Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Biol Macromol ; 271(Pt 1): 132563, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38782313

RESUMO

Staphylococcus aureus possesses the ability to become pathogenic, leading to severe and life-threatening infections. Its methicillin-resistant variant MRSA has garnered high-priority status due to its increased morbidity and associated mortality. This emphasizes the urgency for novel anti-staphylococcal agents. The bacteriocin lysostaphin stands out for its remarkable bactericidal activity against S. aureus, including MRSA, outperforming conventional antibiotics. However, the clinical application of lysostaphin faces challenges, including enzymatic activity loss under physiological conditions and potential immunogenicity. This study introduces a novel approach by encapsulating lysostaphin within polylactic-co-glycolic acid (PLGA) nanoparticles, a biodegradable copolymer known for its biocompatibility and sustained drug release ability. The study assesses the antimicrobial activity of lysostaphin-loaded PLGA nanoparticles against different S. aureus strains, and we also used GFP-expressing S. aureus for facilitating its traceability in planktonic, biofilm, and intracellular infection models. The results showed the significant reduction in bacteria viability both in planktonic and biofilm states. The in vitro intracellular infection model demonstrated the significantly enhanced efficiency of the developed nanoparticles compared to the treatment with the free bacteriocin. This research presents lysostaphin encapsulation within PLGA nanoparticles and offers promising avenues for enhancing lysostaphin's therapeutic efficacy against S. aureus infections.


Assuntos
Antibacterianos , Biofilmes , Lisostafina , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Infecções Estafilocócicas , Staphylococcus aureus , Lisostafina/química , Lisostafina/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Nanopartículas/química , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/efeitos dos fármacos , Antibacterianos/farmacologia , Antibacterianos/química , Biofilmes/efeitos dos fármacos , Testes de Sensibilidade Microbiana , Humanos , Portadores de Fármacos/química
2.
Nat Commun ; 14(1): 6706, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37872144

RESUMO

Peptidoglycan, a gigadalton polymer, functions as the scaffold for bacterial cell walls and provides cell integrity. Peptidoglycan is remodelled by a large and diverse group of peptidoglycan hydrolases, which control bacterial cell growth and division. Over the years, many studies have focused on these enzymes, but knowledge on their action within peptidoglycan mesh from a molecular basis is scarce. Here, we provide structural insights into the interaction between short peptidoglycan fragments and the entire sacculus with two evolutionarily related peptidases of the M23 family, lysostaphin and LytM. Through nuclear magnetic resonance, mass spectrometry, information-driven modelling, site-directed mutagenesis and biochemical approaches, we propose a model in which peptidoglycan cross-linking affects the activity, selectivity and specificity of these two structurally related enzymes differently.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Peptidoglicano/química , Hidrolases , Lisostafina/análise , Lisostafina/química , Espectrometria de Massas/métodos , Parede Celular/química
3.
Biochem Biophys Res Commun ; 668: 111-117, 2023 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-37245291

RESUMO

Lysostaphin endopeptidase cleaves pentaglycine cross-bridges found in staphylococcal cell-wall peptidoglycans and proves very effective in combatting methicillin-resistant Staphylococcus aureus. Here, we revealed the functional importance of two loop residues, Tyr270 in loop 1 and Asn372 in loop 4, which are highly conserved among the M23 endopeptidase family and are found close to the Zn2+-coordinating active site. Detailed analyses of the binding groove architecture together with protein-ligand docking showed that these two loop residues potentially interact with the docked ligand-pentaglycine. Ala-substituted mutants (Y270A and N372A) were generated and over-expressed in Escherichia coli as a soluble form at levels comparable to the wild type. A drastic decrease in staphylolytic activity against S. aureus was observed for both mutants, suggesting an essential role of the two loop residues in lysostaphin function. Further substitutions with an uncharged polar Gln side-chain revealed that only the Y270Q mutation caused a dramatic reduction in bioactivity. In silico predicting the effect of binding site mutations revealed that all mutations displayed a large ΔΔGbind value, signifying requirements of the two loop residues for efficient binding to pentaglycine. Additionally, MD simulations revealed that Y270A and Y270Q mutations induced large flexibility of the loop 1 region, showing markedly increased RMSF values. Further structural analysis suggested that Tyr270 conceivably participated in the oxyanion stabilization of the enzyme catalysis. Altogether, our present study disclosed that two highly conserved loop residues, loop 1-Tyr270 and loop 4-Asn372, located near the lysostaphin active site are crucially involved in staphylolytic activity toward binding and catalysis of pentaglycine cross-links.


Assuntos
Lisostafina , Staphylococcus aureus Resistente à Meticilina , Lisostafina/química , Lisostafina/metabolismo , Lisostafina/farmacologia , Staphylococcus aureus , Domínio Catalítico , Ligantes , Endopeptidases/genética , Endopeptidases/metabolismo , Catálise
4.
J Agric Food Chem ; 71(13): 5293-5301, 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-36967580

RESUMO

Glycine-rich flexible peptide linkers have been widely adopted in fusion protein engineering; however, they can hardly be cleaved for the separation of fusion partners unless specific protease recognition sites are introduced. Herein, we report the use of the peptidoglycan-targeting staphylolytic enzyme lysostaphin to directly digest the glycine-rich flexible linkers of various lengths including oligoglycine linkers and (G4S)x linkers, without the incorporation of extra amino acids. Using His-MBP-linker-LbCpf1 as a model substrate, we show that both types of linkers could be digested by lysostaphin, and the digestion efficiency improved with increasing linker length. The enzyme LbCpf1 retained full activity after tag removal. We further demonstrated that the proteolytic activity of lysostaphin could be well maintained under different environmental conditions and in the presence of a series of chemical reagents at various concentrations that are frequently used in protein purification and stabilization. In addition, such a digestion strategy could also be applied to remove the SUMO domain linked to LwCas13a via an octaglycine linker. This study extends the applications of lysostaphin beyond an antimicrobial reagent and demonstrates its potential as a novel, efficient, and robust protease for protein engineering.


Assuntos
Lisostafina , Peptídeo Hidrolases , Lisostafina/química , Lisostafina/metabolismo , Peptidoglicano/química , Peptidoglicano/metabolismo , Glicina , Parede Celular/metabolismo
5.
J Agric Food Chem ; 70(37): 11441-11457, 2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36082619

RESUMO

Lysostaphin is a potent bacteriolytic enzyme with endopeptidase activity against the common pathogen Staphylococcus aureus. By digesting the pentaglycine crossbridge in the cell wall peptidoglycan of S. aureus including the methicillin-resistant strains, lysostaphin initiates rapid lysis of planktonic and sessile cells (biofilms) and has great potential for use in agriculture, food industries, and pharmaceutical industries. In the past few decades, there have been tremendous efforts in potentiating lysostaphin for better applications in these fields, including engineering of the enzyme for higher potency and lower immunogenicity with longer-lasting effects, formulation and immobilization of the enzyme for higher stability and better durability, and recombinant expression for low-cost industrial production and in situ biocontrol. These achievements are extensively reviewed in this article focusing on applications in disease control, food preservation, surface decontamination, and pathogen detection. In addition, some basic properties of lysostaphin that have been controversial and only elucidated recently are summarized, including the substrate-binding properties, the number of zinc-binding sites, the substrate range, and the cleavage site in the pentaglycine crossbridge. Resistance to lysostaphin is also highlighted with a focus on various mechanisms. This article is concluded with a discussion on the limitations and future perspectives for the actual applications of lysostaphin.


Assuntos
Lisostafina , Staphylococcus aureus , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Bacteriólise , Lisostafina/química , Lisostafina/metabolismo , Lisostafina/farmacologia , Peptidoglicano/química , Peptidoglicano/metabolismo , Staphylococcus aureus/metabolismo , Zinco/metabolismo
6.
Bioconjug Chem ; 33(5): 767-772, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35499914

RESUMO

Bacterial cell walls represent one of the most prominent targets of antibacterial agents. These agents include natural products (e.g., vancomycin) and proteins stemming from the innate immune system (e.g., peptidoglycan-recognition proteins and lysostaphin). Among bacterial pathogens that infect humans, Staphylococcus aureus (S. aureus) continues to impose a tremendous healthcare burden across the globe. S. aureus has evolved countermeasures that can directly restrict the accessibility of innate immune proteins, effectively protecting itself from threats that target key cell well components. We recently described a novel assay that directly reports on the accessibility of molecules to the peptidoglycan layer within the bacterial cell wall of S. aureus. The assay relies on site-specific chemical remodeling of the peptidoglycan with a biorthogonal handle. Here, we disclose the application of our assay to a screen of a nonredundant transposon mutant library for susceptibility of the peptidoglycan layer with the goal of identifying genes that contribute to the control of cell surface accessibility. We discovered several genes that resulted in higher accessibility levels to the peptidoglycan layer and showed that these genes modulate sensitivity to lysostaphin. These results indicate that this assay platform can be leveraged to gain further insight into the biology of bacterial cell surfaces.


Assuntos
Lisostafina , Staphylococcus aureus , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Parede Celular/química , Humanos , Lisostafina/química , Lisostafina/metabolismo , Lisostafina/farmacologia , Peptidoglicano/química , Vancomicina/metabolismo
7.
Biotechnol Appl Biochem ; 68(5): 1058-1066, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32918836

RESUMO

Antibiotic resistance and the colonization of resistant bacteria such as Staphylococcus aureus on surfaces, often in the form of biofilms, prolong hospitalization periods and increase mortality, thus is a significant concern for healthcare providers. To prevent biofilm formation, the inadequate concentration of using nanoparticles as antibacterial coating agents is one of the major obstacles. This study aimed to design a hypervalency TiO2 nanocomposite as a reserved base to carry a high amount of active antibacterial agents such as lysostaphin via a biotin-streptavidin-biotin bridge. The utilization of the streptavidin-biotin system could increase the abundance of lysostaphin. Lysostaphin was expressed in Escherichia coli and purified. Both recombinant lysostaphin and titanium oxide nanocomposite were conjugated with biotin and linked to a streptavidin bridge. The kinetics and activity of the enzyme were examined after each step utilizing N-acetylhexaglycine as a substrate. Physical characteristics of nanoparticles containing lysostaphin were determined using AFM, SEM, FTIR, and zeta potential. The results showed changes in size, charge, and morphology of the nanoparticles following the lysostaphin attachment. Also, the stability and kinetics of the active biological enzymes on nanoparticles were reexamined following 8 months of storage. Exploiting this approach, various biotinylated antibacterial agents could be prepared and rapidly immobilized on a nanoparticle as an active net against related infectious agents.


Assuntos
Antibacterianos/farmacologia , Lisostafina/metabolismo , Nanopartículas/química , Infecções Estafilocócicas/tratamento farmacológico , Titânio/farmacologia , Antibacterianos/química , Biofilmes/efeitos dos fármacos , Biotina/química , Biotina/metabolismo , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Lisostafina/química , Lisostafina/genética , Tamanho da Partícula , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Infecções Estafilocócicas/metabolismo , Estreptavidina/química , Estreptavidina/metabolismo , Propriedades de Superfície , Titânio/química
8.
Biomolecules ; 10(3)2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-32178236

RESUMO

Staphylococcus aureus is a Gram-positive pathogen that is capable of infecting almost every organ in the human body. Alarmingly, the rapid emergence of methicillin-resistant S.aureus strains (MRSA) jeopardizes the available treatment options. Herein, we propose sustainable, low-cost production of recombinant lysostaphin (rLST), which is a native bacteriocin destroying the staphylococcal cell wall through its endopeptidase activity. We combined the use of E. coli BL21(DE3)/pET15b, factorial design, and simple Ni-NTA affinity chromatography to optimize rLST production. The enzyme yield was up to 50 mg/L culture, surpassing reported systems. Our rLST demonstrated superlative biofilm combating ability by inhibiting staphylococcal biofilms formation and detachment of already formed biofilms, compared to vancomycin and linezolid. Furthermore, we aimed at developing a novel rLST topical formula targeting staphylococcal skin infections. The phase inversion composition (PIC) method fulfilled this aim with its simple preparatory steps and affordable components. LST nano-emulgel (LNEG) was able to extend active LST release up to 8 h and cure skin infections in a murine skin model. We are introducing a rapid, convenient rLST production platform with an outcome of pure, active rLST incorporated into an effective LNEG formula with scaling-up potential to satisfy the needs of both research and therapeutic purposes.


Assuntos
Antibacterianos , Biofilmes/efeitos dos fármacos , Lisostafina , Staphylococcus aureus Resistente à Meticilina/fisiologia , Antibacterianos/química , Antibacterianos/farmacologia , Emulsões , Lisostafina/química , Lisostafina/farmacologia
9.
Nat Chem Biol ; 16(1): 24-30, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31686030

RESUMO

Lysostaphin is a bacteriolytic enzyme targeting peptidoglycan, the essential component of the bacterial cell envelope. It displays a very potent and specific activity toward staphylococci, including methicillin-resistant Staphylococcus aureus. Lysostaphin causes rapid cell lysis and disrupts biofilms, and is therefore a therapeutic agent of choice to eradicate staphylococcal infections. The C-terminal SH3b domain of lysostaphin recognizes peptidoglycans containing a pentaglycine crossbridge and has been proposed to drive the preferential digestion of staphylococcal cell walls. Here we elucidate the molecular mechanism underpinning recognition of staphylococcal peptidoglycan by the lysostaphin SH3b domain. We show that the pentaglycine crossbridge and the peptide stem are recognized by two independent binding sites located on opposite sides of the SH3b domain, thereby inducing a clustering of SH3b domains. We propose that this unusual binding mechanism allows synergistic and structurally dynamic recognition of S. aureus peptidoglycan and underpins the potent bacteriolytic activity of this enzyme.


Assuntos
Lisostafina/química , Peptidoglicano/química , Staphylococcus aureus/química , Bacteriólise/efeitos dos fármacos , Biofilmes , Parede Celular/química , Cromatografia Líquida de Alta Pressão , Análise Mutacional de DNA , Glicina/química , Ligantes , Espectroscopia de Ressonância Magnética , Mutagênese Sítio-Dirigida , Peptídeos/química , Ligação Proteica , Domínios Proteicos , Proteínas Recombinantes/química , Domínios de Homologia de src
10.
Biomacromolecules ; 20(10): 4035-4043, 2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31524374

RESUMO

Lytic enzymes have been considered as potential alternatives to antibiotics. These enzymes, particularly those that target Gram-positive bacteria, consist of modular cell wall-binding and catalytic domains, which can be shuffled with those of other lytic enzymes to produce unnatural chimeric enzymes. In this work, we report the in vitro shuffling of two different modular domains using a protein self-assembly methodology. Catalytic domains (CD) and cell wall-binding domains (BD) from the bacteriocin lysostaphin (Lst) and a putative autolysin from Staphylococcus aureus (SA1), respectively, were genetically site-specifically biotinylated and assembled with streptavidin to generate 23 permuted chimeras. The specific assembly of a CD (3 equiv) and a BD (1 equiv) from Lst and SA1, respectively [CDL-BDS (3:1)], on a streptavidin scaffold yielded high lytic activity against S. aureus (at least 5.6 log reduction), which was higher than that obtained with either native Lst or SA1 alone. Moreover, at 37 °C, the initial rate of cell lysis was over 3-fold higher than that with free Lst, thereby revealing the unique catalytic properties of the chimeric proteins. In vitro self-assembly of functional domains from modular lytic enzymes on a protein scaffold likely expands the repertoire of bactericidal enzymes with improved activities.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Staphylococcus aureus/efeitos dos fármacos , Domínio Catalítico/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Quimera , Lisostafina/química , Lisostafina/farmacocinética , N-Acetil-Muramil-L-Alanina Amidase/química , N-Acetil-Muramil-L-Alanina Amidase/farmacologia
11.
Anal Bioanal Chem ; 411(27): 7177-7185, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31522243

RESUMO

Accurate and rapid identification of Staphylococcus aureus (S. aureus) is of great significance for controlling the food poisoning and infectious diseases caused by S. aureus. In this study, a novel strategy that combines lysin cell-binding domain (CBD)-based magnetic separation with fluorescence detection was developed for the specific and sensitive quantification of S. aureus in authentic samples. The S. aureus cells were separated from the sample matrix by lysin CBD-functionalized magnetic beads. Following lysis by lysostaphin, intracellular catalase was released from S. aureus cells and detected by a fluorometric system composed of horseradish peroxidase (HRP), hydrogen peroxide (H2O2), and Amplex Red. S. aureus was quantified via the inhibitory effect of the released intracellular catalase on the fluorometric system since the catalase could decompose the H2O2. Optimized conditions afforded a calibration curve for S. aureus ranging from 1.0 × 102 to 1.0 × 107 CFU mL-1. The detection limit was as low as 78 CFU mL-1 in phosphate-buffered saline (PBS), and the total detection process could be completed in less than 50 min. Other bacteria associated with common food-borne and nosocomial infections negligibly interfered with S. aureus detection, except for Staphylococcus epidermidis, which may have slightly interfered. Moreover, the potential of this proposed method for practical applications has been demonstrated by detection assays of sterilized milk and human serum. Graphical abstract.


Assuntos
Catalase/metabolismo , Peróxido de Hidrogênio/química , Separação Imunomagnética/instrumentação , Lisostafina/química , Oxazinas/química , Staphylococcus aureus/isolamento & purificação , Animais , Bacteriemia/microbiologia , Sítios de Ligação , Fluorescência , Humanos , Leite/microbiologia , Domínios Proteicos
12.
Molecules ; 24(16)2019 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-31395814

RESUMO

Antibacterial lysins are promising proteins that are active against both antibiotic-susceptible and antibiotic-resistant bacterial strains. However, a major limitation of antibacterial lysins is their fast elimination from systemic circulation. PEGylation increases the plasma half-life of lysins but renders them inactive. Here we report the construction of a fusion protein of lysostaphin, a potent anti-staphylococcal lysin, and an albumin-binding domain from streptococcal protein G. The resulting fusion protein was less active than the parent enzyme lysostaphin, but it still retained significant antibacterial activity even when bound to serum albumin. The terminal half-life of the fusion protein in rats was five-fold greater than that of lysostaphin (7.4 vs. 1.5 h), and the area under the curve increased more than 115 times. Most importantly, this increase in systemic circulation time compensated for the decrease in activity. The plasma from rats that received an injection of the fusion protein retained bactericidal activity for up to 7 h, while plasma from rats that received plain lysostaphin lacked any detectable activity after 4 h. To the best of our knowledge, this is the first report of an antibacterial lysin with both improved pharmacokinetic parameters and prolonged bactericidal activity in the systemic circulation.


Assuntos
Proteínas de Bactérias , Lisostafina , Proteínas Recombinantes de Fusão , Albumina Sérica/química , Staphylococcus aureus/crescimento & desenvolvimento , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/farmacocinética , Proteínas de Bactérias/farmacologia , Feminino , Lisostafina/química , Lisostafina/genética , Lisostafina/farmacocinética , Lisostafina/farmacologia , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia
13.
Molecules ; 24(10)2019 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-31100806

RESUMO

The increasing prevalence of antibiotic-resistant strains of pathogenic bacteria is a major healthcare problem. Antibacterial lysins are enzymes that cleave the peptidoglycan of the bacterial cell wall. These proteins hold potential as a supplement or an alternative to traditional antibiotics since they are active against antibiotic resistant strains. However, antibacterial lysins are rapidly eliminated from the systemic circulation, which limits their application. Dimerization of an anti-pneumococcal lysin Cpl-1 has been demonstrated to decrease the clearance rate of this protein in mice. In the present work, we constructed a dimer of an anti-staphylococcal lysin lysostaphin by fusing it with an anti-parallel α-helical dimerization domain. Lysostaphin dimer had a more favorable pharmacokinetic profile with increased terminal half-life and area under the curve (AUC) values compared to monomeric lysostaphin. However, the staphylolytic activity of dimerized lysostaphin was decreased. This decrease in activity was likely caused by the dimerization; since the catalytic efficacy of lysostaphin dimer towards pentaglycine peptide was unaltered. Our results demonstrate that, although dimerization is indeed beneficial for the pharmacokinetics of antibacterial lysins, this approach might not be suitable for all lysins, as it can negatively affect the lysin activity.


Assuntos
Antibacterianos/química , Antibacterianos/farmacocinética , Lisostafina/química , Lisostafina/farmacocinética , Multimerização Proteica , Sequência de Aminoácidos , Área Sob a Curva , Catálise , Ativação Enzimática , Lisostafina/metabolismo , Testes de Sensibilidade Microbiana , Modelos Moleculares , Conformação Proteica , Staphylococcus/efeitos dos fármacos
14.
Sci Adv ; 5(5): eaaw1228, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31114804

RESUMO

Staphylococcus aureus is the most common pathogen associated with bacterial infections in orthopedic procedures. Infections often lead to implant failure and subsequent removal, motivating the development of bifunctional materials that both promote repair and prevent failure due to infection. Lysostaphin is an anti-staphylococcal enzyme resulting in bacterial lysis and biofilm reduction. Lysostaphin use is limited by the lack of effective delivery methods to provide sustained, high doses of enzyme to infection sites. We engineered a BMP-2-loaded lysostaphin-delivering hydrogel that simultaneously prevents S. aureus infection and repairs nonhealing segmental bone defects in the murine radius. Lysostaphin-delivering hydrogels eradicated S. aureus infection and resulted in mechanically competent bone. Cytokine and immune cell profiling demonstrated that lysostaphin-delivering hydrogels restored the local inflammatory environment to that of a sterile injury. These results show that BMP-2-loaded lysostaphin-delivering hydrogel therapy effectively eliminates S. aureus infection while simultaneously regenerating functional bone resulting in defect healing.


Assuntos
Antibacterianos/uso terapêutico , Proteína Morfogenética Óssea 2/uso terapêutico , Regeneração Óssea/efeitos dos fármacos , Lisostafina/uso terapêutico , Procedimentos Ortopédicos/efeitos adversos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/etiologia , Staphylococcus aureus/efeitos dos fármacos , Fator de Crescimento Transformador beta/uso terapêutico , Animais , Antibacterianos/química , Proteína Morfogenética Óssea 2/química , Sistemas de Liberação de Medicamentos , Hidrogéis/química , Inflamação/imunologia , Inflamação/microbiologia , Lisostafina/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Próteses e Implantes , Proteínas Recombinantes/química , Proteínas Recombinantes/uso terapêutico , Infecções Estafilocócicas/microbiologia , Fator de Crescimento Transformador beta/química
15.
Sci Rep ; 9(1): 5965, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979923

RESUMO

Staphylococcus simulans lysostaphin cleaves pentaglycine cross-bridges between stem peptides in the peptidoglycan of susceptible staphylococci, including S. aureus. This enzyme consists of an N-terminal catalytic domain and a cell wall binding domain (SH3b), which anchors the protein to peptidoglycan. Although structures of SH3bs from lysostaphin are available, the binding modes of peptidoglycan to these domains are still unclear. We have solved the crystal structure of the lysostaphin SH3b domain in complex with a pentaglycine peptide representing the peptidoglycan cross-bridge. The structure identifies a groove between ß1 and ß2 strands as the pentaglycine binding site. The structure suggests that pentaglycine specificity of the SH3b arises partially directly by steric exclusion of Cß atoms in the ligand and partially indirectly due to the selection of main chain conformations that are easily accessible for glycine, but not other amino acid residues. We have revealed further interactions of SH3b with the stem peptides with the support of bioinformatics tools. Based on the structural data we have attempted engineering of the domain specificity and have investigated the relevance of the introduced substitutions on the domain binding and specificity, also in the contexts of the mature lysostaphin and of its bacteriolytic activity.


Assuntos
Lisostafina/química , Peptidoglicano/química , Sequência de Aminoácidos , Biologia Computacional , Simulação por Computador , Escherichia coli , Lisostafina/genética , Lisostafina/metabolismo , Modelos Moleculares , Peptidoglicano/metabolismo , Ligação Proteica , Conformação Proteica , Domínios Proteicos , Engenharia de Proteínas , Staphylococcus
16.
Int J Biol Macromol ; 110: 157-166, 2018 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-29410001

RESUMO

Lysostaphin (LST) is a bacteriocin that cleaves within the pentaglycine cross bridge of Staphylococcus aureus peptidoglycan. Previous studies have reported the high efficiency of LST even against multi drug resistant S. aureus including methicillin resistant S. aureus (MRSA). In this study, we have developed a new chitosan based hydrogel formulation of LST to exploit its anti-staphylococcal activity. The atomic interactions of LST with chitosan were studied by molecular docking studies. The rheology and the antibacterial properties of the developed LSTC gel were evaluated. The developed LST containing chitosan hydrogel (LSTC gel) was flexible, flows smoothly and remains stable at physiological temperature. The in vitro studies by agar well diffusion and ex vivo studies in porcine skin model exhibited a reduction in S. aureus survival by ∼3 Log10CFU/mL in the presence of LSTC gel. The cytocompatibility of the gel was tested in vitro using macrophage RAW 264.7 cell line and in vivo in Drosophila melanogaster. A gradual disruption of S. aureus biofilms with the increase of LST concentrations in the LSTC gel was observed which was confirmed by SEM analysis. We conclude that LSTC gel could be highly effectual and advantageous over antibiotics in treating staphylococcal-topical and biofilm infections.


Assuntos
Biofilmes/efeitos dos fármacos , Quitosana , Hidrogéis , Lisostafina , Staphylococcus aureus Resistente à Meticilina/fisiologia , Infecções Estafilocócicas/tratamento farmacológico , Animais , Quitosana/química , Quitosana/farmacologia , Drosophila melanogaster , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Lisostafina/química , Lisostafina/farmacologia , Camundongos , Simulação de Acoplamento Molecular , Células RAW 264.7 , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/patologia , Suínos
17.
Sci Rep ; 7(1): 6020, 2017 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-28729697

RESUMO

We introduce LytU, a short member of the lysostaphin family of zinc-dependent pentaglycine endopeptidases. It is a potential antimicrobial agent for S. aureus infections and its gene transcription is highly upregulated upon antibiotic treatments along with other genes involved in cell wall synthesis. We found this enzyme to be responsible for the opening of the cell wall peptidoglycan layer during cell divisions in S. aureus. LytU is anchored in the plasma membrane with the active part residing in the periplasmic space. It has a unique Ile/Lys insertion at position 151 that resides in the catalytic site-neighbouring loop and is vital for the enzymatic activity but not affecting the overall structure common to the lysostaphin family. Purified LytU lyses S. aureus cells and cleaves pentaglycine, a reaction conveniently monitored by NMR spectroscopy. Substituting the cofactor zinc ion with a copper or cobalt ion remarkably increases the rate of pentaglycine cleavage. NMR and isothermal titration calorimetry further reveal that, uniquely for its family, LytU is able to bind a second zinc ion which is coordinated by catalytic histidines and is therefore inhibitory. The pH-dependence and high affinity of binding carry further physiological implications.


Assuntos
Endopeptidases/química , Lisostafina/química , Sequência de Aminoácidos , Antibacterianos/química , Sítios de Ligação , Domínio Catalítico , Membrana Celular/química , Membrana Celular/metabolismo , Endopeptidases/genética , Endopeptidases/metabolismo , Concentração de Íons de Hidrogênio , Lisostafina/metabolismo , Mutação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteólise , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Staphylococcus aureus/ultraestrutura , Relação Estrutura-Atividade , Zinco/metabolismo
18.
Biomol NMR Assign ; 11(1): 69-73, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27943001

RESUMO

Lysostaphin family endopeptidases, produced by Staphylococcus genus, are zinc-dependent enzymes that cleave pentaglycine bridges of cell wall peptidoglycan. They act as autolysins to maintain cell wall metabolism or as toxins and weapons against competing strains. Consequently, these enzymes are compelling targets for new drugs as well as are potential antimicrobial agents themselves against Staphylococcus pathogens, which depend on cell wall to retain their immunity against antibiotics. The rapid spread of methicillin and vancomycin-resistant Staphylococcus aureus strains draws demand for new therapeutic approaches. S. aureus gene sa0205 was found to be implicated in resistance to vancomycin and synthesis of the bacteria cell wall. The gene encodes for a catalytic domain of a lysostaphin-type endopeptidase. We aim to obtain the structure of the Sa0205 catalytic domain, the first solution structure of the catalytic domain of the lysostaphin family enzymes. In addition, we are to investigate the apparent binding of the second zinc ion, which has not been previously reported for the enzyme group. Herein, we present the backbone and side chain resonance assignments of Sa0205 endopeptidase catalytic domain in its one and two zinc-bound forms.


Assuntos
Domínio Catalítico , Lisostafina/química , Ressonância Magnética Nuclear Biomolecular , Staphylococcus aureus/enzimologia , Sequência de Aminoácidos , Lisostafina/metabolismo
19.
J Control Release ; 245: 108-115, 2017 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-27908758

RESUMO

Staphylococcus aureus infections of the skin and soft tissue pose a major concern to public health, largely owing to the steadily increasing prevalence of drug resistant isolates. As an alternative mode of treatment both bacteriophage endolysins and bacteriocins have been shown to possess antimicrobial efficacy against multiple species of bacteria including otherwise drug resistant strains. Despite this, the administration and exposure of such antimicrobials should be restricted until required in order to discourage the continued evolution of bacterial resistance, whilst maintaining the activity and stability of such proteinaceous structures. Utilising the increase in skin temperature during infection, the truncated bacteriophage endolysin CHAPK and the staphylococcal bacteriocin lysostaphin have been co-administered in a thermally triggered manner from Poly(N-isopropylacrylamide) (PNIPAM) nanoparticles. The thermoresponsive nature of the PNIPAM polymer has been employed in order to achieve the controlled expulsion of a synergistic enzybiotic cocktail consisting of CHAPK and lysostaphin. The point at which this occurs is modifiable, in this case corresponding to the threshold temperature associated with an infected wound. Consequently, bacterial lysis was observed at 37°C, whilst growth was maintained at the uninfected skin temperature of 32°C.


Assuntos
Antibacterianos/administração & dosagem , Bacteriocinas/administração & dosagem , Endopeptidases/administração & dosagem , Lisostafina/administração & dosagem , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Nanopartículas/administração & dosagem , Resinas Acrílicas/administração & dosagem , Resinas Acrílicas/química , Antibacterianos/química , Bacteriocinas/química , Bacteriófagos , Endopeptidases/química , Temperatura Alta , Lisostafina/química , Staphylococcus aureus Resistente à Meticilina/ultraestrutura , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Nanopartículas/química , Nanopartículas/ultraestrutura
20.
Proteins ; 85(1): 177-181, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27699884

RESUMO

Zoocin A is a Zn-metallopeptidase secreted by Streptococcus zooepidemicus strain 4881. Its catalytic domain is responsible for cleaving the D-alanyl-L-alanine peptide bond in streptococcal peptidoglycan. The solution NMR structure of the Cys74 to Ala74 mutant of the recombinant catalytic domain (rCAT C74A) has been determined. With a previous structure determination for the recombinant target recognition domain (rTRD), this completes the 3D structure of zoocin A. While the structure of rCAT C74A resembles those of the catalytic domains of lysostaphin and LytM, the substrate binding groove is wider and no tyrosine residue was observed in the active site. Proteins 2016; 85:177-181. © 2016 Wiley Periodicals, Inc.


Assuntos
Alanina/química , Proteínas de Bactérias/química , Bacteriocinas/química , Cisteína/química , Mutação , Streptococcus equi/química , Alanina/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteriocinas/genética , Bacteriocinas/metabolismo , Domínio Catalítico , Clonagem Molecular , Cisteína/metabolismo , Endopeptidases/química , Endopeptidases/genética , Endopeptidases/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Cinética , Lisostafina/química , Lisostafina/metabolismo , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Streptococcus equi/enzimologia , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...