Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 67(5): e0035523, 2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-37125913

RESUMO

The treatment of bacterial infections is becoming increasingly challenging with the emergence of antimicrobial resistance. Thus, the development of antimicrobials with novel mechanisms of action is much needed. Previously, we designed several cationic main-chain imidazolium compounds and identified the polyimidazolium PIM1 as a potent antibacterial against a wide panel of multidrug-resistant nosocomial pathogens, and it had relatively low toxicity against mammalian epithelial cells. However, little is known about the mechanism of action of PIM1. Using an oligomeric version of PIM1 with precisely six repeating units (OIM1-6) to control for consistency, we showed that OIM1-6 relies on an intact membrane potential for entry into the bacterial cytoplasm, as resistant mutants to OIM1-6 have mutations in their electron transport chains. These mutants demonstrate reduced uptake of the compound, which can be circumvented through the addition of a sub-MIC dose of colistin. Once taken up intracellularly, OIM1-6 exerts double-stranded DNA breaks. Its potency and ability to kill represents a promising class of drugs that can be combined with membrane-penetrating drugs to potentiate activity and hedge against the rise of resistant mutants. In summary, we discovered that cationic antimicrobial OIM1-6 exhibits an antimicrobial property that is dissimilar to the conventional cationic antimicrobial compounds. Its killing mechanism does not involve membrane disruption but instead depends on the membrane potential for uptake into bacterial cells so that it can exert its antibacterial effect intracellularly.


Assuntos
Anti-Infecciosos , Peptídeos Catiônicos Antimicrobianos , Animais , DNA Bacteriano , Potenciais da Membrana , Peptídeos Catiônicos Antimicrobianos/farmacologia , Antibacterianos/farmacologia , Bactérias , Testes de Sensibilidade Microbiana , Mamíferos
2.
J Antimicrob Chemother ; 78(10): 2581-2590, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37671807

RESUMO

OBJECTIVES: The rise of MDR Gram-negative bacteria (GNB), especially those resistant to last-resort drugs such as carbapenems and colistin, is a global health risk and calls for increased efforts to discover new antimicrobial compounds. We previously reported that polyimidazolium (PIM) compounds exhibited significant antimicrobial activity and minimal mammalian cytotoxicity. However, their mechanism of action is relatively unknown. We examined the efficacy and mechanism of action of a hydrophilic PIM (PIM5) against colistin- and meropenem-resistant clinical isolates. METHODS: MIC and time-kill testing was performed for drug-resistant Escherichia coli and Klebsiella pneumoniae clinical isolates. N-phenyl-1-naphthylamine and propidium iodide dyes were employed to determine membrane permeabilization. Spontaneous resistant mutants and single deletion mutants were generated to understand potential resistance mechanisms to the drug. RESULTS: PIM5 had the same effectiveness against colistin- and meropenem-resistant strains as susceptible strains of GNB. PIM5 exhibited a rapid bactericidal effect independent of bacterial growth phase and was especially effective in water. The polymer disrupts both the outer and cytoplasmic membranes. PIM5 binds and intercalates into bacterial genomic DNA upon entry of cells. GNB do not develop high resistance to PIM5. However, the susceptibility and uptake of the polymer is moderately affected by mutations in the two-component histidine kinase sensor BaeS. PIM5 has negligible cytotoxicity on human cells at bacterial-killing concentrations, comparable to the commercial antibiotics polymyxin B and colistin. CONCLUSIONS: PIM5 is a potent broad-spectrum antibiotic targeting GNB resistant to last-resort antibiotics.


Assuntos
Antibacterianos , Anti-Infecciosos , Animais , Humanos , Antibacterianos/farmacologia , Colistina/farmacologia , Meropeném/farmacologia , Bactérias Gram-Negativas , Anti-Infecciosos/farmacologia , Escherichia coli/genética , Testes de Sensibilidade Microbiana , Farmacorresistência Bacteriana Múltipla , Mamíferos
3.
Proc Natl Acad Sci U S A ; 117(49): 31376-31385, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229526

RESUMO

For a myriad of different reasons most antimicrobial peptides (AMPs) have failed to reach clinical application. Different AMPs have different shortcomings including but not limited to toxicity issues, potency, limited spectrum of activity, or reduced activity in situ. We synthesized several cationic peptide mimics, main-chain cationic polyimidazoliums (PIMs), and discovered that, although select PIMs show little acute mammalian cell toxicity, they are potent broad-spectrum antibiotics with activity against even pan-antibiotic-resistant gram-positive and gram-negative bacteria, and mycobacteria. We selected PIM1, a particularly potent PIM, for mechanistic studies. Our experiments indicate PIM1 binds bacterial cell membranes by hydrophobic and electrostatic interactions, enters cells, and ultimately kills bacteria. Unlike cationic AMPs, such as colistin (CST), PIM1 does not permeabilize cell membranes. We show that a membrane electric potential is required for PIM1 activity. In laboratory evolution experiments with the gram-positive Staphylococcus aureus we obtained PIM1-resistant isolates most of which had menaquinone mutations, and we found that a site-directed menaquinone mutation also conferred PIM1 resistance. In similar experiments with the gram-negative pathogen Pseudomonas aeruginosa, PIM1-resistant mutants did not emerge. Although PIM1 was efficacious as a topical agent, intraperitoneal administration of PIM1 in mice showed some toxicity. We synthesized a PIM1 derivative, PIM1D, which is less hydrophobic than PIM1. PIM1D did not show evidence of toxicity but retained antibacterial activity and showed efficacy in murine sepsis infections. Our evidence indicates the PIMs have potential as candidates for development of new drugs for treatment of pan-resistant bacterial infections.


Assuntos
Antibacterianos/farmacologia , Drogas Desenhadas/farmacologia , Imidazóis/farmacologia , Animais , Antibacterianos/química , Antibacterianos/uso terapêutico , Morte Celular/efeitos dos fármacos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Drogas Desenhadas/química , Drogas Desenhadas/uso terapêutico , Humanos , Interações Hidrofóbicas e Hidrofílicas , Imidazóis/química , Imidazóis/uso terapêutico , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Testes de Sensibilidade Microbiana , Viabilidade Microbiana/efeitos dos fármacos , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/efeitos dos fármacos , Sepse/tratamento farmacológico , Sepse/prevenção & controle , Pele/efeitos dos fármacos , Pele/microbiologia , Pele/patologia
4.
Antimicrob Agents Chemother ; 66(10): e0059722, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36094258

RESUMO

Frequent outbreaks of Salmonella Typhimurium infection, in both animal and human populations and with the potential for zoonotic transmission, pose a significant threat to the public health sector. The rapid emergence and spread of more invasive multidrug-resistant clinical isolates of Salmonella further highlight the need for the development of new drugs with effective broad-spectrum bactericidal activities. The synthesis and evaluation of main-chain cationic polyimidazolium 1 (PIM1) against several Gram-positive and Gram-negative bacteria have previously demonstrated the efficacy profile of PIM1. The present study focuses on the antibacterial and anti-biofilm activities of PIM1 against Salmonella in both in vitro and in ovo settings. In vitro, PIM1 exhibited bactericidal activity against three strains of Salmonella at a low dosage of 8 µg/mL. The anti-biofilm activity of PIM1 was evident by its elimination of planktonic cells within preformed biofilms in a dose-dependent manner. During the host cell infection process, PIM1 reduces the extracellular bacterial load, which reduces adhesion and invasion to limit the establishment of infection. Once intracellular, Salmonella strains were tolerant and protected from PIM1 treatment. In a chicken egg infection model, PIM1 exhibited therapeutic activity for both Salmonella strains, using stationary-phase and exponential-phase inocula. Moreover, PIM1 showed a remarkable efficacy against the stationary-phase inocula of drug-resistant Salmonella by eliminating the bacterial burden in >50% of the infected chicken egg embryos. Collectively, our results highlight the potential for PIM1 as a replacement therapy for existing antibiotic applications on the poultry farm, given the efficiency and low toxicity profile demonstrated in our agriculturally relevant chicken embryo model.


