Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Cont Lens Anterior Eye ; 47(2): 102124, 2024 Apr.
Article En | MEDLINE | ID: mdl-38341309

BACKGROUND: Ocular infections caused by antibiotic-resistant pathogens can result in partial or complete vision loss. The development of pan-resistant microbial strains poses a significant challenge for clinicians as there are limited antimicrobial options available. Synthetic peptoids, which are sequence-specific oligo-N-substituted glycines, offer potential as alternative antimicrobial agents to target multidrug-resistant bacteria. METHODS: The antimicrobial activity of synthesised peptoids against multidrug-resistant (MDR) ocular pathogens was evaluated using the microbroth dilution method. Hemolytic propensity was assessed using mammalian erythrocytes. Peptoids were also incubated with proteolytic enzymes, after which their minimum inhibitory activity against bacteria was re-evaluated. RESULTS: Several alkylated and brominated peptoids showed good inhibitory activity against multidrug-resistant Pseudomonas aeruginosa strains at concentrations of ≤15 µg mL-1 (≤12 µM). Similarly, most brominated compounds inhibited the growth of methicillin-resistant Staphylococcus aureus at 1.9 to 15 µg mL-1 (12 µM). The N-terminally alkylated peptoids caused less toxicity to erythrocytes. The peptoid denoted as TM5 had a high therapeutic index, being non-toxic to either erythrocytes or corneal epithelial cells, even at 15 to 22 times its MIC. Additionally, the peptoids were resistant to protease activity. CONCLUSIONS: Peptoids studied here demonstrated potent activity against various multidrug-resistant ocular pathogens. Their properties make them promising candidates for controlling vision-related morbidity associated with eye infections by antibiotic-resistant strains.


Anti-Infective Agents , Methicillin-Resistant Staphylococcus aureus , Peptoids , Animals , Humans , Peptoids/pharmacology , Microbial Sensitivity Tests , Anti-Infective Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Mammals
2.
ACS Infect Dis ; 8(8): 1480-1490, 2022 08 12.
Article En | MEDLINE | ID: mdl-35771275

The escalating issue of multidrug-resistant (MDR) bacteria indicates the urgent need for new and effective strategies to combat this global health challenge. Here, we describe a new combinatorial approach that can be put forward for experimental therapy application against MDR bacteria. Specifically, we have developed a tri-system that includes the coadministration of two different membrane-disrupting-type antimicrobial agents─a synthetic antimicrobial polymer P and an antimicrobial peptide (AMP) colistin methanesulfonate (Col)─in conjunction with an antibiotic [doxycycline (Dox), rifampicin (Rif), or azithromycin (Azi)]. Traditionally, the administration of membrane-disrupting antimicrobial agents causes toxicity, but, in comparison, we demonstrated synergy and biocompatibility using this combinatorial approach. Checkerboard assays showed the occurrence of synergistic interactions in Col-Dox-P, Col-Rif-P, and Col-Azi-P tri-systems against wild-type and MDR Pseudomonas aeruginosa, with the Col-Dox-P system being the most effective. The ability to synergize thus enables the use of a lower dosage in combinations compared to the standalone agents. The tri-systems not only demonstrated bacteriostatic activity but were also bactericidal. For example, the Col-Dox-P system (at 8, 4, and 8 µg mL-1, respectively) and the Col-Rif-P system (at 4, 8, and 16 µg mL-1, respectively) were able to kill >99.999% of planktonic P. aeruginosa cells within 3 h of treatment. More importantly, an improvement of the therapeutic/selectivity index was achieved via combination therapy. Taking the Col-Dox-P system as an example, its biocompatibility with murine embryonic fibroblast cells was found to be comparable to that of polymer P alone despite the synergistic enhancement in antimicrobial activity of the combination. This resulted in a significant increase in selectivity by 16-fold for the Col-Dox-P combination system compared to P alone. Furthermore, the broad applicability of this tri-system strategy was demonstrated via the successful application of the AMP melittin in place of Col or P. Overall, this study sheds new insights on the application of membrane-disrupting antimicrobial agents in combination therapy and their potential for safer clinical use. Additionally, the information gathered in this study could inform the development of future combination therapy systems involving the simultaneous employment of multiple AMPs with antibiotics.


Anti-Bacterial Agents , Anti-Infective Agents , Animals , Mice , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Doxycycline , Drug Synergism , Microbial Sensitivity Tests , Polymers , Rifampin/pharmacology
3.
Adv Mater ; 34(2): e2105063, 2022 Jan.
Article En | MEDLINE | ID: mdl-34611948

Synthetic polymers are omnipresent in society as textiles and packaging materials, in construction and medicine, among many other important applications. Alternatively, natural polymers play a crucial role in sustaining life and allowing organisms to adapt to their environments by performing key biological functions such as molecular recognition and transmission of genetic information. In general, the synthetic and natural polymer worlds are completely separated due to the inability for synthetic polymers to perform specific biological functions; in some cases, synthetic polymers cause uncontrolled and unwanted biological responses. However, owing to the advancement of synthetic polymerization techniques in recent years, new synthetic polymers have emerged that provide specific biological functions such as targeted molecular recognition of peptides, or present antiviral, anticancer, and antimicrobial activities. In this review, the emergence of this generation of bioactive synthetic polymers and their bioapplications are summarized. Finally, the future opportunities in this area are discussed.


Peptides , Polymers , Polymerization , Polymers/chemistry , Textiles
4.
Angew Chem Int Ed Engl ; 60(45): 24248-24256, 2021 11 02.
Article En | MEDLINE | ID: mdl-34453390

This work presents the synthesis of a novel photosensitive acrylate monomer for use as both a self-catalyst in the photoinduced electron/energy transfer-reversible addition fragmentation chain transfer (PET-RAFT) polymerisation process and a photosensitiser (PS) for antibacterial applications. Hydrophilic, cationic, and antimicrobial formulations are explored to compare the antibacterial effects between charged and non-charged polymers. Covalent attachment of the catalyst to well-defined linear polymer chains has no effect on polymerisation control or singlet oxygen generation. The addition of the PS to polymers provides activity against S. aureus for all polymer formulations, resulting in up to a 99.99999 % killing efficacy in 30 min. Antimicrobial peptide mimetic polymers previously active against P. aeruginosa, but not S. aureus, gain significant bactericidal activity against S. aureus through the inclusion of PS groups, with 99.998 % killing efficiency after 30 min incubation with light. Thus, a broader spectrum of antimicrobial activity is achieved using two distinct mechanisms of bactericidal activity via the incorporation of a photosensitiser monomer into an antimicrobial polymer.


Anti-Bacterial Agents/pharmacology , Antimicrobial Peptides/pharmacology , Photosensitizing Agents/pharmacology , Polymers/pharmacology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antimicrobial Peptides/chemical synthesis , Antimicrobial Peptides/chemistry , Microbial Sensitivity Tests , Photochemical Processes , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Polymers/chemical synthesis , Polymers/chemistry
5.
ACS Infect Dis ; 7(2): 215-253, 2021 02 12.
Article En | MEDLINE | ID: mdl-33433995

Antibiotic resistance is a critical global healthcare issue that urgently needs new effective solutions. While small molecule antibiotics have been safeguarding us for nearly a century since the discovery of penicillin by Alexander Fleming, the emergence of a new class of antimicrobials in the form of synthetic antimicrobial polymers, which was driven by the advances in controlled polymerization techniques and the desire to mimic naturally occurring antimicrobial peptides, could play a key role in fighting multidrug resistant bacteria in the near future. By harnessing the ability to control chemical and structural properties of polymers almost at will, synthetic antimicrobial polymers can be strategically utilized in combination therapy with various antimicrobial coagents in different formats to yield more potent (synergistic) outcomes. In this review, we present a short summary of the different combination therapies involving synthetic antimicrobial polymers, focusing on their combinations with nitric oxide, antibiotics, essential oils, and metal- and carbon-based inorganics.


Anti-Bacterial Agents , Anti-Infective Agents , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Polymers
6.
Sci Rep ; 11(1): 1690, 2021 01 18.
Article En | MEDLINE | ID: mdl-33462270

An antimicrobial peptide, nisin Z, was embedded within polyelectrolyte multilayers (PEMs) composed of natural polysaccharides in order to explore the potential of forming a multilayer with antimicrobial properties. Using attenuated total reflection Fourier transform infrared spectroscopy (ATR FTIR), the formation of carrageenan/chitosan multilayers and the inclusion of nisin Z in two different configurations was investigated. Approximately 0.89 µg cm-2 nisin Z was contained within a 4.5 bilayer film. The antimicrobial properties of these films were also investigated. The peptide containing films were able to kill over 90% and 99% of planktonic and biofilm cells, respectively, against Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA) strains compared to control films. Additionally, surface topography and wettability studies using atomic force microscopy (AFM) and the captive bubble technique revealed that surface roughness and hydrophobicity was similar for both nisin containing multilayers. This suggests that the antimicrobial efficacy of the peptide is unaffected by its location within the multilayer. Overall, these results demonstrate the potential to embed and protect natural antimicrobials within a multilayer to create functionalised coatings that may be desired by industry, such as in the food, biomaterials, and pharmaceutical industry sectors.


Anti-Bacterial Agents/pharmacology , Biofilms/growth & development , Carrageenan/chemistry , Chitosan/chemistry , Coated Materials, Biocompatible/chemistry , Nisin/analogs & derivatives , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Nisin/chemistry , Nisin/pharmacology , Staphylococcus aureus/physiology , Surface Properties
7.
Biomacromolecules ; 21(12): 5241-5255, 2020 12 14.
Article En | MEDLINE | ID: mdl-33186496

Antimicrobial polymers have emerged as a potential solution to the growing problem of antimicrobial resistance. Although several studies have examined the effects of various parameters on the antimicrobial and hemolytic activity of statistical copolymers, there are still numerous parameters to be explored. Therefore, in this study, we developed a library of 36 statistical amphiphilic ternary copolymers prepared via photoinduced electron transfer-reversible addition-fragmentation chain transfer polymerization to systematically evaluate the influence of hydrophobic groups [number of carbons (5, 7, and 9)] and chain type of the hydrophobic monomer (cyclic, aromatic, linear, or branched), monomer ratio, and degree of polymerization (DPn) on antimicrobial and hemolytic activity. To guide our synthetic strategy, we developed a pre-experimental screening approach using C log P values of oligomer models, which correspond to the logarithm of the partition coefficient of compounds between n-octanol and water. This method enabled correlation of polymer hydrophobicity with antimicrobial and hemolytic activity. In addition, this study revealed that minimizing hydrophobicity and hydrophobic content were key factors in controlling hemolysis, whereas optimizing antimicrobial activity was more complex. High antimicrobial activity required hydrophobicity (i.e., C log P, hydrophobicity index) that was neither too high nor too low, an appropriate cationic/hydrophobic balance, and structural compatibility between the chosen monomers. Furthermore, these findings could guide the design of future antimicrobial ternary copolymers and suggest that C log P values between 0 and 2 have the best balance of high antimicrobial activity and low hemolytic activity.


Anti-Infective Agents , Hemolysis , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Humans , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Polymers
8.
Biomacromolecules ; 21(1): 262-272, 2020 01 13.
Article En | MEDLINE | ID: mdl-31657209

The development of potent strategies to counter microbial biofilm is an urgent priority in healthcare. The majority of bacterial infections in humans are biofilm related, however, effective treatments are still lacking especially for combating multidrug-resistant (MDR) strains. Herein, we report an effective antibiofilm platform based on the use of synthetic antimicrobial polymers in combination with essential oils, where the antimicrobial polymers play a secondary role as delivery vehicle for essential oils. Two ternary antimicrobial polymers consisting of cationic primary amines, low-fouling oligo(ethylene glycol) and hydrophobic ethylhexyl groups were synthesized in the form of random and block copolymers, and mixed with either carvacrol or eugenol. Coadministration of these compounds improved the efficacy against Pseudomonas aeruginosa biofilms compared to the individual compounds. We observed about a 60-75% and 70-85% biofilm inhibition effect for all tested combinations against wild-type P. aeruginosa PAO1 and MDR strain PA37, respectively, upon 6.5 h of incubation time. While both random and block copolymers demonstrated similar biofilm inhibition potencies in combination with essential oils, only the block copolymer acted synergistically with essential oils in killing biofilm. Treatment of PAO1 biofilm for 20 min with the block copolymer-oil combinations resulted in the killing of >99.99% of biofilm bacteria. This synergistic bactericidal activity is attributed to the targeted delivery of essential oils to the biofilm, driven by the electrostatic interaction between positively charged delivery vehicles, in the form of polymeric micelles, and negatively charged bacteria. This study thus highlights the advantage of combining essential oils and antimicrobial polymers as an effective avenue for antibacterial applications.


Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Oils, Volatile/pharmacology , Polymers/chemistry , Anti-Bacterial Agents/chemistry , Anti-Infective Agents/pharmacology , Biofilms/growth & development , Cymenes/chemistry , Cymenes/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Combinations , Drug Delivery Systems/methods , Drug Resistance, Multiple, Bacterial/drug effects , Dynamic Light Scattering , Eugenol/chemistry , Eugenol/pharmacology , Hemolysis/drug effects , Humans , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Oils, Volatile/administration & dosage , Oils, Volatile/chemistry , Polymers/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Spectrophotometry, Ultraviolet
9.
ACS Infect Dis ; 5(8): 1357-1365, 2019 08 09.
Article En | MEDLINE | ID: mdl-30939869

The failure of many antibiotics in the treatment of chronic infections caused by multidrug-resistant (MDR) bacteria necessitates the development of effective strategies to combat this global healthcare issue. Here, we report an antimicrobial platform based on the synergistic action between commercially available antibiotics and a potent synthetic antimicrobial polymer that consists of three key functionalities: low-fouling oligoethylene glycol, hydrophobic ethylhexyl, and cationic primary amine groups. Checkerboard assays with Pseudomonas aeruginosa (P. aeruginosa) and Escherichia coli demonstrated synergy between our synthetic antimicrobial polymer and two antibiotics, doxycycline and colistin. Coadministration of these compounds significantly improved the bacteriostatic efficacy especially against MDR P. aeruginosa strains PA32 and PA37, where the minimal inhibitory concentrations (MICs) of polymer and antibiotics were reduced by at least 4-fold. A synergistic killing activity was observed when the antimicrobial polymer was used in combination with doxycycline, killing >99.999% of planktonic and biofilm P. aeruginosa PAO1 upon a 20 min treatment at a polymer concentration of 128 µg mL-1 (4.6 µM) and doxycycline concentration of 64 µg mL-1 (133.1 µM). In addition, this synergistic combination reduced the rate of resistance development in P. aeruginosa compared to individual compounds and was also capable of reviving susceptibility to treatment in the resistant strains.


Anti-Infective Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Multiple, Bacterial , Polymers/pharmacology , Biofilms/drug effects , Colistin/pharmacology , Doxycycline/pharmacology , Drug Synergism , Escherichia coli/drug effects , Microbial Sensitivity Tests , Polymers/chemical synthesis , Pseudomonas aeruginosa/drug effects
10.
ACS Appl Mater Interfaces ; 11(7): 7320-7329, 2019 Feb 20.
Article En | MEDLINE | ID: mdl-30688429

The growing number of patient morbidity related to nosocomial infections has placed an importance on the development of new antibacterial coatings for medical devices. Here, we utilize the versatile adhesion property of polydopamine (pDA) to design an antibacterial coating that possesses low-fouling and nitric oxide (NO)-releasing capabilities. To demonstrate this, glass substrates were functionalized with pDA via immersion in alkaline aqueous solution containing dopamine, followed by grafting of low-fouling polymer (poly(ethylene glycol) (PEG)) via Michael addition and subsequent formation of N-diazeniumdiolate functionalities (NO precursors) by purging with NO gas. X-ray photoelectron spectroscopy confirmed the successful grafting of PEG and formation of N-diazeniumdiolate on polydopamine-coated substrates. NO release from the coating was observed over 2 days, and NO loading is tunable by the pDA film thickness. The antibacterial efficiency of the coatings was assessed using Gram-negative Pseudomonas aeruginosa (i.e., wild-type PAO1 and multidrug-resistant PA37) and Gram-positive Staphylococcus aureus (ATCC 29213). The NO-releasing PEGylated pDA film inhibited biofilm attachment by 96 and 70% after exposure to bacterial culture solution for 24 and 36 h, respectively. In contrast, films that do not contain NO failed to prevent biofilm formation on the surfaces at these time points. Furthermore, this coating also showed 99.9, 97, and 99% killing efficiencies against surface-attached PAO1, PA37, and S. aureus bacteria. Overall, the combination of low-fouling PEG and antibacterial activity of NO in pDA films makes this coating a potential therapeutic option to inhibit biofilm formation on medical devices.


Anti-Bacterial Agents , Biofilms/drug effects , Coated Materials, Biocompatible , Indoles , Nitric Oxide , Polymers , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/physiology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/growth & development , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Indoles/chemistry , Indoles/pharmacology , Nitric Oxide/chemistry , Nitric Oxide/pharmacology , Polymers/chemistry , Polymers/pharmacology
11.
ACS Biomater Sci Eng ; 5(11): 5881-5887, 2019 Nov 11.
Article En | MEDLINE | ID: mdl-33405678

The development of novel strategies for the prevention of bacterial infections is of utmost importance because of the exponential growth in the number of patient morbidity related to nosocomial and chronic infections. Nitric oxide (NO) is known to be a potent inhibitor of bacterial growth and adhesion to surfaces. Here, we develop an antibiofilm coating that possesses S-nitrosothiol NO donors via plasma polymerization (PP) for biofilm prevention applications. Cell culture dishes of four different film thicknesses ranging from 125 to 1000 nm were coated via PP using a thiol monomer. The thiol functionality on the substrates was converted to S-nitrosothiol NO precursors using tert-butyl nitrite. The successful conjugation of thiol and subsequent formation of S-nitrosothiol functionalities on the substrates were confirmed using X-ray photoelectron spectroscopy and UV-vis analysis. These coatings are capable of releasing NO over 2 days, and the NO loading is tunable by the polymer film thickness. The antibiofilm activity of the surfaces was assessed using Gram-negative bacteria, Pseudomonas aeruginosa. Higher film thickness (and hence, higher NO loading) demonstrate better antibiofilm activity, and the best performing coating shows 81 and 60% inhibition of bacterial attachment to the surface after exposure to bacterial culture solution for 24 and 36 h, respectively. Overall, the NO-releasing plasma-modified surfaces present a potential viable strategy to inhibit bacterial biofilm formation.

12.
Sci Rep ; 8(1): 7965, 2018 05 21.
Article En | MEDLINE | ID: mdl-29785029

Access to clean and safe water supply remains inadequate in many developing countries. One of the key challenges is to remove pathogenic bacteria from the water supply via effective water disinfection technologies to prevent the spread of diseases and to ensure the safety of consumers. Herein, a highly effective point-of-use (on-demand) water disinfection technology, in the form of a polymeric scaffold called macroporous antimicrobial polymeric gel (MAPG), is demonstrated. MAPG is easy to fabricate, completely organic and possess inherent antimicrobial property which makes it non-reliant on inorganic compounds such as silver where the long-term toxicity remains unknown. MAPG is highly bactericidal and can disinfect bacteria-contaminated water (ca. 108 CFU mL-1) at a capacity of about >50 times the mass of the organic material used, inactivating >99% of both Gram-negative and Gram-positive bacteria including Escherichia coli, Vibrio cholerae and Staphylococcus aureus within 20 minutes of treatment. When fabricated in a syringe, MAPG eliminates E. coli from contaminated water source by >8.0 log10 reduction in bacteria counts (i.e., no viable bacteria were detected after treatment), and the syringe can be reused multiple times without losing potency. The MAPG technology is not only restricted to water disinfection but may also be applicable in other bacteria inactivation applications.


Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Disinfectants/pharmacology , Point-of-Care Testing/statistics & numerical data , Polymers/pharmacology , Water Purification/methods , Gels , Water Microbiology
13.
Macromol Rapid Commun ; 39(13): e1800159, 2018 Jul.
Article En | MEDLINE | ID: mdl-29806143

In this study, an antimicrobial platform in the form of nitric oxide (NO) gas-releasing polydopamine (PDA)-coated iron oxide nanoparticles (IONPs) is developed for combating bacterial biofilms. NO is bound to the PDA-coated IONPs via the reaction between NO and the secondary amine moieties on PDA to form N-diazeniumdiolate (NONOate) functionality. To impart colloidal stability to the nanoparticles in aqueous solutions (e.g., phosphate buffered saline (PBS) and bacteria cell culture media M9), a polymer bearing hydrophilic and amine pendant groups, P(OEGMA)-b-P(ABA), is synthesized via reversible addition-fragmentation chain transfer (RAFT) polymerization and is subsequently grafted onto the PDA-coated IONPs by employing the Schiff base/Michael addition reaction between o-quinone and a primary amine. These nanoparticles are able to effectively disperse Pseudomonas aeruginosa biofilms (up to 79% dispersal) at submicromolar NO concentrations. In addition, the nanoparticles demonstrate excellent bactericidal activity toward P. aeruginosa planktonic and biofilm cells (up to 5-log10 reduction).


Biofilms/drug effects , Coated Materials, Biocompatible , Drug Carriers , Indoles/chemistry , Nanoparticles/chemistry , Nitric Oxide , Polymers/chemistry , Pseudomonas aeruginosa/physiology , Biofilms/growth & development , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Nitric Oxide/chemistry , Nitric Oxide/pharmacology
14.
Angew Chem Int Ed Engl ; 57(17): 4559-4564, 2018 04 16.
Article En | MEDLINE | ID: mdl-29441657

Synthetic polymers have shown promise in combating multidrug-resistant bacteria. However, the biological effects of sequence control in synthetic antimicrobial polymers are currently not well understood. As such, we investigate the antimicrobial effects of monomer distribution within linear high-order quasi-block copolymers consisting of aminoethyl, phenylethyl, and hydroxyethyl acrylamides made in a one-pot synthesis approach via photoinduced electron transfer-reversible addition-fragmentation chain transfer polymerisation (PET-RAFT). Through different combinations of monomer/polymer block order, antimicrobial and haemolytic activities are tuneable in a manner comparable to antimicrobial peptides.


Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Escherichia coli/drug effects , Polymers/pharmacology , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemical synthesis , Antimicrobial Cationic Peptides/chemistry , Microbial Sensitivity Tests , Molecular Structure , Polymers/chemical synthesis , Polymers/chemistry , Structure-Activity Relationship
15.
J Mater Chem B ; 6(19): 2945-2959, 2018 May 21.
Article En | MEDLINE | ID: mdl-32254331

The nitric oxide (NO) molecule has gained increasing attention in biological applications to combat biofilm-associated bacterial infections. However, limited NO loading, relatively short half-lives of low molecular weight NO donor compounds, and difficulties in targeted delivery of NO have hindered their practical clinical administration. To overcome these drawbacks, the combination of NO and scaffolds based on biocompatible polymers is an effective way towards realizing the practical utility of NO in biomedical applications. In this regard, the present overview highlights the recent developments in NO-releasing polymeric biomaterials for antimicrobial applications, focusing on antibiofilm treatments and the challenges that need to be overcome.

16.
ACS Macro Lett ; 7(5): 592-597, 2018 May 15.
Article En | MEDLINE | ID: mdl-35632937

Bacterial biofilms are often difficult to treat and represent the main cause of chronic and recurrent infections. In this study, we report the synthesis of a novel antimicrobial/antibiofilm polymer that consists of biocompatible oligoethylene glycol, hydrophobic ethylhexyl, cationic primary amine, and nitric oxide (NO)-releasing functional groups. The NO-loaded polymer has dual-action capability as it can release NO which triggers the dispersion of biofilm, whereas the polymer can induce bacteria cell death via membrane wall disruption. By functionalizing the polymers with NO, we observed a synergistic effect in biofilm dispersal, planktonic and biofilm killing activities against Pseudomonas aeruginosa. The NO-loaded polymer results in 80% reduction in biofilm biomass and kills >99.999% of planktonic and biofilm P. aeruginosa cells within 1 h of treatment at a polymer concentration of 64 µg mL-1. To achieve this synergistic effect, it is imperative that the NO donors and antimicrobial polymer exist as a single chemical entity, instead of a cocktail physical mixture of two individual components. The excellent antimicrobial/antibiofilm activity of this dual-action polymer suggests the advantages of combination therapy in combating bacterial biofilms.

17.
Bioconjug Chem ; 28(7): 1859-1866, 2017 07 19.
Article En | MEDLINE | ID: mdl-28548819

Particle-cell interactions are governed by, among other factors, the composition and surface properties of the particles. Herein, we report the preparation of various polymer capsules with different compositions and properties via atom transfer radical polymerization mediated continuous assembly of polymers (CAPATRP), where the cellular interactions of these capsules, particularly fouling and specific targeting, are examined by flow cytometry and deconvolution microscopy. Acrylated eight-arm poly(ethylene glycol) (8-PEG) and poly(N-(2-hydroxypropyl)-methacrylamide) (PHPMA) as well as methacrylated hyaluronic acid (HA), poly(glutamic acid) (PGA), and poly(methacrylic acid) (PMA) are used as macro-cross-linkers to obtain a range of polymer capsules with different compositions (PEG, PHPMA, HA, PGA, and PMA). Capsules composed of low-fouling polymers, PEG and PHPMA, show negligible association with macrophage Raw 264.7, monocyte THP-1, and HeLa cells. HA capsules, although moderately low-fouling (<22%) to HeLa, BT474, Raw 264.7, and THP-1 cells, exhibit high targeting specificity to CD44-over-expressing MDA-MB-231 cells. In contrast, PGA and PMA capsules show high cellular association toward phagocytic Raw 264.7 and THP-1 cells. These findings demonstrate the capability of the CAPATRP technique in preparing polymer capsules with specific cellular interactions.


Cell Membrane/metabolism , Polymers/chemistry , Animals , Capsules/chemical synthesis , Capsules/chemistry , Capsules/metabolism , Cross-Linking Reagents/chemistry , Flow Cytometry , HeLa Cells , Humans , Mice , Microscopy , Polymers/chemical synthesis , Polymers/metabolism , RAW 264.7 Cells , Surface Properties
18.
ACS Infect Dis ; 3(3): 237-248, 2017 03 10.
Article En | MEDLINE | ID: mdl-28135798

Infections caused by multidrug-resistant bacteria are on the rise and, therefore, new antimicrobial agents are required to prevent the onset of a postantibiotic era. In this study, we develop new antimicrobial compounds in the form of single-chain polymeric nanoparticles (SCPNs) that exhibit excellent antimicrobial activity against Gram-negative bacteria (e.g., Pseudomonas aeruginosa) at micromolar concentrations (e.g., 1.4 µM) and remarkably kill ≥99.99% of both planktonic cells and biofilm within an hour. Linear random copolymers, which comprise oligoethylene glycol (OEG), hydrophobic, and amine groups, undergo self-folding in aqueous systems due to intramolecular hydrophobic interactions to yield these SCPNs. By systematically varying the hydrophobicity of the polymer, we can tune the extent of cell membrane wall disruption, which in turn governs the antimicrobial activity and rate of resistance acquisition in bacteria. We also show that the incorporation of OEG groups into the polymer design is essential in preventing complexation with proteins in biological medium, thereby maintaining the antimicrobial efficacy of the compound even in in vivo mimicking conditions. In comparison to the last-resort antibiotic colistin, our lead agents have a higher therapeutic index (by ca. 2-3 times) and hence better biocompatibility. We believe that the SCPNs developed here have potential for clinical applications and the information pertaining to their structure-activity relationship will be valuable toward the general design of synthetic antimicrobial (macro)molecules.


Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Gram-Negative Bacteria/drug effects , Plankton/drug effects , Polymers/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Cell Line , Drug Design , Drug Resistance, Bacterial , Gram-Negative Bacteria/physiology , Hydrophobic and Hydrophilic Interactions , Microbial Sensitivity Tests , Nanoparticles/chemistry , Polymers/pharmacology , Rats , Structure-Activity Relationship
19.
ACS Appl Mater Interfaces ; 8(49): 33446-33456, 2016 Dec 14.
Article En | MEDLINE | ID: mdl-27960388

'Structurally nanoengineered antimicrobial peptide polymers' (SNAPPs), in the form of star-shaped peptide polymer nanoparticles, have been recently demonstrated as a new class of antimicrobial agents with superior in vitro and in vivo efficacy against Gram-negative pathogens, including multidrug-resistant species. Herein, we present a detailed bionano interaction study on SNAPPs by assessing their antimicrobial activities against several Gram-negative bacteria in complex biological matrices. Simulated body fluid and animal serum were used as test media to reveal factors that influence the antimicrobial efficacy of SNAPPs. With the exception of Acinetobacter baumannii, the presence of divalent cations at physiological concentrations reduced the antimicrobial efficacy of SNAPPs from minimum inhibitory concentrations (MICs) within the nanomolar range (40-300 nM) against Escherichia coli, Pseudomanas aeruginosa, and Klebsiella pneumoniae to 0.6-4.7 µM. By using E. coli as a representative bacterial species, we demonstrated that the reduction in activity was due to a decrease in the ability of SNAPPs to cause outer and inner membrane disruption. This effect could be reversed through coadministration with a chelating agent. Interestingly, the potency of SNAPPs against A. baumannii was retained even under high salt concentrations. The presence of serum proteins was also found to affect the interaction of SNAPPs with bacterial membranes, possibly through intermolecular binding. Collectively, this study highlights the need to consider the possible interactions of (bio)molecules present in vivo with any new antimicrobial agent under development. We also demonstrate that outer membrane disruption/destabilization is an important but hitherto under-recognized target for the antimicrobial action of peptide-based agents, such as antimicrobial peptides (AMPs). Overall, the findings presented herein could aid in the design of more efficient peptide-based antimicrobial agents with uncompromised potency even under physiological conditions.


Nanoparticles , Animals , Anti-Bacterial Agents , Antimicrobial Cationic Peptides , Escherichia coli , Microbial Sensitivity Tests , Polymers
20.
Nat Microbiol ; 1(11): 16162, 2016 Sep 12.
Article En | MEDLINE | ID: mdl-27617798

With the recent emergence of reports on resistant Gram-negative 'superbugs', infections caused by multidrug-resistant (MDR) Gram-negative bacteria have been named as one of the most urgent global health threats due to the lack of effective and biocompatible drugs. Here, we show that a class of antimicrobial agents, termed 'structurally nanoengineered antimicrobial peptide polymers' (SNAPPs) exhibit sub-µM activity against all Gram-negative bacteria tested, including ESKAPE and colistin-resistant and MDR (CMDR) pathogens, while demonstrating low toxicity. SNAPPs are highly effective in combating CMDR Acinetobacter baumannii infections in vivo, the first example of a synthetic antimicrobial polymer with CMDR Gram-negative pathogen efficacy. Furthermore, we did not observe any resistance acquisition by A. baumannii (including the CMDR strain) to SNAPPs. Comprehensive analyses using a range of microscopy and (bio)assay techniques revealed that the antimicrobial activity of SNAPPs proceeds via a multimodal mechanism of bacterial cell death by outer membrane destabilization, unregulated ion movement across the cytoplasmic membrane and induction of the apoptotic-like death pathway, possibly accounting for why we did not observe resistance to SNAPPs in CMDR bacteria. Overall, SNAPPs show great promise as low-cost and effective antimicrobial agents and may represent a weapon in combating the growing threat of MDR Gram-negative bacteria.


Antimicrobial Cationic Peptides/pharmacology , Drug Resistance, Multiple, Bacterial , Gram-Negative Bacteria/drug effects , Nanoparticles , Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/pathogenicity , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/therapeutic use , Escherichia coli/drug effects , Escherichia coli/pathogenicity , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/microbiology , Mice , Nanoparticles/chemistry , Nanotechnology/economics , Nanotechnology/methods , Peritonitis/drug therapy , Peritonitis/microbiology
...