scholarly journals A Broad-Spectrum Antibiofilm Peptide Enhances Antibiotic Action against Bacterial Biofilms

2014 ◽  
Vol 58 (9) ◽  
pp. 5363-5371 ◽  
Author(s):  
Fany Reffuveille ◽  
César de la Fuente-Núñez ◽  
Sarah Mansour ◽  
Robert E. W. Hancock

ABSTRACTBiofilm-related infections account for at least 65% of all human infections, but there are no available antimicrobials that specifically target biofilms. Their elimination by available treatments is inefficient since biofilm cells are between 10- and 1,000-fold more resistant to conventional antibiotics than planktonic cells. Here we describe the synergistic interactions, with different classes of antibiotics, of a recently characterized antibiofilm peptide, 1018, to potently prevent and eradicate bacterial biofilms formed by multidrug-resistant ESKAPE (Enterococcus faecium,Staphylococcus aureus,Klebsiella pneumoniae,Acinetobacter baumannii,Pseudomonas aeruginosa, andEnterobacterspecies) pathogens. Combinations of peptide 1018 and the antibiotic ceftazidime, ciprofloxacin, imipenem, or tobramycin were synergistic in 50% of assessments and decreased by 2- to 64-fold the concentration of antibiotic required to treat biofilms formed byPseudomonas aeruginosa,Escherichia coli,Acinetobacter baumannii,Klebsiella pneumoniae,Salmonella enterica, and methicillin-resistantStaphylococcus aureus. Furthermore, in flow cell biofilm studies, combinations of low, subinhibitory levels of the peptide (0.8 μg/ml) and ciprofloxacin (40 ng/ml) decreased dispersal and triggered cell death in matureP. aeruginosabiofilms. In addition, short-term treatments with the peptide in combination with ciprofloxacin prevented biofilm formation and reducedP. aeruginosaPA14 preexisting biofilms. PCR studies indicated that the peptide suppressed the expression of various antibiotic targets in biofilm cells. Thus, treatment with the peptide represents a novel strategy to potentiate antibiotic activity against biofilms formed by multidrug-resistant pathogens.

mBio ◽  
2016 ◽  
Vol 7 (2) ◽  
Author(s):  
George G. Zhanel ◽  
James A. Karlowsky

ABSTRACT Clay minerals are naturally occurring layered phyllosilicates which consist of fine particles and possess antimicrobial activity. In a recent article, Behroozian et al. obtained Kisameet clay (KC) from Kisameet, from the central coast of British Columbia, Canada, northwest of Vancouver and assessed its antimicrobial activity versus 16 selected ESKAPE pathogens ( Enterococcus faecium , Staphylococcus aureus , Klebsiella pneumoniae , Acinetobacter baumannii , Pseudomonas aeruginosa , and Enterobacter spp.) possessing a variety of different resistance profiles [S. Behroozian, S. L. Svensson, and J. Davies, mBio 7(1):e01842-15, 2016, http://dx.doi.org/10.1128/mBio.01842-15]. KC demonstrated complete bacterial eradication of Klebsiella pneumoniae , Acinetobacter baumannii , Pseudomonas aeruginosa , and Staphylococcus aureus within 24 h. For Enterobacter spp., the organisms were eradicated with 1% KC within 5 h, while for Enterococcus faecium , it took 48 h to kill all organisms. Although many questions need to be answered, these exciting findings highlight the importance of testing natural substances/products from around the globe to assess whether they possess antimicrobial activity and potential for usage as topical, oral, or systemic agents for the treatment of multidrug-resistant pathogens.


2012 ◽  
Vol 56 (9) ◽  
pp. 4856-4861 ◽  
Author(s):  
Céline Vidaillac ◽  
Lothaire Benichou ◽  
Raphaël E. Duval

ABSTRACTColistin resistance, although uncommon, is increasingly being reported among Gram-negative clinical pathogens, and an understanding of its impact on the activity of antimicrobials is now evolving. We evaluated the potential for synergy of colistin plus trimethoprim, trimethoprim-sulfamethoxazole (1/19 ratio), or vancomycin against 12 isolates ofAcinetobacter baumannii(n= 4),Pseudomonas aeruginosa(n= 4), andKlebsiella pneumoniae(n= 4). The strains included six multidrug-resistant clinical isolates,K. pneumoniaeATCC 700603,A. baumanniiATCC 19606,P. aeruginosaATCC 27853, and their colistin-resistant derivatives (KPm1, ABm1, and PAm1, respectively). Antimicrobial susceptibilities were assessed by broth microdilution and population analysis profiles. The potential for synergy of colistin combinations was evaluated using a checkerboard assay, as well as static time-kill experiments at 0.5× and 0.25× MIC. The MIC ranges of vancomycin, trimethoprim, and trimethoprim-sulfamethoxazole (1/19) were ≥128, 4 to ≥128, and 2/38 to >128/2,432 μg/ml, respectively. Colistin resistance demonstrated little impact on vancomycin, trimethoprim, or trimethoprim-sulfamethoxazole MIC values. Isolates with subpopulations heterogeneously resistant to colistin were observed to various degrees in all tested isolates. In time-kill assays, all tested combinations were synergistic against KPm1 at 0.25× MIC and 0.5× MIC and ABm1 and PAm1 at 0.5× MIC. In contrast, none of the tested combinations demonstrated synergy against any colistin-susceptibleP. aeruginosaisolates and clinical strains ofK. pneumoniaeisolates. Only colistin plus trimethoprim or trimethoprim-sulfamethoxazole was synergistic and bactericidal at 0.5× MIC againstK. pneumoniaeATCC 700603. Colistin resistance seems to promote thein vitroactivity of unconventional colistin combinations. Additional experiments are warranted to understand the clinical significance of these observations.


2020 ◽  
Vol 202 (18) ◽  
Author(s):  
Giulia Orazi ◽  
Fabrice Jean-Pierre ◽  
George A. O’Toole

ABSTRACT The thick mucus within the airways of individuals with cystic fibrosis (CF) promotes frequent respiratory infections that are often polymicrobial. Pseudomonas aeruginosa and Staphylococcus aureus are two of the most prevalent pathogens that cause CF pulmonary infections, and both are among the most common etiologic agents of chronic wound infections. Furthermore, the ability of P. aeruginosa and S. aureus to form biofilms promotes the establishment of chronic infections that are often difficult to eradicate using antimicrobial agents. In this study, we found that multiple LasR-regulated exoproducts of P. aeruginosa, including 2-heptyl-4-hydroxyquinoline N-oxide (HQNO), siderophores, phenazines, and rhamnolipids, likely contribute to the ability of P. aeruginosa PA14 to shift S. aureus Newman norfloxacin susceptibility profiles. Here, we observe that exposure to P. aeruginosa exoproducts leads to an increase in intracellular norfloxacin accumulation by S. aureus. We previously showed that P. aeruginosa supernatant dissipates the S. aureus membrane potential, and furthermore, depletion of the S. aureus proton motive force recapitulates the effect of the P. aeruginosa PA14 supernatant on shifting norfloxacin sensitivity profiles of biofilm-grown S. aureus Newman. From these results, we hypothesize that exposure to P. aeruginosa PA14 exoproducts leads to increased uptake of the drug and/or an impaired ability of S. aureus Newman to efflux norfloxacin. Surprisingly, the effect observed here of P. aeruginosa PA14 exoproducts on S. aureus Newman susceptibility to norfloxacin seemed to be specific to these strains and this antibiotic. Our results illustrate that microbially derived products can alter the ability of antimicrobial agents to kill bacterial biofilms. IMPORTANCE Pseudomonas aeruginosa and Staphylococcus aureus are frequently coisolated from multiple infection sites, including the lungs of individuals with cystic fibrosis (CF) and nonhealing diabetic foot ulcers. Coinfection with P. aeruginosa and S. aureus has been shown to produce worse outcomes compared to infection with either organism alone. Furthermore, the ability of these pathogens to form biofilms enables them to cause persistent infection and withstand antimicrobial therapy. In this study, we found that P. aeruginosa-secreted products dramatically increase the ability of the antibiotic norfloxacin to kill S. aureus biofilms. Understanding how interspecies interactions alter the antibiotic susceptibility of bacterial biofilms may inform treatment decisions and inspire the development of new therapeutic strategies.


2015 ◽  
Vol 59 (5) ◽  
pp. 2583-2587 ◽  
Author(s):  
Robert K. Flamm ◽  
Paul R. Rhomberg ◽  
Nachum Kaplan ◽  
Ronald N. Jones ◽  
David J. Farrell

ABSTRACTStaphylococcus aureusand coagulase-negative staphylococci (CoNS) are responsible for a wide variety of human infections. The investigational antibacterial Debio1450 (previously AFN-1720), a prodrug of Debio1452 (previously AFN-1252), specifically targets staphylococci without significant activity against other Gram-positive or Gram-negative species. Debio1452 inhibits FabI, an enzyme critical to fatty acid biosynthesis in staphylococci. The activity of Debio1452 against CoNS, methicillin-susceptibleS. aureus(MSSA), and methicillin-resistantS. aureus(MRSA), including significant clones, was determined. A globally diverse collection of 574 patient isolates from 35 countries was tested that included CoNS (6 species, 103 strains), MSSA (154 strains), MRSA (163 strains), and molecularly characterized strains (includingspa-typed MRSA clones; 154 strains). The isolates were tested for susceptibility by CLSI broth microdilution methods against Debio1452 and 10 comparators. The susceptibility rates for the comparators were determined using CLSI and EUCAST breakpoint criteria. AllS. aureusand CoNS strains were inhibited by Debio1452 concentrations of ≤0.12 and ≤0.5 μg/ml, respectively. The MIC50s for MSSA, MRSA, and molecularly characterized MRSA strains were 0.004 μg/ml, and the MIC90s ranged from 0.008 to 0.03 μg/ml. The MICs were higher for the CoNS isolates (MIC50/90, 0.015/0.12 μg/ml). AmongS. aureusstrains, resistance was common for erythromycin (61.6%), levofloxacin (49.0%), clindamycin (27.6%), tetracycline (15.7%), and trimethoprim-sulfamethoxazole (7.0%). Debio1452 demonstrated potent activity against MSSA, MRSA, and CoNS. Debio1452 showed significantly greater activity overall (MIC50, 0.004 μg/ml) than the other agents tested against these staphylococcal species, which included dominant MRSA clones and strains resistant to currently utilized antimicrobial agents.


Author(s):  
I. I. Myrko ◽  
T. I. Chaban ◽  
V. V. Ogurtsov ◽  
V. S. Matiychuk

Мета роботи. Здійснити синтез деяких нових піразолзаміщених 7H-[1,2,4]триазоло[3,4-b][1,3,4]тіадіазинів та провести дослідження антимікробних властивостей синтезованих сполук. Матеріали і методи. Органічний синтез, ЯМР-спектроскопія, елементний аналіз, фармакологічний скринінг. Результати й обговорення. У результаті взаємодії eтил (2Z)-хлоро(фенілгідразоно)ацетатів з ацетилацетоном було отримано етил 4-ацетил-5-метил-1-феніл-1H-піразол-3-карбоксилати. Зазначені сполуки піддали бромуванню, що дозволило одержати цільові бромкетони. Синтезовані на даній стадії етил 1-арил-4-(бромацетил)-5-метил-1Н-піразол-3-карбоксилати було введено у взаємодію з 4-аміно-5-арил(гетарил)-2,4-дигідро-3Н-1,2,4-триазол-3-тіонами з подальшим формуванням 1,3,4-тіадіазольного циклу та отриманням відповідних етил 1-арил-4-{3-арил(гетарил)-7H-[1,2,4]триазоло[3,4-b][1,3,4]тіадіазин-6-іл)}-5-метил-1H-піразол-3-карбоксилатів. Структура синтезованих сполук підтверджена даними елементного аналізу та ЯМР спектроскопією. В рамках міжнародного проекту "The Community for Antimicrobial Drug Discovery" (CO-ADD) за підтримки Wellcome Trust (Великобританія) і університету Квінсленда (Австралія) для синтезованих сполук здійснено скринінг антимікробної активності. Як тестові мікроорганізми використовували п'ять штамів бактерій: Escherichia coli ATCC 25922, Klebsiella pneumoniae ATCC 700603, Acinetobacter baumannii ATCC 19606, Pseudomonas aeruginosa ATCC 27853, Staphylococcus aureus ATCC 43300 та двох штамів грибків: Candida albicans ATCC 90028 і Cryptococcus neoformans ATCC 208821. Встановлено, що досліджувані сполуки виявляють різноманітну дію, від практично повної її відсутності до виразного антимікробного ефекту. Висновки. Здійснено синтез 12 нових етил 1-арил-4-{3-арил(гетарил)-7H-[1,2,4]триазоло[3,4-b][1,3,4]тіадіазин-6-іл)}-5-метил-1H-піразол-3-карбоксилатів. Зазначені речовини отримані шляхом взаємодії відповідних етил 1-арил-4-(бромацетил)-5-метил-1Н-піразол-3-карбоксилатів з 4-аміно-5-арил(гетарил)-2,4-дигідро-3Н-1,2,4-триазол-3-тіонами. Дослідження антимікробної активності синтезованих сполук демонструють потенціал пошуку антимікробних агентів серед зазначеного класу сполук.


2015 ◽  
Vol 59 (4) ◽  
pp. 2280-2285 ◽  
Author(s):  
Robert K. Flamm ◽  
Paul R. Rhomberg ◽  
Ronald N. Jones ◽  
David J. Farrell

ABSTRACTRX-P873 is a novel antibiotic from the pyrrolocytosine series which exhibits high binding affinity for the bacterial ribosome and broad-spectrum antibiotic properties. The pyrrolocytosines have shownin vitroactivity against multidrug-resistant Gram-negative and Gram-positive strains of bacteria known to cause complicated urinary tract, skin, and lung infections, as well as sepsis.Enterobacteriaceae(657),Pseudomonas aeruginosa(200), andAcinetobacter baumannii(202) isolates from North America and Europe collected in 2012 as part of a worldwide surveillance program were testedin vitroby broth microdilution using Clinical and Laboratory Standards Institute (CLSI) methodology. RX-P873 (MIC90, 0.5 μg/ml) was >32-fold more active than ceftazidime and inhibited 97.1% and 99.5% ofEnterobacteriaceaeisolates at MIC values of ≤1 and ≤4 μg/ml, respectively. There were only three isolates with an MIC value of >4 μg/ml (all were indole-positiveProtea). RX-P873 (MIC50/90, 2/4 μg/ml) was highly active againstPseudomonas aeruginosaisolates, including isolates which were nonsusceptible to ceftazidime or meropenem. RX-P873 was 2-fold less active againstP. aeruginosathan tobramycin (MIC90, 2 μg/ml; 91.0% susceptible) and colistin (MIC90, 2 μg/ml; 99.5% susceptible) and 2-fold more potent than amikacin (MIC90, 8 μg/ml; 93.5% susceptible) and meropenem (MIC90, 8 μg/ml; 76.0% susceptible). RX-P873, the most active agent againstAcinetobacter baumannii(MIC90, 1 μg/ml), was 2-fold more active than colistin (MIC90, 2 μg/ml; 97.0% susceptible) and 4-fold more active than tigecycline (MIC90, 4 μg/ml). This novel agent merits further exploration of its potential against multidrug-resistant Gram-negative bacteria.


2020 ◽  
Author(s):  
M. R. Tuttobene ◽  
J. F. Pérez ◽  
E. Pavesi ◽  
B. Perez Mora ◽  
D. Biancotti ◽  
...  

Light sensing has been extensively characterized in the human pathogen Acinetobacter baumannii at environmental temperatures. However, the influence of light on the physiology and pathogenicity of human bacterial pathogens at temperatures found in warm-blooded hosts is still poorly understand. In this work, we show that ESKAPE priority pathogens, such as Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter spp., which have been recognized by the WHO and the CDC as critical, can also sense and respond to light at temperatures found in human hosts. Most interestingly, in these pathogens light modulates important pathogenicity determinants as well as virulence in an epithelial infection model, which could have implications in human infections. In fact, we found that alpha-toxin-dependent hemolysis, motility and growth under iron deprived conditions are modulated by light in S. aureus. Light also regulates persistence, metabolism and the ability to kill competitors, in some of these microorganisms. Finally, light exerts a profound effect on the virulence of these pathogens in an epithelial infection model, though the response is not the same in the different species: virulence was enhanced by light in A. baumannii and S. aureus, while in A. nosocomialis and P. aeruginosa it was reduced. Neither the BlsA photoreceptor nor the type VI secretion system (T6SS) are involved in virulence modulation by light in A. baumannii. Overall, this fundamental knowledge highlights the potential use of light to control pathogen's virulence, either directly or by manipulating the light regulatory switch toward the lowest virulence/persistence configuration. IMPORTANCE Pathogenic bacteria are microorganisms capable of producing disease. Dangerous bacterial pathogens such as Staphylococcus aureus, Pseudomonas aeruginosa and Acinetobacter baumannii are responsible for serious intrahospital and community infections in humans. Therapeutics is often complicated due to resistance to multiple antibiotics, rendering them ineffective. In this work, we show that these pathogens sense natural light and respond to it by modulating aspects related to their ability to cause disease: in the presence of light some of them become more aggressive while others show an opposite response. Overall, we provide new understanding on the behavior of these pathogens, which could contribute to control infections caused by them. Since the response is distributed in diverse pathogens, this notion could prove a general concept.


2014 ◽  
Vol 58 (7) ◽  
pp. 4113-4122 ◽  
Author(s):  
Mohamed F. Mohamed ◽  
Maha I. Hamed ◽  
Alyssa Panitch ◽  
Mohamed N. Seleem

ABSTRACTThe seriousness of microbial resistance combined with the lack of new antimicrobials has increased interest in the development of antimicrobial peptides (AMPs) as novel therapeutics. In this study, we evaluated the antimicrobial activities of two short synthetic peptides, namely, RRIKA and RR. These peptides exhibited potent antimicrobial activity againstStaphylococcus aureus, and their antimicrobial effects were significantly enhanced by addition of three amino acids in the C terminus, which consequently increased the amphipathicity, hydrophobicity, and net charge. Moreover, RRIKA and RR demonstrated a significant and rapid bactericidal effect against clinical and drug-resistantStaphylococcusisolates, including methicillin-resistantStaphylococcus aureus(MRSA), vancomycin-intermediateS. aureus(VISA), vancomycin-resistantS. aureus(VRSA), linezolid-resistantS. aureus, and methicillin-resistantStaphylococcus epidermidis. In contrast to many natural AMPs, RRIKA and RR retained their activity in the presence of physiological concentrations of NaCl and MgCl2. Both RRIKA and RR enhanced the killing of lysostaphin more than 1,000-fold and eradicated MRSA and VRSA isolates within 20 min. Furthermore, the peptides presented were superior in reducing adherent biofilms ofS. aureusandS. epidermidiscompared to results with conventional antibiotics. Our findings indicate that the staphylocidal effects of our peptides were through permeabilization of the bacterial membrane, leading to leakage of cytoplasmic contents and cell death. Furthermore, peptides were not toxic to HeLa cells at 4- to 8-fold their antimicrobial concentrations. The potent and salt-insensitive antimicrobial activities of these peptides present an attractive therapeutic candidate for treatment of multidrug-resistantS. aureusinfections.


2020 ◽  
Vol 3 (2) ◽  
pp. 96
Author(s):  
Isna Romadhona ◽  
Fauna Herawati ◽  
Rika Yulia

Antibiotik merupakan obat yang digunakan untuk mengatasi dan mencegah infeksi bakteri. Penggunaan antibiotik yang tidak tepat dapat menimbulkan berbagai masalah, diantaranya pengobatan akan lebih mahal dan juga risiko terjadinya resistensi bakteri terhadap antibiotik. Penelitian ini bertujuan untuk mengetahui profil penggunaan antibiotik dan profil peta kuman pada pasien gangren diabetes melitus di sebuah RSUD di Kabupaten Gresik serta untuk mengetahui kesesuaian penggunaan antibiotik dengan mengacu pada Permenkes Republik Indonesia No. 2406/Menkes/PER/XII/2011. Data penggunaan antibiotik diperoleh dari catatan Rekam Medis pada periode Januari – November 2017. Data penggunaan antibiotik dihitung dengan menggunakan rumus DDD/100 pasien-hari rawat. Hasil perhitungan DDD/100 pasien-hari rawat menunjukkan hasil sebesar 470,11 DDD/100 pasien-hari rawat. Peta kuman pada pasien gangren, melaporkan adanya bakteri Enterobacter cloacae 24%, Escherichia coli 18%, Staphylococcus aureus 15%, Acinetobacter baumannii 9%, Pseudomonas aeruginosa 6%, Citrobacter youngae 6%, Enterobacter aerogenes 6%, Proteus vulgaris 6%, Staphylococcus schleiferi 6%, Klebsiella pneumoniae 3%, dan Proteus mirabilis 3% . Penggunaan antibiotik seftriakson dan metronidazol pada pasien gangren diabetes melitus di sebuah RSUD di Kabupaten Gresik pada periode Januari – November 2017 telah sesuai dengan pedoman penggunaan antibiotik berdasarkan Permenkes Republik Indonesia No. 2406/Menkes/PER/ XII/2011, yaitu antibiotik golongan sefalosporin generasi III yang lebih aktif terhadap Enterobacteriaceae dan antibiotik golongan nitroimidazol yang dapat mengobati infeksi bakteri basil anerob Gram-Negatif.


Sign in / Sign up

Export Citation Format

Share Document