Assuntos
Salmonelose Animal , Infecções por Salmonella , Embrião de Galinha , Animais , Humanos , Salmonella typhimurium , Antibacterianos/farmacologia , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Biofilmes , Galinhas , Salmonelose Animal/tratamento farmacológico , Salmonelose Animal/prevenção & controle , Salmonelose Animal/microbiologia
5.
Biomacromolecules ; 23(5): 1873-1891, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35471022

RESUMO

In recent years, infectious diseases have again become a critical threat to global public health largely due to the challenges posed by antimicrobial resistance. Conventional antibiotics have played a crucial role in combating bacterial infections; however, their efficacy is significantly impaired by widespread drug resistance. Natural antimicrobial peptides (AMPs) and their polymeric mimics demonstrate great potential for killing bacteria with low propensity of resistance as they target the microbial membrane rather than a specific molecular target, but they are also toxic to the host eukaryotic cells. To minimize antibiotics systemic spread and the required dose that promote resistance and to advocate practical realization of the promising activity of AMPs and polymers, smart systems to target bacteria are highly sought after. This review presents bacterial recognition by various specific targeting molecules and the delivery systems of active components in supramolecules. Bacteria-induced activations of antimicrobial-based nanoformulations are also included. Recent advances in the bacteria targeting and delivery of synthetic antimicrobial agents may assist in developing new classes of highly selective antimicrobial systems which can improve bactericidal efficacy and greatly minimize the spread of bacterial resistance.


Assuntos
Anti-Infecciosos , Infecções Bacterianas , Polímeros Responsivos a Estímulos , Antibacterianos/química , Anti-Infecciosos/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bactérias , Infecções Bacterianas/tratamento farmacológico , Humanos , Polímeros/química , Polímeros/farmacologia
6.
Macromol Rapid Commun ; 43(10): e2100812, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35394089

RESUMO

Cationic polymers are under intense research to achieve prominent antimicrobial activity. However, the cellular and in vivo toxicity caused by nonspecific electrostatic interaction has become a major challenge for their practical applications. Here, the development of a "caging" strategy based on the use of a block copolymer consisting of a stealth block and an anionic block that undergoes degradation in presence of enzymes secreted by selective bacterial pathogens of interest is reported. The results have shown that antimicrobial cationic polymer brushes-coated gold nanorods (AuNRs) can be caged by the block polymer of poly(ethylene glycol) and anionic, lipase-degradable block of ε-caprolactone and methacrylic acid copolymer to afford neutrally charged surfaces. The caged AuNRs are activated by lipase released by bacteria of interest to endow an excellent bactericidal effect but show minimal binding and toxicity against mammalian cells and nonspecific bacteria that do not produce lipase. In this design, AuNRs play multifunctional roles as the scaffolds for polymer brushes, photothermal transducers, and imaging probes for traceable delivery of the activation and delivery of bactericidal cationic polymer brushes. The caging strategy opens new opportunities for the safe delivery of antimicrobial materials for the treatment of bacterial infections.


Assuntos
Nanoestruturas , Polímeros , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Bactérias , Cátions , Lipase , Mamíferos , Nanoestruturas/química , Polietilenoglicóis/química , Polímeros/química , Polímeros/farmacologia
7.
Nano Lett ; 21(2): 899-906, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33448223

RESUMO

Antimicrobial peptides that target the integrity of bacterial envelopes can eradicate pathogens with little development of resistance, but they often inflict nonselective toxicity toward mammalian cells. The prevailing approach to optimize the selectivity of cationic peptides has been to modify their composition. Instead, we invent a new generation of broad-spectrum antibacterial nanoconstructs with negligible mammalian cell toxicity through a competitive displacement of counter polyanions from the complementary polycations. The nanoconstruct, which has a highly cationic Au nanoparticles (NPs) core shielded by polymeric counterions, is inert in nonbacterial environments. When exposed to negatively charged bacterial envelopes, this construct sheds its polyanions, triggering a cationic Au NP/bacterial membrane interaction that rapidly kills Gram-positive and Gram-negative bacteria. The anionic charge and hydrophilicity of the polyanion provides charge neutralization for the peptide-decorated Au NP core, but it is also bacteria-displaceable. These results provide a foundation for the development of other cationic particles and polymeric counterion combinations with potent antimicrobial activity without toxicity.


Assuntos
Peptídeos Catiônicos Antimicrobianos , Nanopartículas Metálicas , Animais , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Ouro , Bactérias Gram-Negativas , Bactérias Gram-Positivas , Testes de Sensibilidade Microbiana
8.
Small ; 17(2): e2006357, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33325629

RESUMO

Nanoparticles have been widely used in detection and killing of bacteria; however, targeting bacteria is still challenging. Delicate design of nanoparticles is required for simultaneous targeting, detection, and therapeutic functions. Here the use of Au/MnFe2 O4 (Au/MFO) Janus nanoparticles to target Gram-positive bacteria via metabolic labeling is reported and realize integrated self-reporting and thermal killing of bacteria. In these nanoparticles, the Au component is functionalized with tetrazine to target trans-cyclooctene group anchored on bacterial cell wall by metabolic incorporation of d-amino acids, and the MFO part exhibits peroxidase activity, enabling self-reporting of bacteria before treatment. The spatial separation of targeting and reporting functions avoids the deterioration of catalytic activity after surface modification. Also important is that MFO facilitates magnetic separation and magnetic heating, leading to easy enrichment and magnetic thermal therapy of labeled bacteria. This method demonstrates that metabolic labeling with d-amino acids is a promising strategy to specifically target and kill Gram-positive bacteria.


Assuntos
Nanopartículas de Magnetita , Nanopartículas Multifuncionais , Nanopartículas , Bactérias Gram-Positivas , Magnetismo
9.
Macromol Rapid Commun ; 42(20): e2100255, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34418208

RESUMO

Pseudomonas aeruginosa (P. aeruginosa) is a life-threatening pathogen associated with multiantibiotic resistance, which is largely caused by its strong ability to form biofilms. Recent research has revealed that gallium (III) shows an activity against the biofilm of P. aeruginosa by interfering with Fe metabolism. The antibacterial activity of the combination of Ga3+ ion and antibiotic rifampicin (RMP) against P. aeruginosa PAO1 is investigated. An anionic polymer poly{{2-[(2-methylprop-2-enoyl)oxy]ethyl}phosphonic acid} (PDMPOH) is exploited to form complexes (GaPD) with Ga3+ . The GaPD complexes act as a carrier of Ga3+ and release Ga3+ via enzymatic degradation by bacterial lipases. GaPD is found to damage the outer membrane, leading to enhanced cellular uptake of RMP and Ga3+ due to increased outer membrane permeability, which inhibits the RNA polymerase and interferes with Fe metabolism. The antibiofilm activity and biocompatibility of the GaPD system offer a promising treatment option for P. aeruginosa biofilm-related infections.


Assuntos
Gálio , Antibacterianos/farmacologia , Biofilmes , Gálio/farmacologia , Testes de Sensibilidade Microbiana , Polímeros , Pseudomonas aeruginosa
10.
Angew Chem Int Ed Engl ; 60(31): 16900-16905, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34018295

RESUMO

Real-time monitoring of the evolution of bacterial infection-associated multiple radical species is critical to accurately profile the pathogenesis and host-defense mechanisms. Here, we present a unique dual wavelength near-infrared (NIR) cyanine-dyad molecular probe (HCy5-Cy7) for simultaneous monitoring of reactive oxygen and nitrogen species (RONS) variations both in vitro and in vivo. HCy5-Cy7 specifically turns on its fluorescence at 660 nm via superoxide or hydroxyl radical (O2.- , . OH)-mediated oxidation of reduced HCy5 moiety to Cy5, while peroxynitrite or hypochlorous species (ONOO- , ClO- )-induced Cy7 structural degradation causes the emission turn-off at 800 nm. Such multispectral but reverse signal responses allow multiplex manifestation of in situ oxidative and nitrosative stress events during the pathogenic and defensive processes in both bacteria-infected macrophage cells and living mice. Most importantly, this study may also provide new perspectives for understanding the bacterial pathogenesis and advancing the precision medicine against infectious diseases.


Assuntos
Infecções Bacterianas/diagnóstico por imagem , Carbocianinas/química , Corantes/química , Animais , Camundongos , Células RAW 264.7 , Espécies Reativas de Nitrogênio/análise , Espécies Reativas de Oxigênio/análise
11.
Macromol Rapid Commun ; 41(21): e2000279, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32840324

RESUMO

The sensitive, safe, and portable detection of food spoilage is becoming unprecedentedly important because it is closely related to the public health and economic development, particularly given the globalization of food supply chain. However, the existing approaches for food monitoring are still limited to meet these requirements. To address this challenge, much research has been done to develop an ideal food sensor that can indicate food quality in real-time in a sensitive and reliable way. So far, many sensors such as time-temperature indicators, smart trademarks, colorimetric tags, electronic noses, and electronic tongues, have been developed and even commercialized. In this feature article, the recent progress of food sensors based on functional polymers, including the molecular design of polymer structures, sensing mechanisms, and relevant processing techniques to fabricate a variety of food sensor devices is reviewed.


Assuntos
Técnicas Biossensoriais , Polímeros
12.
Macromol Rapid Commun ; 41(21): e2000175, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32803821

RESUMO

Contact lenses are medical devices commonly used to correct refractive errors and to maintain ocular health. Microorganisms such as bacteria that grow on the lens surface cause irritation to the eyes and can even cause loss of vision. In this paper, two different coating strategies are employed to form an efficient antimicrobial coating on contact lenses. In the first method, a presynthesized copolymer of polyethyleneimine-graft-polyethylene glycol methacrylate (PEI-PEGMA) is used and the coated lenses show antimicrobial activity (in vitro) against methicillin-resistant Staphylococcus aureus (MRSA) bacteria with killing efficacy >99.99% and log reduction of 5.1 and proxy host cell viability of 79%. In the second method, commercially available monomers/polymers such as glycidyl methacrylate (GMA), sulfobetaine methacrylate, and polyethyleneimine are used. A typical formulation consisting of 1% GMA shows antibacterial activity against MRSA with killing efficacy >99.99% and log reduction of 6.3. Proxy host cell viability for the coated lenses is found to be 90% indicating that the coating is nontoxic. Antibacterial coating reported here is very effective in killing gram-positive bacteria such as MRSA and S. aureus. The second method using commercially available monomers/polymers involving a simple coating procedure is also easily scalable.


Assuntos
Anti-Infecciosos , Lentes de Contato , Staphylococcus aureus Resistente à Meticilina , Antibacterianos/farmacologia , Anti-Infecciosos/farmacologia , Materiais Revestidos Biocompatíveis , Compostos de Epóxi , Metacrilatos , Polietilenoimina , Polímeros , Silicones , Staphylococcus aureus
13.
Nano Lett ; 19(11): 7712-7724, 2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31565943

RESUMO

The corona phase-the adsorbed layer of polymer, surfactant, or stabilizer molecules around a nanoparticle-is typically utilized to disperse nanoparticles into a solution or solid phase. However, this phase also controls molecular access to the nanoparticle surface, a property important for catalytic activity and sensor applications. Unfortunately, few methods can directly probe the structure of this corona phase, which is subcategorized as either a hard, immobile corona or a soft, transient corona in exchange with components in the bulk solution. In this work, we introduce a molecular probe adsorption (MPA) method for measuring the accessible nanoparticle surface area using a titration of a quenchable fluorescent molecule. For example, riboflavin is utilized to measure the surface area of gold nanoparticle standards, as well as corona phases on dispersed single-walled carbon nanotubes and graphene sheets. A material balance on the titration yields certain surface coverage parameters, including the ratio of the surface area to dissociation constant of the fluorophore, q/KD, as well as KD itself. Uncertainty, precision, and the correlation of these parameters across different experimental systems, preparations, and modalities are all discussed. Using MPA across a series of corona phases, we find that the Gibbs free energy of probe binding scales inversely with the cube root of surface area, q. In this way, MPA is the only technique to date capable of discerning critical structure-property relationships for such nanoparticle surface phases. Hence, MPA is a rapid quantitative technique that should prove useful for elucidating corona structure for nanoparticles across different systems.


Assuntos
Nanopartículas/química , Nanotubos de Carbono/química , Coroa de Proteína/análise , Adsorção , Corantes Fluorescentes/análise , Ouro/química , Grafite/química , Nanoestruturas/química , Riboflavina/análise , Espectrometria de Fluorescência/métodos , Propriedades de Superfície
14.
Angew Chem Int Ed Engl ; 59(17): 6819-6826, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32011781

RESUMO

Carbapenem-resistant Gram-negative bacteria (GNB) are heading the list of pathogens for which antibiotics are the most critically needed. Many antibiotics are either unable to penetrate the outer-membrane or are excluded by efflux mechanisms. Here, we report a cationic block ß-peptide (PAS8-b-PDM12) that reverses intrinsic antibiotic resistance in GNB by two distinct mechanisms of action. PAS8-b-PDM12 does not only compromise the integrity of the bacterial outer-membrane, it also deactivates efflux pump systems by dissipating the transmembrane electrochemical potential. As a result, PAS8-b-PDM12 sensitizes carbapenem- and colistin-resistant GNB to multiple antibiotics in vitro and in vivo. The ß-peptide allows the perfect alternation of cationic versus hydrophobic side chains, representing a significant improvement over previous antimicrobial α-peptides sensitizing agents. Together, our results indicate that it is technically possible for a single adjuvant to reverse innate antibiotic resistance in all pathogenic GNB of the ESKAPE group, including those resistant to last resort antibiotics.


Assuntos
Antibacterianos/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Peptídeos/química , Peptídeos/farmacologia , Carbapenêmicos/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Glicosilação , Testes de Sensibilidade Microbiana , Conformação Proteica em Folha beta
15.
Biomacromolecules ; 20(2): 949-958, 2019 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-30629424

RESUMO

Most synthetic antimicrobial polymers are not biodegradable, thus limiting their potential for large-scale applications in personal care disinfection and environmental contaminations. Poly(ε-caprolactone) (PCL) is known to be both biodegradable and biocompatible, thus representing an ideal candidate biopolymer for antimicrobial applications. Here we successfully grafted alkylimidazolium (Im) onto PCL to mimic the cationic properties of antimicrobial peptides. The poly(ε-caprolactone)- graft-butylimidazolium had only moderate MICs (32 µg/mL), reasonably good red blood cell selectivity (36) and relatively good fibroblast compatibility (81% cell viability at 100 µg/mL), indicating that combining the hydrophobic PCL backbone with the most hydrophilic butylimidazolium gives a good balance of MIC and cytotoxicity. On the other hand, the PCL- graft-hexylimidazolium and -octylimidazolium demonstrated better MICs (4-32 µg/mL), but considerably worse cytotoxicity. We postulated that the worse hydrophilicity of hexylimidazolium and octylimidazolium was responsible for their higher cytotoxicity and sought to moderate their cytotoxicity with different sugar compositions and lengths. Through our screening, we identified a candidate polymer, P(C6Im)0.35CL- co-P(Man)0.65CL, that demonstrated both superior MIC and very low cytotoxicity. We further demonstrated that our biopolymer hit had superior antimicrobial kinetics compared to the antibiotic vancomycin. This work paves the way forward for the use of biodegradable polyesters as the backbone scaffold for biocompatible antibacterial agents, by clicking with different types and ratios of alkylimidazolium and carbohydrate moieties.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Hemólise/efeitos dos fármacos , Poliésteres/química , Células 3T3 , Animais , Peptídeos Catiônicos Antimicrobianos/química , Materiais Biocompatíveis/química , Biopolímeros/química , Caproatos/química , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/química , Fibroblastos/efeitos dos fármacos , Humanos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Lactonas/química , Camundongos , Polímeros/química
16.
Biomacromolecules ; 20(8): 2922-2933, 2019 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-31305998

RESUMO

To combat the increasing risk of infection by pathogenic bacteria, the new generation of antimicrobial agents is expected to exhibit nonmetabolic killing mechanisms, high potency and biocompatibility. In this work, cysteine-terminated antimicrobial peptide (AMP) was employed directly as a reducing ligand to synthesize AMP-coated gold nanoclusters (Au NCs), bypassing the use of other reagents which might interfere with the efficacy of the resulting NCs. In addition to the use of a biocompatible Au core, the primary amines of AMP coating were functionalized with anionic citraconyl moieties to further reduce cytotoxicity. The citraconyl amides could autocleave to re-expose the cationic amines at low pH. As a result, the AMP-coated Au NCs with citraconyl protection were stable and cytocompatible under physiological conditions as determined by fluorescamine, hemolytic, cytotoxicity, and in vivo toxicology studies, but would switch into a cationic bactericidal mode in an acidic environment commonly encountered at bacterial infection sites. Furthermore, the AMP-coated Au NCs system exhibited bacterial binding and photoluminescence features as determined by flow cytometry and confocal microscopy, which were useful for the detection and imaging of bacterial contamination. The AMP-coated Au NCs with citraconyl moieties therefore represent a "smart" design of pH-responsive antimicrobial agents that can serve multiple functions of bacterial detection, bacterial imaging, and anti-infection therapy.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bactérias/efeitos dos fármacos , Ouro/química , Hemólise/efeitos dos fármacos , Nanopartículas Metálicas/administração & dosagem , Células 3T3 , Animais , Antibacterianos/química , Peptídeos Catiônicos Antimicrobianos/química , Bactérias/química , Proliferação de Células , Humanos , Nanopartículas Metálicas/química , Camundongos , Espécies Reativas de Oxigênio/metabolismo
17.
J Chem Inf Model ; 59(4): 1575-1583, 2019 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-30855952

RESUMO

Ever since the discovery of the new antibiotic teixobactin, studies of its structure-activity relationships have never ceased. Here we focus on the chirality of the threonine (Thr) residue, which belongs to the ring motif of teixobactin and plays an important role in the binding with its target, lipid II molecule. We study the structural propensity of the open and closed ring motifs with different chiral Thr residues as well as the teixobactin-lipid II complex with the help of molecular dynamics simulations. Our results suggest that different chiralities lead to different NH orientations of Thr with respect to the ring plane. Only in the closed ring motif with d-Thr is a favored binding cavity achievable with all four NH groups facing the same side of the ring plane. This study develops a deeper understanding of the binding mechanism of teixobactin and lipid II and is expected to be beneficial to new teixobactin-based drug design.


Assuntos
Antibacterianos/química , Antibacterianos/metabolismo , Depsipeptídeos/química , Depsipeptídeos/metabolismo , Simulação de Dinâmica Molecular , Treonina , Motivos de Aminoácidos , Conformação Proteica , Uridina Difosfato Ácido N-Acetilmurâmico/análogos & derivados , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo
18.
Nano Lett ; 18(7): 4180-4187, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29902011

RESUMO

Biofilms and the rapid evolution of multidrug resistance complicate the treatment of bacterial infections. Antibiofilm agents such as metallic-inorganic nanoparticles or peptides act by exerting antibacterial effects and, hence, do not combat biofilms of antibiotics-resistant strains. In this Letter, we show that the block copolymer DA95B5, dextran- block-poly((3-acrylamidopropyl) trimethylammonium chloride (AMPTMA)- co-butyl methacrylate (BMA)), effectively removes preformed biofilms of various clinically relevant multidrug-resistant Gram-positive bacteria including methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococci (VRE V583), and Enteroccocus faecalis (OG1RF). DA95B5 self-assembles into core-shell nanoparticles with a nonfouling dextran shell and a cationic core. These nanoparticles diffuse into biofilms and attach to bacteria but do not kill them; instead, they promote the gradual dispersal of biofilm bacteria, probably because the solubility of the bacteria-nanoparticle complex is enhanced by the nanoparticle dextran shell. DA95B5, when applied as a solution to a hydrogel pad dressing, shows excellent in vivo MRSA biofilm removal efficacy of 3.6 log reduction in a murine excisional wound model, which is significantly superior to that for vancomycin. Furthermore, DA95B5 has very low in vitro hemolysis and negligible in vivo acute toxicity. This new strategy for biofilm removal (nanoscale bacterial debridement) is orthogonal to conventional rapidly developing resistance traits in bacteria so that it is as effective toward resistant strains as it is toward sensitive strains and may have widespread applications.


Assuntos
Biofilmes/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Nanopartículas/administração & dosagem , Enterococos Resistentes à Vancomicina/efeitos dos fármacos , Antibacterianos/efeitos adversos , Dextranos/administração & dosagem , Dextranos/química , Humanos , Metacrilatos/administração & dosagem , Metacrilatos/química , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Staphylococcus aureus Resistente à Meticilina/patogenicidade , Testes de Sensibilidade Microbiana , Nanopartículas/química , Enterococos Resistentes à Vancomicina/crescimento & desenvolvimento , Enterococos Resistentes à Vancomicina/patogenicidade
19.
Biomacromolecules ; 19(6): 2156-2165, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29672023

RESUMO

The rapid spread of multidrug-resistant bacteria has called for effective antimicrobial agents which work on a more direct mechanism of killing. Cationic peptidopolysaccharides are developed in the present work to mimic the peptidoglycan structure of bacteria and to enhance the membrane-compromising bactericidal efficacy. Antimicrobial CysHHC10 peptide was grafted to the C-2 (amino) or C-6 (hydroxyl) position of chitosan backbone via thiol-maleimide "click" conjugation, utilizing the maleimidohexanoic linkers. The peptidopolysaccharide with primary amino backbone intact (CSOHHC) exhibited higher bactericidal activity toward Gram-positive and Gram-negative bacteria, in comparison to that with amino backbone grafted with the peptide (CSNHHC). Both peptidopolysaccharides also exhibited lower hemolytic activity and cytotoxicity than free CysHHC10 peptide due to the moderation effect contributed by the chitosan backbone. For targeting the Gram-positive bacteria in particular, the CSOHHC expressed 4- and 2-fold increases in hemo- and cytoselectivity, respectively, as compared to the CysHHC10 peptide. In an extended application, peptidopolysaccharide antibacterial coatings were formed via layer-by-layer assembly with tannic acid. The peptidopolysaccharide coatings readily killed the adhered bacteria upon contact while being cytocompatible by maintaining more than 60% viability for the adhered fibroblasts. Therefore, the peptidoglycan-mimetic peptidopolysaccharides are potential candidates for anti-infective drugs in biomedical applications.


Assuntos
Antibacterianos , Peptídeos Catiônicos Antimicrobianos , Quitosana , Materiais Revestidos Biocompatíveis , Bactérias Gram-Negativas/crescimento & desenvolvimento , Bactérias Gram-Positivas/crescimento & desenvolvimento , Polissacarídeos , Células 3T3 , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Quitosana/química , Quitosana/farmacologia , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Teste de Materiais , Camundongos , Polissacarídeos/química , Polissacarídeos/farmacologia
20.
Biomacromolecules ; 18(1): 44-55, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-28009508

RESUMO

Cationic polymethacrylates are interesting candidates for bacterial disinfectants since they can be made in large-scale by various well-established polymerization techniques such as atom transfer radical polymerization (ATRP). However, they are usually toxic or ineffective in serum and various strategies to improve their biocompatibility or nonfouling property have often resulted in compromised bactericidal activity. Also, star-shaped polymers are less explored than linear polymers for application as antibacterial compounds. In this paper, star polymers with poly[2-(dimethylamino)ethyl methacrylate] (PDMA) as the arms and polyhedral oligomeric silsesquioxane (POSS) as the core (POSS-g-PDMA) were successfully synthesized by ATRP. The minimum inhibition concentrations (MICs) of the synthesized POSS-g-PDMA are in the range of 10-20 µg/mL. POSS-g-PDMA was further modified by various hydrophilization strategies in attempting to reduce hemolysis. With quaternization of POSS-g-PDMA, the antibacterial activities of the obtained quaternary polymers are almost unchanged and the copolymers become relatively nonhemolytic. We also copolymerized sulfobetaine (SB) with POSS-g-PDMA to obtain random and block PDMA-co-PSB arm structures, where the PDMA and poly(sulfobetaine) were the cationic and zwitterionic blocks, respectively. The random cationic-zwitterionic POSS-g-(PDMA-r-PSB) copolymers showed poor antibacterial activity, while the block POSS-g-(PDMA-b-PSB) copolymers retained the antibacterial and hemolytic activity of the pristine POSS-g-PDMA. Further, the block copolymers of POSS-g-(PDMA-b-PSB) showed enhanced antifouling property and serum stability as seen by their nanoparticle size stability in the presence of serum and reduced red blood cell aggregation; the POSS-g-(PDMA-b-PSB) also somewhat retained its MIC in blood unlike the quaternized or random zwitterionic copolymers. The antibacterial kinetics study showed that Escherichia coli can be killed within 30 min by both random and block copolymers of POSS-g-(PDMA-co-PSB). Finally, our POSS star polymers showed low toxicity to zebrafish embryo and could be potentially used in aquaculture antibacterial applications.


Assuntos
Antibacterianos/síntese química , Antibacterianos/farmacologia , Betaína/análogos & derivados , Metacrilatos/química , Polímeros/síntese química , Polímeros/farmacologia , Compostos de Amônio Quaternário/química , Animais , Betaína/química , Embrião não Mamífero/citologia , Embrião não Mamífero/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Células NIH 3T3 , Peixe-Zebra/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA