Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(2): 259-270.e11, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29937224

RESUMEN

Many community- and hospital-acquired bacterial infections are caused by antibiotic-resistant pathogens. Methicillin-resistant Staphylococcus aureus (MRSA) predisposes humans to invasive infections that are difficult to eradicate. We designed a closed-loop gene network programming mammalian cells to autonomously detect and eliminate bacterial infections. The genetic circuit contains human Toll-like receptors as the bacterial sensor and a synthetic promoter driving reversible and adjustable expression of lysostaphin, a bacteriolytic enzyme highly lethal to S. aureus. Immunomimetic designer cells harboring this genetic circuit exhibited fast and robust sense-and-destroy kinetics against live staphylococci. When tested in a foreign-body infection model in mice, microencapsulated cell implants prevented planktonic MRSA infection and reduced MRSA biofilm formation by 91%. Notably, this system achieved a 100% cure rate of acute MRSA infections, whereas conventional vancomycin treatment failed. These results suggest that immunomimetic designer cells could offer a therapeutic approach for early detection, prevention, and cure of pathogenic infections in the post-antibiotic era.


Asunto(s)
Biomimética/métodos , Staphylococcus aureus Resistente a Meticilina/fisiología , Infecciones Estafilocócicas/prevención & control , Fosfatasa Alcalina/sangre , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Pruebas Antimicrobianas de Difusión por Disco , Femenino , Células HEK293 , Humanos , Receptores de Lipopolisacáridos/genética , Lisostafina/metabolismo , Lisostafina/farmacología , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Plásmidos/genética , Plásmidos/metabolismo , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/veterinaria , Receptor Toll-Like 1/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 6/genética , Factor de Transcripción AP-1/metabolismo
2.
Antimicrob Agents Chemother ; 68(9): e0023524, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39072634

RESUMEN

In response to the antimicrobial resistance crisis, we have developed a powerful and versatile therapeutic platform, the Antibacterial Drone (ABD) system. The ABD consists of a highly mobile staphylococcal pathogenicity island re-purposed to deliver genes encoding antibacterial proteins. The chromosomally located island is induced by a co-resident helper phage, packaged in phage-like particles, and released in very high numbers upon phage-induced lysis. ABD particles specifically adsorb to bacteria causing an infection and deliver their DNA to these bacteria, where the bactericidal cargo genes are expressed, kill the bacteria, and cure the infection. Here, we report a major advance of the system, incorporation of the gene encoding a secreted, bactericidal, species-specific lytic enzyme, lysostsphin. This ABD not only kills the bacterium that has been attacked by the ABD, but also any surrounding bacteria that are sensitive to the lytic enzyme which is released by secretion and by lysis of the doomed cell. So while the killing field is thus expanded, there are no civilian casualties (bacteria that are insensitive to the ABD and its cargo protein(s) are not inadvertently killed). Without amplifying the number of ABD particles (which are not re-packaged), the expression and release of the cargo gene's product dramatically extend the effective reach of the ABD. A cargo gene that encodes a secreted bactericidal protein also enables the treatment of a mixed bacterial infection in which one of the infecting organisms is insensitive to the ABD delivery system but is sensitive to the ABD's secreted cargo protein.


Asunto(s)
Antibacterianos , Lisostafina , Antibacterianos/farmacología , Lisostafina/farmacología , Islas Genómicas/genética , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Bacteriófagos/genética
3.
Molecules ; 29(17)2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39274911

RESUMEN

The cell wall is an indispensable element of bacterial cells and a long-known target of many antibiotics. Penicillin, the first discovered beta-lactam antibiotic inhibiting the synthesis of cell walls, was successfully used to cure many bacterial infections. Unfortunately, pathogens eventually developed resistance to it. This started an arms race, and while novel beta-lactams, either natural or (semi)synthetic, were discovered, soon upon their application, bacteria were developing resistance. Currently, we are facing the threat of losing the race since more and more multidrug-resistant (MDR) pathogens are emerging. Therefore, there is an urgent need for developing novel approaches to combat MDR bacteria. The cell wall is a reasonable candidate for a target as it differentiates not only bacterial and human cells but also has a specific composition unique to various groups of bacteria. This ensures the safety and specificity of novel antibacterial agents that target this structure. Due to the shortage of low-molecular-weight candidates for novel antibiotics, attention was focused on peptides and proteins that possess antibacterial activity. Here, we describe proteinaceous agents of various origins that target bacterial cell wall, including bacteriocins and phage and bacterial lysins, as alternatives to classic antibiotic candidates for antimicrobial drugs. Moreover, advancements in protein chemistry and engineering currently allow for the production of stable, specific, and effective drugs. Finally, we introduce the concept of selective targeting of dangerous pathogens, exemplified by staphylococci, by agents specifically disrupting their cell walls.


Asunto(s)
Antibacterianos , Pared Celular , Bacterias Grampositivas , Pared Celular/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Bacterias Grampositivas/efectos de los fármacos , Humanos , Bacteriocinas/farmacología , Bacteriocinas/química , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Infecciones por Bacterias Grampositivas/microbiología , Bacteriófagos
4.
Biochem Biophys Res Commun ; 668: 111-117, 2023 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-37245291

RESUMEN

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.


Asunto(s)
Lisostafina , Staphylococcus aureus Resistente a Meticilina , Lisostafina/química , Lisostafina/metabolismo , Lisostafina/farmacología , Staphylococcus aureus , Dominio Catalítico , Ligandos , Endopeptidasas/genética , Endopeptidasas/metabolismo , Catálisis
5.
BMC Biotechnol ; 23(1): 39, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37723466

RESUMEN

Staphylococcus aureus is a unique challenge for the healthcare system because it can form biofilms, is resistant to the host's immune system, and is resistant to numerous antimicrobial therapies. The aim of this study was to investigate the effect of poly (lactic-co-glycolic acid) (PLGA) polymer nanoparticles loaded with vancomycin and conjugated with lysostaphin (PLGA-VAN-LYS) on inhibiting S. aureus biofilm formation. Nano drug carriers were produced using the double emulsion evaporation process. we examined the physicochemical characteristics of the nanoparticles, including particle size, polydispersity index (PDI), zeta potential, drug loading (DL), entrapment efficiency (EE), Lysostaphin conjugation efficiency (LCE), and shape. The effect of the nano drug carriers on S. aureus strains was evaluated by determining the minimum inhibitory concentration (MIC), conducting biofilm formation inhibition studies, and performing agar well diffusion tests. The average size, PDI, zeta potential, DL, EE, and LCE of PLGA-VAN-LYS were 320.5 ± 35 nm, 0.270 ± 0.012, -19.5 ± 1.3 mV, 16.75 ± 2.5%, 94.62 ± 2.6%, and 37% respectively. Both the agar well diffusion and MIC tests did not show a distinction between vancomycin and the nano drug carriers after 72 h. However, the results of the biofilm analysis demonstrated that the nano drug carrier had a stronger inhibitory effect on biofilm formation compared to the free drug. The use of this technology for treating hospital infections caused by the Staphylococcus bacteria may have favorable effects on staphylococcal infections, considering the efficacy of the nano medicine carrier developed in this study.


Asunto(s)
Infecciones Estafilocócicas , Vancomicina , Humanos , Vancomicina/farmacología , Glicoles , Staphylococcus aureus , Agar , Lisostafina , Polímeros , Biopelículas
6.
Biotechnol Bioeng ; 120(6): 1694-1701, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36810983

RESUMEN

We describe a facile strategy to identify sites for the incorporation of noncanonical amino acids into lysostaphin-an enzyme that degrades the cell wall of Staphylococcus aureus-while retaining stapholytic activity. We used this strategy to generate active variants of lysostaphin incorporating para-azidophenylalanine. The incorporation of this "reactive handle" enabled the orthogonal site-specific modification of the enzyme variants with polyethylene glycol (PEG) using copper-free click cycloaddition. PEGylated lysostaphin variants could retain their stapholytic activity, with the extent of retention depending on the site of modification and the PEG molecular weight. The site-specific modification of lysostaphin could be useful not only for PEGylation to improve biocompatibility but also for the incorporation of the enzyme into hydrogels and other biomaterials and for studies of protein structure and dynamics. Moreover, the approach described herein could be readily applied to identify suitable sites for the incorporation of reactive handles into other proteins of interest.


Asunto(s)
Lisostafina , Infecciones Estafilocócicas , Humanos , Lisostafina/farmacología , Aminoácidos/química , Proteínas , Staphylococcus aureus/metabolismo
7.
Protein Expr Purif ; 207: 106274, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37084838

RESUMEN

Lysostaphin is a zinc-dependent endopeptidase that is effective against both antibiotic-sensitive and antibiotic-resistant strains of Staphylococcus aureus. Lysostaphin is typically purified on cation-exchange or metal-chelate affinity resins, and there are data indicating potential influence of the chromatographic resin on the lysostaphin activity. In this study, we systematically investigated the impact of the resin used to purify the recombinant lysostaphin on its activity. To this end, recombinant lysostaphin with an additional histidine tag at the C-terminus was purified using a cation-exchange resin, three types of nickel-chelate resins with different strength of metal ion binding, or a zinc-chelate resin. Lysostaphin samples purified on the cation-exchange resin (WorkBeads 40S), the nickel-chelate resin with a strong nickel ion binding (WorkBeads NiMAC), and the zinc-chelate resin (WorkBeads NTA with immobilized zinc ions) had equal activity. On the contrary, the activity of lysostaphin preparations purified on nickel-chelate resins with medium (WorkBeads Ni-NTA) and relatively weak (WorkBeads Ni-IDA) nickel ion binding was significantly reduced. The decrease in activity can be explained by the interaction of lysostaphin with the nickel ions leached from the resin and is caused by either the exchange of the zinc ion in the lysostaphin active center with a nickel ion from the resin, or binding of an additional ion that inhibits the enzymatic activity. Removal of the metal ions from the active site of lysostaphin and subsequent incorporation of the native zinc ions lead to complete restoration of the activity of the enzyme.


Asunto(s)
Lisostafina , Níquel , Níquel/química , Metales/química , Quelantes/química , Zinc/química , Cromatografía de Afinidad/métodos , Antibacterianos
8.
Appl Microbiol Biotechnol ; 106(13-16): 5023-5033, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35790549

RESUMEN

Mature lysostaphin (mLst) is a glycineglycine endopeptidase, capable of specifically cleaving penta-glycine crosslinker in the peptidoglycan of Staphylococcus aureus cell wall. It is a very effective therapeutic enzyme to kill the multidrug-resistant S. aureus often encountered in hospital acquired infections. Fusing cellulose binding domain (CBD) to mLst significantly reduced the insoluble expression of mLst in E. coli. Employing mLst-cleavable peptides as fusion linkers leaded to an effective self-cleavage expression that CBD and mLst could be completely cleaved off from the fusions during the expression process. The presence of residue linker fragment at N-terminus of the cleaved-off mLst strongly inhibited the cell lytic activity of the recovered recombinant mLst, and only ~ 50% of the wild-type mLst activity could be retained. Intact CBD-Lst fusions were obtained when uncleavable peptide linkers were employed. With CBD at N-terminus of mLst, the intact fusion completely lost its cell lytic activity but the dipeptidase activity still remained. In contrast, approximately 10% cell lytic activity of mLst still could be maintained for the fusion with CBD at C-terminus of mLst. KEY POINTS: • CBD fusion enhanced soluble expression of recombinant lysostaphin. • In vivo self-cleavage of fusion linkers by the expressed lysostaphin fusions. • Self-cleaved lysostaphin fusions retain most of dipeptidase but lose 50% cell lytic activity.


Asunto(s)
Dipeptidasas , Staphylococcus aureus Resistente a Meticilina , Celulosa , Escherichia coli/genética , Escherichia coli/metabolismo , Lisostafina/farmacología , Tipificación de Secuencias Multilocus , Peptidoglicano/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-33318001

RESUMEN

There is an urgent need for novel agents to treat drug-resistant bacterial infections, such as multidrug-resistant Staphylococcus aureus (MRSA). Desirable properties for new antibiotics include high potency, narrow species selectivity, low propensity to elicit new resistance phenotypes, and synergy with standard-of-care (SOC) chemotherapies. Here, we describe analysis of the antibacterial potential exhibited by F12, an innovative anti-MRSA lysin that has been genetically engineered to evade detrimental antidrug immune responses in human patients. F12 possesses high potency and rapid onset of action, it has narrow selectivity against pathogenic staphylococci, and it manifests synergy with numerous SOC antibiotics. Additionally, resistance to F12 and ß-lactam antibiotics appears mutually exclusive, and, importantly, we provide evidence that F12 resensitizes normally resistant MRSA strains to ß-lactams both in vitro and in vivo These results suggest that combinations of F12 and SOC antibiotics are a promising new approach to treating refractory S. aureus infections.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Sinergismo Farmacológico , Humanos , Lisostafina/farmacología , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus , beta-Lactamas/farmacología
10.
Artículo en Inglés | MEDLINE | ID: mdl-33468459

RESUMEN

Drug-resistant bacterial pathogens are a serious threat to global health, and antibacterial lysins are at the forefront of innovative treatments for these life-threatening infections. While lysins' general mechanism of action is well understood, the design principles that might enable engineering of performance-enhanced variants are still being formulated. Here, we report a detailed analysis of molecular determinants underlying the in vivo efficacy of lysostaphin, a canonical anti-MRSA (methicillin-resistant Staphylococcus aureus) lysin. Systematic analysis of bacterial binding, growth inhibition, lysis kinetics, and in vivo therapeutic efficacy revealed that binding affinity, and not inherent catalytic firepower, is the dominant driver of lysostaphin efficacy. This insight enabled electrostatic affinity tuning of lysostaphin to produce a single point mutant that manifested dramatically enhanced processivity and lysis kinetics and trended toward improved in vivo efficacy. More generally, these studies provide important insights into the complex relationships between lysin electrostatics, bacterial targeting, cell lysis efficiency, and in vivo efficacy. The lessons learned may enable engineering of other high-performance antibacterial biocatalysts.


Asunto(s)
Lisostafina , Staphylococcus aureus Resistente a Meticilina , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Cinética , Lisostafina/metabolismo , Lisostafina/farmacología , Staphylococcus aureus Resistente a Meticilina/metabolismo , Electricidad Estática
11.
Protein Expr Purif ; 177: 105753, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32950627

RESUMEN

Staphylococcus aureus (S. aureus), which has developed multidrug resistance, leads to many healthcare-associated infections resulting in significant medical and economic losses. Therefore, the development of new efficient strategies to deal with these bacteria has been gaining importance. Lysostaphin is a peptidoglycan hydrolase that has considerable potential as a bacteriocin. However, there have been few reported optimization and scale-up studies of the lysostaphin bioproduction process. Our preliminary results have revealed that the composition of auto-induction media at 30 °C increases the produced lysostaphin around 10-fold in shake flasks. In this study, achieving higher yields for recombinant lysostaphin in E. coli at a laboratory scale has been the aim, through the use of auto-induction media. Optimized medium composition and fermentation parameters were transferred to a laboratory-scale bioreactor. The tested conditions improved protein yields up to 184 mg/L in a 3 L stirred bioreactor and the productivity was improved 2-fold in comparison to previously published reports. Furthermore, this study also showed that lysostaphin is an effective bacteriocin on both commercially available and isolated S. aureus strains. These results will contribute to future larger-scale production of lysostaphin via the proposed fermentation conditions.


Asunto(s)
Antibacterianos/biosíntesis , Lisostafina/biosíntesis , Staphylococcus aureus/efectos de los fármacos , Staphylococcus/metabolismo , Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , Reactores Biológicos , Clonación Molecular , Medios de Cultivo/química , Medios de Cultivo/farmacología , Escherichia coli/efectos de los fármacos , Escherichia coli/genética , Escherichia coli/metabolismo , Fermentación , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Glicerol/metabolismo , Glicerol/farmacología , Lisostafina/genética , Lisostafina/aislamiento & purificación , Lisostafina/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Staphylococcus/genética , Staphylococcus aureus/crecimiento & desarrollo
12.
J Appl Microbiol ; 131(3): 1072-1082, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33382154

RESUMEN

Staphylococcus aureus, an opportunistic pathogen, causes diverse community and nosocomial-acquired human infections, including folliculitis, impetigo, sepsis, septic arthritis, endocarditis, osteomyelitis, implant-associated biofilm infections and contagious mastitis in cattle. In recent days, both methicillin-sensitive and methicillin-resistant S. aureus infections have increased. Highly effective anti-staphylococcal agents are urgently required. Lysostaphin is a 27 kDa zinc metallo antimicrobial lytic enzyme that is produced by Staphylococcus simulans biovar staphylolyticus and was first discovered in the 1960s. Lysostaphin is highly active against S. aureus strains irrespective of their drug-resistant patterns with a minimum inhibitory concentration of ranges between 0·001 and 0·064 µg ml-1 . Lysostaphin has activity against both dividing and non-dividing S. aureus cells; and can seep through the extracellular matrix to kill the biofilm embedded S. aureus. In spite of having excellent anti-staphylococcal activity, its clinical application is hindered because of its immunogenicity and reduced bio-availability. Extensive research with lysostaphin lead to the development of several engineered lysostaphin derivatives with reduced immunogenicity and increased serum half-life. Therapeutic efficacy of both native and engineered lysostaphin derivatives was studied by several research groups. This review provides an overview of the therapeutic applications of native and engineered lysostaphin derivatives developed to eradicate S. aureus infections.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Infecciones Estafilocócicas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Bovinos , Femenino , Lisostafina/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus , Staphylococcus aureus
13.
Biotechnol Appl Biochem ; 68(5): 1058-1066, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32918836

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Lisostafina/metabolismo , Nanopartículas/química , Infecciones Estafilocócicas/tratamiento farmacológico , Titanio/farmacología , Antibacterianos/química , Biopelículas/efectos de los fármacos , Biotina/química , Biotina/metabolismo , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Lisostafina/química , Lisostafina/genética , Tamaño de la Partícula , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Infecciones Estafilocócicas/metabolismo , Estreptavidina/química , Estreptavidina/metabolismo , Propiedades de Superficie , Titanio/química
14.
Proc Natl Acad Sci U S A ; 115(22): E4960-E4969, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29760099

RESUMEN

Orthopedic implant infections are a significant clinical problem, with current therapies limited to surgical debridement and systemic antibiotic regimens. Lysostaphin is a bacteriolytic enzyme with high antistaphylococcal activity. We engineered a lysostaphin-delivering injectable PEG hydrogel to treat Staphylococcus aureus infections in bone fractures. The injectable hydrogel formulation adheres to exposed tissue and fracture surfaces, ensuring efficient, local delivery of lysostaphin. Lysostaphin encapsulation within this synthetic hydrogel maintained enzyme stability and activity. Lysostaphin-delivering hydrogels exhibited enhanced antibiofilm activity compared with soluble lysostaphin. Lysostaphin-delivering hydrogels eradicated S. aureus infection and outperformed prophylactic antibiotic and soluble lysostaphin therapy in a murine model of femur fracture. Analysis of the local inflammatory response to infections treated with lysostaphin-delivering hydrogels revealed indistinguishable differences in cytokine secretion profiles compared with uninfected fractures, demonstrating clearance of bacteria and associated inflammation. Importantly, infected fractures treated with lysostaphin-delivering hydrogels fully healed by 5 wk with bone formation and mechanical properties equivalent to those of uninfected fractures, whereas fractures treated without the hydrogel carrier were equivalent to untreated infections. Finally, lysostaphin-delivering hydrogels eliminate methicillin-resistant S. aureus infections, supporting this therapy as an alternative to antibiotics. These results indicate that lysostaphin-delivering hydrogels effectively eliminate orthopedic S. aureus infections while simultaneously supporting fracture repair.


Asunto(s)
Antibacterianos/administración & dosificación , Curación de Fractura/efectos de los fármacos , Hidrogeles/uso terapéutico , Lisostafina/administración & dosificación , Infecciones Relacionadas con Prótesis , Infecciones Estafilocócicas , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Materiales Biocompatibles/uso terapéutico , Modelos Animales de Enfermedad , Fracturas del Fémur/cirugía , Lisostafina/farmacología , Lisostafina/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Diseño de Prótesis , Infecciones Relacionadas con Prótesis/tratamiento farmacológico , Infecciones Relacionadas con Prótesis/prevención & control , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus
15.
Int J Mol Sci ; 21(23)2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33266291

RESUMEN

Gaining an insight into the mechanism underlying antimicrobial-resistance development in Staphylococcus aureus is crucial for identifying effective antimicrobials. We isolated S. aureus sequence type 72 from a patient in whom the S. aureus infection was highly resistant to various antibiotics and lysostaphin, but no known resistance mechanisms could explain the mechanism of lysostaphin resistance. Genome-sequencing followed by subtractive and functional genomics revealed that serine hydroxymethyltransferase (glyA or shmT gene) plays a key role in lysostaphin resistance. Serine hydroxymethyltransferase (SHMT) is indispensable for the one-carbon metabolism of serine/glycine interconversion and is linked to folate metabolism. Functional studies revealed the involvement of SHMT in lysostaphin resistance, as ΔshmT was susceptible to the lysostaphin, while complementation of the knockout expressing shmT restored resistance against lysostaphin. In addition, the ΔshmT showed reduced virulence under in vitro (mammalian cell lines infection) and in vivo (wax-worm infection) models. The SHMT inhibitor, serine hydroxymethyltransferase inhibitor 1 (SHIN1), protected the 50% of the wax-worm infected with wild type S. aureus. These results suggest SHMT is relevant to the extreme susceptibility to lysostaphin and the host immune system. Thus, the current study established that SHMT plays a key role in lysostaphin resistance development and in determining the virulence potential of multiple drug-resistant S. aureus.


Asunto(s)
Antiinfecciosos Locales/farmacología , Farmacorresistencia Bacteriana , Glicina Hidroximetiltransferasa/genética , Lisostafina/farmacología , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Animales , Genoma Bacteriano , Genómica/métodos , Redes y Vías Metabólicas , Fenotipo , Staphylococcus aureus/ultraestructura , Virulencia/genética , Factores de Virulencia/genética
16.
Infect Immun ; 87(5)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30782857

RESUMEN

Precise enumeration of living intracellular bacteria is the key step to estimate the invasion potential of pathogens and host immune responses to understand the mechanism and kinetics of bacterial pathogenesis. Therefore, quantitative assessment of host-pathogen interactions is essential for development of novel antibacterial therapeutics for infectious disease. The gentamicin protection assay (GPA) is the most widely used method for these estimations by counting the CFU of intracellular living pathogens. Here, we assess the longstanding drawbacks of the GPA by employing an antistaphylococcal endopeptidase as a bactericidal agent to kill extracellular Staphylococcus aureus We found that the difference between the two methods for the recovery of intracellular CFU of S. aureus was about 5 times. We prove that the accurate number of intracellular CFU could not be precisely determined by the GPA due to the internalization of gentamicin into host cells during extracellular bacterial killing. We further demonstrate that lysostaphin-mediated extracellular bacterial clearance has advantages for measuring the kinetics of bacterial internalization on a minute time scale due to the fast and tunable activity and the inability of protein to permeate the host cell membrane. From these results, we propose that accurate quantification of intracellular bacteria and measurement of internalization kinetics can be achieved by employing enzyme-mediated killing of extracellular bacteria (enzyme protection assay [EPA]) rather than the host-permeative drug gentamicin, which is known to alter host physiology.


Asunto(s)
Carga Bacteriana , Bioensayo/métodos , Pruebas de Enzimas/métodos , Gentamicinas/análisis , Interacciones Huésped-Patógeno , Infecciones Estafilocócicas/fisiopatología , Staphylococcus aureus/aislamiento & purificación
17.
Biotechnol Bioeng ; 116(12): 3149-3159, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31433061

RESUMEN

Bacteriolytic enzymes (cell lytic enzymes) are promising alternatives to antibiotics especially in killing drug-resistant bacteria. However, some bacteria slowly become resistant to various classes of peptidoglycan hydrolases, for reasons not well studied, in the presence of growth-supporting nutrients, which are prevalent at sites of infection. Here, we show that Staphylococcus aureus, a human and animal pathogen, while susceptible to the potent staphylolytic enzyme lysostaphin (Lst) in buffered saline, is highly resistant in the rich medium tryptic soy broth (TSB). Through a series of biochemical analysis, we identified that the resistance was due to prevention of Lst-cell binding mediated by the wall teichoic acids (WTAs) present on the cell surface. Inhibition or deletion of the gene tarO responsible for the first step of WTA biosynthesis greatly reduced S. aureus resistance to Lst in TSB. To overcome the resistance, we took advantage of the gene regulation potential of CRISPR-dCas9 and demonstrated that downregulation of tarO, tarH, and/or tarG gene expression, the latter two encoding enzymes that anchor WTAs in the outer layer of cell wall peptidoglycan, sensitized S. aureus to Lst and enabled eradication of the bacterium in TSB in 24 hr. As a result, we elucidate a key mechanism of Lst resistance in metabolically active S. aureus and provide a potential approach for treating life-threatening or hard-to-treat infections caused by Gram-positive pathogens.


Asunto(s)
Sistemas CRISPR-Cas , Farmacorresistencia Bacteriana , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica , Lisostafina/farmacología , Staphylococcus aureus , Animales , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Bacteriana de la Expresión Génica/genética , Genes Bacterianos , Humanos , Staphylococcus aureus/genética , Staphylococcus aureus/crecimiento & desarrollo
18.
Appl Microbiol Biotechnol ; 103(17): 7041-7054, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31203418

RESUMEN

The role that the complex microbial communities play in human and environmental health cannot be understated. The increased information about community complexity, as well as the overuse of broad-spectrum antibiotics, suggest that new approaches to target specific organisms within a community context are essential towards new antimicrobial therapies. Here, we have assessed the activity and selectivity of two cell wall lytic enzymes, lysostaphin (Lst) and PlyPH, in the presence of multiple bacteria and under varied media conditions. Lst and PlyPH target the clinically relevant pathogens Staphylococcus aureus and Bacillus cereus, respectively. Lst was effective under all conditions resulting in ~ 4-log and ~ 3-log reduction at 100 µg/mL in actively growing monoculture and co-culture, respectively. PlyPH was also selective but less active and more susceptible to media and cell population changes. Lst and PlyPH activities could be increased in supernatants from actively growing cultures in the presence of a protease inhibitor cocktail, suggesting a possible role played by proteases secreted during cell growth in reducing lytic enzyme activity. This work demonstrates the utility of cell wall lytic enzymes for targeted pathogen killing or microbial community remodeling.


Asunto(s)
Antiinfecciosos/farmacología , Pared Celular/efectos de los fármacos , Consorcios Microbianos/efectos de los fármacos , Bacillus cereus/efectos de los fármacos , Bacillus cereus/crecimiento & desarrollo , Bacillus cereus/metabolismo , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Técnicas de Cocultivo , Medios de Cultivo/química , Lisostafina/farmacología , Péptido Hidrolasas/análisis , Péptido Hidrolasas/metabolismo , Inhibidores de Proteasas/farmacología , Especificidad de la Especie , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo
19.
Molecules ; 24(16)2019 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-31395814

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Lisostafina , Proteínas Recombinantes de Fusión , Albúmina Sérica/química , Staphylococcus aureus/crecimiento & desarrollo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/farmacocinética , Proteínas Bacterianas/farmacología , Femenino , Lisostafina/química , Lisostafina/genética , Lisostafina/farmacocinética , Lisostafina/farmacología , Ratas , Ratas Wistar , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/farmacología
20.
Molecules ; 24(10)2019 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-31100806

RESUMEN

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.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacocinética , Lisostafina/química , Lisostafina/farmacocinética , Multimerización de Proteína , Secuencia de Aminoácidos , Área Bajo la Curva , Catálisis , Activación Enzimática , Lisostafina/metabolismo , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación Proteica , Staphylococcus/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA