degrading activity against the cell wall fraction of E. faecalis in the absence of BacA. Furthermore, MALDI-TOF MS analysis revealed that BacL1 has a peptidoglycan D-isoglutamyl-L-lysine endopeptidase activity via a NlpC/P60 homology domain. These results collectively suggest that BacL1 serves as a peptidoglycan hydrolase and, when BacA is present, results in the lysis of viable E. faecalis cells.


Assuntos
Bacteriocinas/química , Endopeptidases/química , Enterococcus faecalis/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/química , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteriocinas/genética , Bacteriocinas/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo , Enterococcus faecalis/genética , N-Acetil-Muramil-L-Alanina Amidase/genética , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Estrutura Terciária de Proteína
15.
Biochem J ; 448(2): 201-11, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22931054

RESUMO

Tse1 (Tse is type VI secretion exported), an effector protein produced by Pseudomonas aeruginosa, is an amidase that hydrolyses the γ-D-glutamyl-DAP (γ-D-glutamyl-L-meso-diaminopimelic acid) linkage of the peptide bridge of peptidoglycan. P. aeruginosa injects Tse1 into the periplasm of recipient cells, degrading their peptidoglycan, thereby helping itself to compete with other bacteria. Meanwhile, to protect itself from injury by Tse1, P. aeruginosa expresses the cognate immunity protein Tsi1 (Tsi is type VI secretion immunity) in its own periplasm to inactivate Tse1. In the present paper, we report the crystal structures of Tse1 and the Tse1-(6-148)-Tsi1-(20-end) complex at 1.4 Å and 1.6 Å (1 Å=0.1 nm) resolutions respectively. The Tse1 structure adopts a classical papain-like α+ß fold. A cysteine-histidine catalytic diad is identified in the reaction centre of Tse1 by structural comparison and mutagenesis studies. Tsi1 binds Tse1 tightly. The HI loop (middle finger tip) from Tsi1 inserts into the large pocket of the Y-shaped groove on the surface of Tse1, and CD, EF, JK and LM loops (thumb, index finger, ring finger and little finger tips) interact with Tse1, thus blocking the binding of enzyme to peptidoglycan. The catalytic and inhibition mechanisms provide new insights into how P. aeruginosa competes with others and protects itself.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Pseudomonas aeruginosa/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Domínio Catalítico , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multiproteicos/química , Mutagênese Sítio-Dirigida , N-Acetil-Muramil-L-Alanina Amidase/genética , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Pseudomonas aeruginosa/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
16.
PLoS One ; 7(6): e40353, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22768285

RESUMO

BACKGROUND: Staphylococcus aureus is a frequent cause of serious and life-threatening infections, such as endocarditis, osteomyelitis, pneumonia, and sepsis. Its adherence to various host structures is crucial for the establishment of diseases. Adherence may be mediated by a variety of adhesins, among them the autolysin/adhesins Atl and Aaa. Aaa is composed of three N-terminal repeated sequences homologous to a lysin motif (LysM) that can confer cell wall attachment and a C-terminally located cysteine, histidine-dependent amidohydrolase/peptidase (CHAP) domain having bacteriolytic activity in many proteins. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show by surface plasmon resonance that the LysM domain binds to fibrinogen, fibronectin, and vitronectin respresenting a novel adhesive function for this domain. Moreover, we demonstrated that the CHAP domain not only mediates the bacteriolytic activity, but also adherence to fibrinogen, fibronectin, and vitronectin, thus demonstrating for the first time an adhesive function for this domain. Adherence of an S. aureus aaa mutant and the complemented aaa mutant is slightly decreased and increased, respectively, to vitronectin, but not to fibrinogen and fibronectin, which might at least in part result from an increased expression of atl in the aaa mutant. Furthermore, an S. aureus atl mutant that showed enhanced adherence to fibrinogen, fibronectin, and endothelial cells also demonstrated increased aaa expression and production of Aaa. Thus, the redundant functions of Aaa and Atl might at least in part be interchangeable. Lastly, RT-PCR and zymographic analysis revealed that aaa is negatively regulated by the global virulence gene regulators agr and SarA. CONCLUSIONS/SIGNIFICANCE: We identified novel functions for two widely distributed protein domains, LysM and CHAP, i.e. the adherence to the extracellular matrix proteins fibrinogen, fibronectin, and vitronectin. The adhesive properties of Aaa might promote S. aureus colonization of host extracellular matrix and tissue, suggesting a role for Aaa in the pathogenesis of S. aureus infections.


Assuntos
Adesinas Bacterianas/química , N-Acetil-Muramil-L-Alanina Amidase/química , Staphylococcus aureus/metabolismo , Adesinas Bacterianas/metabolismo , Fibrinogênio/metabolismo , Fibronectinas/metabolismo , Teste de Complementação Genética , Humanos , Mutação/genética , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Staphylococcus aureus/patogenicidade , Ressonância de Plasmônio de Superfície , Fatores de Virulência/metabolismo , Vitronectina/metabolismo
17.
J Bacteriol ; 194(6): 1464-73, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22228730

RESUMO

Streptococcus suis serotype 2 (S. suis 2) is an important swine and human pathogen responsible for septicemia and meningitis. A novel gene, designated atl and encoding a major autolysin of S. suis 2 virulent strain HA9801, was identified and characterized in this study. The Atl protein contains 1,025 amino acids with a predicted molecular mass of 113 kDa and has a conserved N-acetylmuramoyl-l-alanine amidase domain. Recombinant Atl was expressed in Escherichia coli, and its bacteriolytic and fibronectin-binding activities were confirmed by zymography and Western affinity blotting. Two bacteriolytic bands were shown in the sodium dodecyl sulfate extracts of HA9801, while both were absent from the atl inactivated mutant. Cell chains of the mutant strain became longer than that of the parental strain. In the autolysis assay, HA9801 decreased to 20% of the initial optical density (OD) value, while the mutant strain had almost no autolytic activity. The biofilm capacity of the atl mutant was reduced ∼30% compared to the parental strain. In the zebrafish infection model, the 50% lethal dose of the mutant strain was increased up to 5-fold. Furthermore, the adherence to HEp-2 cells of the atl mutant was 50% less than that of the parental strain. Based on the functional analysis of the recombinant Atl and observed effects of atl inactivation on HA9801, we conclude that Atl is a major autolysin of HA9801. It takes part in cell autolysis, separation of daughter cells, biofilm formation, fibronectin-binding activity, cell adhesion, and pathogenesis of HA9801.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/genética , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Streptococcus suis/enzimologia , Animais , Proteínas de Bactérias/química , Biofilmes/crescimento & desenvolvimento , Adesão Celular , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Fibronectinas/metabolismo , Deleção de Genes , Expressão Gênica , Células Hep G2 , Humanos , Dose Letal Mediana , Peso Molecular , N-Acetil-Muramil-L-Alanina Amidase/química , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Infecções Estreptocócicas/microbiologia , Streptococcus suis/genética , Streptococcus suis/crescimento & desenvolvimento , Streptococcus suis/metabolismo , Análise de Sobrevida , Virulência , Peixe-Zebra
18.
FEMS Microbiol Lett ; 290(1): 105-13, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19025571

RESUMO

The nucleotide sequence of atlL, a gene encoding a putative Staphylococcus lugdunensis peptidoglycan hydrolase, was determined using degenerate consensus PCR and genome walking. This 3837-bp gene encodes a protein, AtlL, that appears as a putative bifunctional autolysin with a 29-amino acid putative signal peptide and two enzymatic putative centres (N-acetylmuramoyl-l-alanine amidase and N-acetylglucosaminidase) interconnected with three imperfect repeated sequences displaying glycine-tryptophan motifs. In order to determine whether both lytic domains were functional, and verify their exact enzymatic activities, gene fragments harbouring both putative domains, AM (N-acetylmuramoyl-l-alanine amidase enzymatic centre plus two repeated sequences) and GL (N-acetylglucosaminidase enzymatic centre plus one repeated sequence), were isolated, subcloned, and expressed in Escherichia coli. Purified recombinant AM and GL protein truncations exhibited cell wall lytic activity in zymograms performed with cell walls of Micrococcus lysodeikticus, Bacillus subtilis, and S. lugdunensis. AtlL is expressed during the whole growth, with an overexpression in the early-exponential stage. Liquid chromatography-mass spectrometry analysis of muropeptides generated by digestion of B. subtilis cell walls demonstrated the hydrolytic bond specificities and confirmed both of the acetyl domains' activities as predicted by sequence homology data. AtlL is the first autolysin described in S. lugdunensis, with a bifunctional enzymatic activity involved in peptidoglycan hydrolysis.


Assuntos
Acetilglucosaminidase/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Staphylococcus/enzimologia , Acetilglucosaminidase/química , Acetilglucosaminidase/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteriólise , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/genética , Peptidoglicano/química , Peptidoglicano/metabolismo , Reação em Cadeia da Polimerase , Staphylococcus/genética , Staphylococcus/crescimento & desenvolvimento
19.
Artigo em Inglês | MEDLINE | ID: mdl-18776453

RESUMO

The protein-protein interaction presides the various biological events in life. Toward the understanding of their functions and networks, various techniques to regulate the specific protein functions are developed and applied so far. Here we examined the novel method to regulate the protein-protein interactions via coiled-coil assembly.


Assuntos
RNA Polimerases Dirigidas por DNA/química , N-Acetil-Muramil-L-Alanina Amidase/química , Peptídeos/química , Proteínas Virais/química , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , Ligantes , Dobramento de Proteína , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , RNA/biossíntese , Proteínas Virais/antagonistas & inibidores
20.
Biochem Biophys Res Commun ; 374(4): 747-51, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18671939

RESUMO

Gp181 (2237 amino acids) of Pseudomonas aeruginosa bacteriophage phiKZ (Myoviridae) is a structural virion protein, which bears a peptidoglycan hydrolase domain near its C-terminus. This protein is supposed to degrade the peptidoglycan locally during the infection process. Nine deletional mutants allowed delineation of the peptidoglycan hydrolase domain between amino acids 1880-2042 (gp181M8) and analysis of its biochemical properties. Gp181M8 tolerates a high ionic strength (>320mM) and is less sensitive to long thermal treatments compared to the similar phiKZ endolysin. Gp181M8 lysed all tested outer membrane-permeabilized Gram-negative species. The C-terminal distal end (amino acids 2043-2237) enhances the specific activity of gp181M8 threefold, resulting in a twelve times higher activity than commercial hen egg white lysozyme. These biochemical properties suggest that this novel peptidoglycan hydrolase domain may be suitable for enzybiotic applications.


Assuntos
Myoviridae/enzimologia , N-Acetil-Muramil-L-Alanina Amidase/química , Fagos de Pseudomonas/enzimologia , Pseudomonas aeruginosa/virologia , Proteínas Estruturais Virais/química , Catálise , Clonagem Molecular , Estabilidade Enzimática , Hidrólise , N-Acetil-Muramil-L-Alanina Amidase/genética , Concentração Osmolar , Peptidoglicano/química , Estrutura Terciária de Proteína , Deleção de Sequência , Especificidade por Substrato , Proteínas Estruturais Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA