1
|
Zhu L, Zhang H, Xiao X, Sun S, Tong Y, Zhuang S, Sheng Z, Fan Y, Ma W, Liu Y, Qing S, Zhang W. Shuanghuanglian volatile oil exerts antipyretic, anti-inflammatory, and antibacterial synergistic effects through multiple pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118795. [PMID: 39278293 DOI: 10.1016/j.jep.2024.118795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese Medicine (TCM) has a rich history spanning 2000 years. Shuanghuanglian, a traditional Chinese herbal formula composed of three botanicals, is primarily used to treat colds, respiratory infections (including bacterial pneumonia), and pharyngitis. Previous research has found that the volatile oil of Shuanghuanglian is crucial for its efficacy. However, there is a lack of studies investigating its mechanisms. AIM OF THE STUDY This study aims to explore the antibacterial and anti-inflammatory mechanisms of Shuanghuanglian volatile oil and its potential to enhance the antibacterial effects when used in conjunction with antibiotics. METHODS Determination of the GC-MS fingerprint of SVO using Gas Chromatography-Mass Spectrometry (GC-MS), The antibacterial effects of SVO on multidrug-resistant Klebsiella pneumoniae (MDR-KP) were assessed by detecting MIC, checkerboard method assay, time-kill curves, resistance growth curves, transcriptome sequencing analysis, scanning electron microscopy(SEM), purification, and quantitative analysis of extracellular polysaccharides(EPS). In vivo part, an MDR-KP induced mouse pneumonia model was established to evaluate the mitigating effects of SVO on mouse pneumonia, using comprehensive network pharmacology and bioinformatics to identify genes related to bacterial pneumonia and potential targets of SVO. Validation was performed through molecular docking, qPCR, and ELISA tests. RESULTS SVO modulates the expression of MDR-KP mRNA for wecB, wecC, murA, murD, murE, murF, inhibiting the synthesis of O-antigen polysaccharides and peptidoglycans, thereby compromising bacterial cell wall integrity and affecting the synthesis of biofilms. These actions not only exhibit antibacterial effects but also enhance antibacterial activity, restoring the sensitivity of CEF to MDR-KP. SVO suppresses the biological activity of PTGS2, reducing the production of Prostaglandin E2 (PGE2), thereby exerting antipyretic and anti-inflammatory effects, providing new insights for the development of natural non-steroidal anti-inflammatory drugs (NSAIDs). CONCLUSIONS Our research indicates that SVO exerts antipyretic, anti-inflammatory, and antibacterial synergistic effects through multiple pathways.
Collapse
Affiliation(s)
- Leixin Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Hanwen Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Xinglan Xiao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Shaoqiang Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China
| | - Yinchao Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Shen Zhuang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Zhenwei Sheng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Yunpeng Fan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Wuren Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Yingqiu Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China
| | - Suzhu Qing
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China.
| | - Weimin Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, PR China; Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling, 712100, PR China.
| |
Collapse
|
2
|
Polyudova T, Lemkina L, Eroshenko D, Esaev A. Suppression of planktonic and biofilm of Escherichia coli by the synergistic lantibiotics-polymyxins combinations. Arch Microbiol 2024; 206:191. [PMID: 38520490 DOI: 10.1007/s00203-024-03922-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/28/2024] [Accepted: 03/02/2024] [Indexed: 03/25/2024]
Abstract
Escherichia coli are generally resistant to the lantibiotic's action (nisin and warnerin), but we have shown increased sensitivity of E. coli to lantibiotics in the presence of subinhibitory concentrations of polymyxins. Synergistic lantibiotic-polymyxin combinations were found for polymyxins B and M. The killing of cells at the planktonic and biofilm levels was observed for two collection and four clinical multidrug-resistant E. coli strains after treatment with lantibiotic-polymyxin B combinations. Thus, 24-h treatment of E. coli mature biofilms with warnerin-polymyxin B or nisin-polymyxin B leads to five to tenfold decrease in the number of viable cells, depending on the strain. AFM revealed that the warnerin and polymyxin B combination caused the loss of the structural integrity of biofilm and the destruction of cells within the biofilm. It has been shown that pretreatment of cells with polymyxin B leads to an increase of Ca2+ and Mg2+ ions in the culture medium, as detected by atomic absorption spectroscopy. The subsequent exposure to warnerin caused cell death with the loss of K+ ions and cell destruction with DNA and protein release. Thus, polymyxins display synergy with lantibiotics against planktonic and biofilm cells of E. coli, and can be used to overcome the resistance of Gram-negative bacteria to lantibiotics.
Collapse
Affiliation(s)
- Tatyana Polyudova
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Scientific Centre, Ural Branch, Russian Academy of Sciences, Perm, Russia.
| | - Larisa Lemkina
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Scientific Centre, Ural Branch, Russian Academy of Sciences, Perm, Russia
| | - Daria Eroshenko
- Institute of Technical Chemistry, Perm Federal Scientific Centre, Ural Branch, Russian Academy of Science, Perm, Russia
| | - Artem Esaev
- Institute of Ecology and Genetics of Microorganisms, Perm Federal Scientific Centre, Ural Branch, Russian Academy of Sciences, Perm, Russia
| |
Collapse
|
3
|
Baindara P, Mandal SM. Gut-Antimicrobial Peptides: Synergistic Co-Evolution with Antibiotics to Combat Multi-Antibiotic Resistance. Antibiotics (Basel) 2023; 12:1732. [PMID: 38136766 PMCID: PMC10740742 DOI: 10.3390/antibiotics12121732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Due to huge diversity and dynamic competition, the human gut microbiome produces a diverse array of antimicrobial peptides (AMPs) that play an important role in human health. The gut microbiome has an important role in maintaining gut homeostasis by the AMPs and by interacting with other human organs via established connections such as the gut-lung, and gut-brain axis. Additionally, gut AMPs play a synergistic role with other gut microbiota and antimicrobials to maintain gut homeostasis by fighting against multi-antibiotic resistance (MAR) bacteria. Further, conventional antibiotics intake creates a synergistic evolutionary pressure for gut AMPs, where antibiotics and gut AMPs fight synergistically against MAR. Overall, gut AMPs are evolving under a complex and highly synergistic co-evolutionary pressure created by the various interactions between gut microbiota, gut AMPs, and antibiotics; however, the complete mechanism is not well understood. The current review explores the synergistic action of gut AMPs and antibiotics along with possibilities to fight against MAR bacteria.
Collapse
Affiliation(s)
- Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Santi M. Mandal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| |
Collapse
|
4
|
R PA, Anbarasu A. Antimicrobial Peptides as Immunomodulators and Antimycobacterial Agents to Combat Mycobacterium tuberculosis: a Critical Review. Probiotics Antimicrob Proteins 2023; 15:1539-1566. [PMID: 36576687 DOI: 10.1007/s12602-022-10018-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 12/29/2022]
Abstract
Tuberculosis (TB) is a devastating disease foisting a significantly high morbidity, prepotent in low- and middle-income developing countries. Evolution of drug resistance among Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, has made the TB treatment more complicated. The protracted nature of present TB treatment, persistent and tolerant Mtb populations, interaction with antiretroviral therapy and existing toxicity concerned with conventional anti-TB drugs are the four major challenges inflicted with emergence of drug-resistant mycobacterial strains, and the standard medications are unable to combat these strains. These factors emphasize an exigency to develop new drugs to overcome these barriers in current TB therapy. With this regard, antimycobacterial peptides derived from various sources such as human cells, bacterial sources, mycobacteriophages, fungal, plant and animal sources could be considered as antituberculosis leads as most of these peptides are associated with dual advantages of having both bactericidal activity towards Mtb as well as immuno-regulatory property. Some of the peptides possess the additional advantage of interacting synergistically with antituberculosis medications too, thereby increasing their efficiency, underscoring the vigour of antimicrobial peptides (AMPs) as best possible alternative therapeutic candidates or adjuvants in TB treatment. Albeit the beneficiary features of these peptides, few obstacles allied with them like cytotoxicity and proteolytic degradation are matter of concerns too. In this review, we have focused on structural hallmarks, targeting mechanisms and specific structural aspects contributing to antimycobacterial activity and discovered natural and synthetic antimycobacterial peptides along with their sources, anti-TB, immuno-regulatory properties, merits and demerits and possible delivery methods of AMPs.
Collapse
Affiliation(s)
- Preethi A R
- Medical & Biological Computing Laboratory, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore-632014, India
- Department of Biotechnology, SBST, VIT, Vellore-632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical & Biological Computing Laboratory, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore-632014, India.
- Department of Biotechnology, SBST, VIT, Vellore-632014, Tamil Nadu, India.
| |
Collapse
|
5
|
Lennard PR, Hiemstra PS, Nibbering PH. Complementary Activities of Host Defence Peptides and Antibiotics in Combating Antimicrobial Resistant Bacteria. Antibiotics (Basel) 2023; 12:1518. [PMID: 37887219 PMCID: PMC10604037 DOI: 10.3390/antibiotics12101518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/27/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
Due to their ability to eliminate antimicrobial resistant (AMR) bacteria and to modulate the immune response, host defence peptides (HDPs) hold great promise for the clinical treatment of bacterial infections. Whereas monotherapy with HDPs is not likely to become an effective first-line treatment, combinations of such peptides with antibiotics can potentially provide a path to future therapies for AMR infections. Therefore, we critically reviewed the recent literature regarding the antibacterial activity of combinations of HDPs and antibiotics against AMR bacteria and the approaches taken in these studies. Of the 86 studies compiled, 56 featured a formal assessment of synergy between agents. Of the combinations assessed, synergistic and additive interactions between HDPs and antibiotics amounted to 84.9% of the records, while indifferent and antagonistic interactions accounted for 15.1%. Penicillin, aminoglycoside, fluoro/quinolone, and glycopeptide antibiotic classes were the most frequently documented as interacting with HDPs, and Staphylococcus aureus, Pseudomonas aeruginosa, Escherichia coli, and Enterococcus faecium were the most reported bacterial species. Few studies formally evaluated the effects of combinations of HDPs and antibiotics on bacteria, and even fewer assessed such combinations against bacteria within biofilms, in animal models, or in advanced tissue infection models. Despite the biases of the current literature, the studies suggest that effective combinations of HDPs and antibiotics hold promise for the future treatment of infections caused by AMR bacteria.
Collapse
Affiliation(s)
- Patrick R. Lennard
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
- Institute of Immunology and Infection, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FE, UK
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Centre, Leiden 2333, The Netherlands
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden 2333, The Netherlands;
| | - Pieter S. Hiemstra
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Centre, Leiden 2333, The Netherlands
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden 2333, The Netherlands;
| |
Collapse
|
6
|
Tong YC, Li PC, Yang Y, Lin QY, Liu JT, Gao YN, Zhang YN, Jin S, Qing SZ, Xing FS, Fan YP, Liu YQ, Wang WL, Zhang WM, Ma WR. Detection of Antibiotic Resistance in Feline-Origin ESBL Escherichia coli from Different Areas of China and the Resistance Elimination of Garlic Oil to Cefquinome on ESBL E. coli. Int J Mol Sci 2023; 24:ijms24119627. [PMID: 37298578 DOI: 10.3390/ijms24119627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
The development of drug-resistance in the opportunistic pathogen Escherichia coli has become a global public health concern. Due to the share of similar flora between pets and their owners, the detection of pet-origin antibiotic-resistant E. coli is necessary. This study aimed to detect the prevalence of feline-origin ESBL E. coli in China and to explore the resistance elimination effect of garlic oil to cefquinome on ESBL E. coli. Cat fecal samples were collected from animal hospitals. The E. coli isolates were separated and purified by indicator media and polymerase chain reaction (PCR). ESBL genes were detected by PCR and Sanger sequencing. The MICs were determined. The synergistic effect of garlic oil and cefquinome against ESBL E. coli was investigated by checkerboard assays, time-kill and growth curves, drug-resistance curves, PI and NPN staining, and a scanning electronic microscope. A total of 80 E. coli strains were isolated from 101 fecal samples. The rate of ESBL E. coli was 52.5% (42/80). The prevailing ESBL genotypes in China were CTX-M-1, CTX-M-14, and TEM-116. In ESBL E. coli, garlic oil increased the susceptibility to cefquinome with FICIs from 0.2 to 0.7 and enhanced the killing effect of cefquinome with membrane destruction. Resistance to cefquinome decreased with treatment of garlic oil after 15 generations. Our study indicates that ESBL E. coli has been detected in cats kept as pets. The sensitivity of ESBL E. coli to cefquinome was enhanced by garlic oil, indicating that garlic oil may be a potential antibiotic enhancer.
Collapse
Affiliation(s)
- Yin-Chao Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Peng-Cheng Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yang Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Qing-Yi Lin
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jin-Tong Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yi-Nuo Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yi-Ning Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Shuo Jin
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Su-Zhu Qing
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Fu-Shan Xing
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yun-Peng Fan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
- Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Ying-Qiu Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
- Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Wei-Ling Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, China
| | - Wei-Min Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
- Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Wu-Ren Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
- Institute of Traditional Chinese Veterinary Medicine, Northwest A&F University, Yangling 712100, China
- Xi'an Veterinary Teaching Hospital, Northwest A&F University, Xi'an 710065, China
| |
Collapse
|
7
|
Tong YC, Zhang YN, Li PC, Cao YL, Ding DZ, Yang Y, Lin QY, Gao YN, Sun SQ, Fan YP, Liu YQ, Qing SZ, Ma WR, Zhang WM. Detection of antibiotic-resistant canine origin Escherichia coli and the synergistic effect of magnolol in reducing the resistance of multidrug-resistant Escherichia coli. Front Vet Sci 2023; 10:1104812. [PMID: 37008355 PMCID: PMC10057116 DOI: 10.3389/fvets.2023.1104812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/21/2023] [Indexed: 03/17/2023] Open
Abstract
BackgroundThe development of antimicrobial resistance in the opportunistic pathogen Escherichia coli has become a global public health concern. Due to daily close contact, dogs kept as pets share the same E. coli with their owners. Therefore, the detection of antimicrobial resistance in canine E. coli is important, as the results could provide guidance for the future use of antibiotics. This study aimed to detect the prevalence of antibiotic-resistance of canine origin E. coli in Shaanxi province and to explore the inhibition effect of magnolol combined with cefquinome on MDR E. coli, so as to provide evidence for the use of antibiotics.MethodsCanine fecal samples were collected from animal hospitals. The E. coli isolates were separated and purified using various indicator media and polymerase chain reaction (PCR). Drug-resistance genes [aacC2, ant(3')-I, aph(3')-II, aac(6')-Ib-cr, aac(3')-IIe, blaKPC, blaIMP−4, blaOXA, blaCMY, blaTEM−1, blaSHV, blaCTX−M−1, blaCTX−M−9, Qnra, Qnrb, Qnrs, TetA, TetB, TetM, Ermb] were also detected by PCR. The minimum inhibitory concentration (MIC) was determined for 10 antibiotics using the broth-microdilution method. Synergistic activity of magnolol and cefquinome against multidrug-resistant (MDR) E. coli strains was investigated using checkerboard assays, time-kill curves, and drug-resistance curves.ResultsA total of 101 E. coli strains were isolated from 158 fecal samples collected from animal hospitals. MIC determinations showed that 75.25% (76/101) of the E. coli strains were MDR. A total of 22 drug-resistance genes were detected among the 101 strains. The blaTEM−1gene exhibited the highest detection rate (89.77%). The TetA and Sul gene also exhibited high detection rate (66.34 and 53.47%, respectively). Carbapenem-resistant E. coli strains were found in Shangluo and Yan'an. Additionally, in MDR E. coli initially resistant to cefquinome, magnolol increased the susceptibility to cefquinome, with an FICI (Fractional Inhibitory Concentration Index) between 0.125 and 0.5, indicating stable synergy. Furthermore, magnolol enhanced the killing effect of cefquinome against MDR E. coli. Resistance of MDR E. coli to cefquinome decreased markedly after treatment with magnolol for 15 generations.ConclusionOur study indicates that antibiotic-resistance E. coli has been found in domestic dogs. After treatment with magnolol extracted from the Chinese herb Houpo (Magnolia officinalis), the sensitivity of MDR E. coli to cefquinome was enhanced, indicating that magnolol reverses the resistance of MDR E. coli. The results of this study thus provide reference for the control of E. coli resistance.
Collapse
Affiliation(s)
- Yin-Chao Tong
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yi-Ning Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Peng-Cheng Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ya-Li Cao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Dong-Zhao Ding
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yang Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qing-Yi Lin
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Yi-Nuo Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Shao-Qiang Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yun-Peng Fan
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ying-Qiu Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Su-Zhu Qing
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wu-Ren Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- Xi'an Veterinary Teaching Hospital, Northwest A&F University, Xi'an, China
- Wu-Ren Ma
| | - Wei-Min Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
- *Correspondence: Wei-Min Zhang
| |
Collapse
|
8
|
Synergy between Human Peptide LL-37 and Polymyxin B against Planktonic and Biofilm Cells of Escherichia coli and Pseudomonas aeruginosa. Antibiotics (Basel) 2023; 12:antibiotics12020389. [PMID: 36830299 PMCID: PMC9952724 DOI: 10.3390/antibiotics12020389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/01/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The rise in antimicrobial resistant bacteria is limiting the number of effective treatments for bacterial infections. Escherichia coli and Pseudomonas aeruginosa are two of the pathogens with the highest prevalence of resistance, and with the greatest need for new antimicrobial agents. Combinations of antimicrobial peptides (AMPs) and antibiotics that display synergistic effects have been shown to be an effective strategy in the development of novel therapeutic agents. In this study, we investigated the synergy between the AMP LL-37 and various classes of antibiotics against E. coli and P. aeruginosa strains. Of the six antibiotics tested (ampicillin, tetracycline, ciprofloxacin, gentamicin, aztreonam, and polymyxin B (PMB)), LL-37 displayed the strongest synergy against E. coli MG1655 and P. aeruginosa PAO1 laboratory strains when combined with PMB. Given the strong synergy, the PMB + LL-37 combination was chosen for further examination where it demonstrated synergy against multidrug-resistant and clinical E. coli isolates. Synergy of PMB + LL-37 towards clinical isolates of P. aeruginosa varied and showed synergistic, additive, or indifferent effects. The PMB + LL-37 combination treatment showed significant prevention of biofilm formation as well as eradication of pre-grown E. coli and P. aeruginosa biofilms. Using the Galleria mellonella wax worm model, we showed that the PMB + LL-37 combination treatment retained its antibacterial capacities in vivo. Flow analyses were performed to characterize the mode of action. The results of the present study provide proof of principle for the synergistic response between LL-37 and PMB and give novel insights into a promising new antimicrobial combination against gram-negative planktonic and biofilm cells.
Collapse
|
9
|
Biswas S. Synthesis of a Novel Lantibiotic Using Mutacin II Biosynthesis Apparatus. Microbiol Spectr 2023; 11:e0303022. [PMID: 36645288 PMCID: PMC9927145 DOI: 10.1128/spectrum.03030-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/06/2022] [Indexed: 01/17/2023] Open
Abstract
Owing to extensive metagenomic studies, we now have access to numerous sequences of novel bacteriocin-like antimicrobial peptides encoded by various cultivable and noncultivable bacteria. However, relatively rarely, we even have access to these cultivable strains to examine the potency and the targets of the predicted bacteriocins. In this study, we evaluated a heterologous biosynthetic system to produce biologically active nonnative novel lantibiotics, which are modified bacteriocins. We chose Streptococcus mutans, a dental pathogen, as the host organism because it is genetically easy to manipulate and is inherently a prolific producer of various bacteriocins. We chose the S. mutans T8 strain as the host, which produces the lantibiotic mutacin II, to express 10 selected homologs of mutacin II identified from GenBank. These lantibiotic peptides either are novel or have been studied very minimally. The core regions of the selected lantibiotic peptides were fused to the leader sequence of the mutacin II peptide and integrated into the chromosome such that the core region of the native mutacin II was replaced with the new core sequences. By this approach, using the mutacin II biosynthesis machinery, we obtained one bioactive novel lantibiotic peptide with 52% different residues compared to the mutacin II core region. This unknown lantibiotic is encoded by Streptococcus agalactiae and Streptococcus ovuberis strains. Since this peptide displays some homology with nukacin ISK-1, we named it nukacin Spp. 2. This study demonstrated that the mutacin II biosynthesis machinery can be successfully used as an efficient system for the production of biologically active novel lantibiotics. IMPORTANCE In this study, we report for the first time that Streptococcus mutans can be used as a host to produce various nonnative lantibiotics. We showed that in the T8 strain, we could produce bioactive lacticin 481 and nukacin ISK-1, both of which are homologs of mutacin II, using T8's modification and secretion apparatus. Similarly, we also synthesized a novel bioactive lantibiotic, which we named nukacin Spp. 2.
Collapse
Affiliation(s)
- Saswati Biswas
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
10
|
Zuo J, Liu L, Hou S, Liu X, Teng J, Li P, Liu X. Antimicrobial and antibiofilm activity of isoorientin against carbapenem non-sensitive Escherichia coli from raw milk of goats. J Anim Sci 2023; 101:skad047. [PMID: 36762933 PMCID: PMC9985329 DOI: 10.1093/jas/skad047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/09/2023] [Indexed: 02/11/2023] Open
Abstract
Carbapenemase-producing E. coli is a grave public health concern as the potential emergence of resistant strains and their transmission. Isoorientin belongs to a potential antimicrobial flavonoid compound existing in several plants, while the research on the antimicrobial activity of isoorientin is limited thus far. We evaluated the antimicrobial and antibiofilm effects of isoorientin against biofilm-forming carbapenem non-sensitive Escherichia coli (E. coli) from raw milk of goats, and explored its molecular mechanisms. Isoorientin showed obvious antimicrobial ability with the minimum inhibitory concentration (MIC), and it exhibited synergistic activity with traditional antimicrobials against the carbapenem non-sensitive E. coli. Isoorientin could also significantly inhibit the carbapenem non-sensitive E. coli biofilm formation and destroy the established biofilms, with the percentage of inhibition ranging from 27.8% to 75% at MIC, and the corresponding percentage of eradication ranging from 15.3% to 61.6%, respectively. Confocal laser scanning microscopy (CLSM) observation and scanning electron microscopy (SEM) images indicated that the E. coli biofilm reduced in thickness with increasing concentrations of isoorientin. Dose-dependent decrease in eDNA revealed that isoorientin interacted with the extracellular polymeric substances (EPS) of the biofilm. qRT-PCR assay for the biofilm-forming associated genes further confirmed the above results. Overall, these results concluded that the isoorientin has significant antimicrobial and antibiofilm activity against carbapenem non-sensitive E. coli, and has potential application in prevention of food contamination and spoilage.
Collapse
Affiliation(s)
- Jingru Zuo
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lianjie Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Simeng Hou
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaoqiang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Jiang Teng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Pei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xiaotong Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| |
Collapse
|
11
|
Basardeh E, Piri-Gavgani S, Soltanmohammadi B, Ghanei M, Omrani MD, Soezi M, Shokrgozar MA, Azizi M, Fateh A, Vaziri F, Siadat SD, Sharifzadeh Z, Rahimi-Jamnani F. Anti- Acinetobacter baumannii single-chain variable fragments show direct bactericidal activity. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1141-1149. [PMID: 36246061 PMCID: PMC9526879 DOI: 10.22038/ijbms.2022.64062.14106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 08/21/2022] [Indexed: 11/06/2022]
Abstract
Objectives The high resistance rate of Acinetobacter baumannii and the limited number of available antibiotics have prompted a worldwide effort to develop effective antimicrobial agents. Accordingly, identifying single-chain variable fragment antibodies (scFvs), capable of exerting direct antibacterial activity in an immune system-independent manner, may be making immunocompromised patients more susceptible to A. baumannii infections. Materials and Methods To isolate bactericidal scFvs targeting A. baumannii, we panned a large human scFv phage display library against whole-cell extensively drug-resistant (XDR) A. baumannii strains grown as biofilm or cultured with human blood or human peripheral blood mononuclear cells plus plasma. The binding of scFv-phages to A. baumannii was assessed by the dot-blot assay. Soluble scFvs, derived from the selected phages, were assessed based on their ability to bind and inhibit the growth of A. baumannii. Results Five phage clones showed the highest reactivity toward A. baumannii. Among five soluble scFvs, derived from positive phage clones, two scFvs, EB211 and EB279, had high expression yields and displayed strong binding to A. baumannii compared with the controls. Moreover, XDR A. baumannii strains treated with positively-charged scFvs, including EB211, EB279, or a cocktail of EB211 and EB279 (200 µg/ml), displayed lower viability (approximately 50%, 78%, and 40% viability, respectively) compared with PBS-treated bacteria. Conclusion These results suggest that combining last-resort antibiotics with bactericidal scFvs could provide promising outcomes in immunocompromised individuals with A. baumannii infections.
Collapse
Affiliation(s)
- Eilnaz Basardeh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Somayeh Piri-Gavgani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Behnoush Soltanmohammadi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Soezi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Masoumeh Azizi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Rahimi-Jamnani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran ,Corresponding author: Fatemeh Rahimi-Jamnani. Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran. Tel: +98-21-66953311; Fax: +98-21-66465132;
| |
Collapse
|
12
|
High Level Expression and Purification of Cecropin-like Antimicrobial Peptides in Escherichia coli. Biomedicines 2022; 10:biomedicines10061351. [PMID: 35740373 PMCID: PMC9220022 DOI: 10.3390/biomedicines10061351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/05/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
Cecropins are a family of antimicrobial peptides (AMPs) that are widely found in the innate immune system of Cecropia moths. Cecropins exhibit a broad spectrum of antimicrobial and anticancer activities. The structures of Cecropins are composed of 34–39 amino acids with an N-terminal amphipathic α-helix, an AGP hinge and a hydrophobic C-terminal α-helix. KR12AGPWR6 was designed based on the Cecropin-like structural feature. In addition to its antimicrobial activities, KR12AGPWR6 also possesses enhanced salt resistance, antiendotoxin and anticancer properties. Herein, we have developed a strategy to produce recombinant KR12AGPWR6 through a salt-sensitive, pH and temperature dependent intein self-cleavage system. The His6-Intein-KR12AGPWR6 was expressed by E. coli and KR12AGPWR6 was released by the self-cleavage of intein under optimized ionic strength, pH and temperature conditions. The molecular weight and structural feature of the recombinant KR12AGPWR6 was determined by MALDI-TOF mass, CD, and NMR spectroscopy. The recombinant KR12AGPWR6 exhibited similar antimicrobial activities compared to the chemically synthesized KR12AGPWR6. Our results provide a potential strategy to obtain large quantities of AMPs and this method is feasible and easy to scale up for commercial production.
Collapse
|
13
|
Costa SS, Lago LAB, Silva A, Graças DAD, Lameira J, Baraúna RA. Diversity of bacteriocins in the microbiome of the Tucuruí Hydroelectric Power Plant water reservoir and three-dimensional structure prediction of a zoocin. Genet Mol Biol 2022; 45:e20210204. [PMID: 35037933 PMCID: PMC8762718 DOI: 10.1590/1678-4685-gmb-2021-0204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022] Open
Abstract
Bacteriocins are antimicrobial peptides expressed by bacteria through ribosomal activity. In this study, we analyzed the diversity of bacteriocin-like genes in the Tucuruí-HPP using a whole-metagenome shotgun sequencing approach. Three layers of the water column were analyzed (photic, aphotic and sediment). Detection of bacteriocin-like genes was performed with blastx using the BAGEL4 database as subject sequences. In order to calculate the abundance of bacteriocin-like genes we also determined the number of 16S rRNA genes using blastn. Taxonomic analysis was performed using RAST server and the metagenome was assembled using IDBA-UD in order to recover the full sequence of a zoocin which had its three-dimensional structure determined. The photic zone presented the highest number of reads affiliated to bacteriocins. The most abundant bacteriocins were sonorensin, Klebicin D , pyocin and colicin. The zoocin model was composed of eight anti-parallel β-sheets and two α-helices with a Zn2+ ion in the active site. This model was considerably stable during 10 ns of molecular dynamics simulation. We observed a high diversity of bacteriocins in the Tucuruí-HPP, demonstrating that the environment is an inexhaustible source for prospecting these molecules. Finally, the zoocin model can be used for further studies of substrate binding and molecular mechanisms involving peptidoglycan degradation.
Collapse
Affiliation(s)
- Sávio S Costa
- Parque de Ciência e Tecnologia Guamá, Laboratório de Engenharia Biológica, Belém, PA, Brazil
| | - Leticia A B Lago
- Parque de Ciência e Tecnologia Guamá, Laboratório de Engenharia Biológica, Belém, PA, Brazil
| | - Artur Silva
- Parque de Ciência e Tecnologia Guamá, Laboratório de Engenharia Biológica, Belém, PA, Brazil
| | - Diego A das Graças
- Parque de Ciência e Tecnologia Guamá, Laboratório de Engenharia Biológica, Belém, PA, Brazil
| | - Jerônimo Lameira
- Universidade Federal do Pará, Instituto de Ciências Exatas e Naturais, Laboratório de Planejamento e Desenvolvimento de Fármacos, Belém, PA, Brazil
| | - Rafael A Baraúna
- Parque de Ciência e Tecnologia Guamá, Laboratório de Engenharia Biológica, Belém, PA, Brazil
| |
Collapse
|
14
|
Ghadaksaz A, Nodoushan SM, Sedighian H, Behzadi E, Fooladi AAI. Evaluation of the Role of Probiotics As a New Strategy to Eliminate Microbial Toxins: a Review. Probiotics Antimicrob Proteins 2022; 14:224-237. [PMID: 35031968 DOI: 10.1007/s12602-021-09893-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2021] [Indexed: 01/17/2023]
Abstract
Probiotics are living microorganisms that have favorable effects on human and animal health. The most usual types of microorganisms recruited as probiotics are lactic acid bacteria (LAB) and bifidobacteria. To date, numerous utilizations of probiotics have been reported. In this paper, it is suggested that probiotic bacteria can be recruited to remove and degrade different types of toxins such as mycotoxins and algal toxins that damage host tissues and the immune system causing local and systemic infections. These microorganisms can remove toxins by disrupting, changing the permeability of the plasma membrane, producing metabolites, inhibiting the protein translation, hindering the binding to GTP binding proteins to GM1 receptors, or by preventing the interaction between toxins and adhesions. Here, we intend to review the mechanisms that probiotic bacteria use to eliminate and degrade microbial toxins.
Collapse
Affiliation(s)
- Abdolamir Ghadaksaz
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Somayeh Mousavi Nodoushan
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Vanak Sq. Molasadra St, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Vanak Sq. Molasadra St, Tehran, Iran
| | - Elham Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-E-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Vanak Sq. Molasadra St, Tehran, Iran.
| |
Collapse
|
15
|
Darbandi A, Asadi A, Mahdizade Ari M, Ohadi E, Talebi M, Halaj Zadeh M, Darb Emamie A, Ghanavati R, Kakanj M. Bacteriocins: Properties and potential use as antimicrobials. J Clin Lab Anal 2021; 36:e24093. [PMID: 34851542 PMCID: PMC8761470 DOI: 10.1002/jcla.24093] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/03/2021] [Accepted: 10/24/2021] [Indexed: 12/12/2022] Open
Abstract
A variety of bacteriocins originate from lactic acid bacteria, which have recently been modified by scientists. Many strains of lactic acid bacteria related to food groups could produce bacteriocins or antibacterial proteins highly effective against foodborne pathogens such as Staphylococcus aureus, Pseudomonas fluorescens, P. aeruginosa, Salmonella typhi, Shigella flexneri, Listeria monocytogenes, Escherichia coli O157:H7, and Clostridium botulinum. A wide range of bacteria belonging primarily to the genera Bifidobacterium and Lactobacillus have been characterized with different health‐promoting attributes. Extensive studies and in‐depth understanding of these antimicrobials mechanisms of action could enable scientists to determine their production in specific probiotic lactic acid bacteria, as they are potentially crucial for the final preservation of functional foods or for medicinal applications. In this review study, the structure, classification, mode of operation, safety, and antibacterial properties of bacteriocins as well as their effect on foodborne pathogens and antibiotic‐resistant bacteria were extensively studied.
Collapse
Affiliation(s)
- Atieh Darbandi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Arezoo Asadi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Elnaz Ohadi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Masoume Halaj Zadeh
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Darb Emamie
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Kakanj
- Food and Drug Laboratory Research Center, Food and Drug Administration, MOH&ME, Tehran, Iran
| |
Collapse
|
16
|
Zhang S, Qu X, Tang H, Wang Y, Yang H, Yuan W, Yue B. Diclofenac Resensitizes Methicillin-Resistant Staphylococcus aureus to β-Lactams and Prevents Implant Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2100681. [PMID: 34258168 PMCID: PMC8261494 DOI: 10.1002/advs.202100681] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/12/2021] [Indexed: 05/25/2023]
Abstract
Implant infections caused by methicillin-resistant Staphylococcus aureus (MRSA) can cause major complications during the perioperative period. Diclofenac, one of the most widely used nonsteroidal anti-inflammatory drugs, is often used to relieve pain and inflammation. In this study, it is found that high-dose diclofenac can inhibit the growth of MRSA, and does not easily induce drug-resistant mutations after continuous passage. However, low-doses diclofenac can resensitize bacteria to β-lactams, which help to circumvent drug resistance and improve the antibacterial efficacy of conventional antibiotics. Further, low-dose diclofenac in combination with β-lactams inhibit MRSA associated biofilm formation in implants. Transcriptomic and proteomic analyses indicate that diclofenac can reduce the expression of genes and proteins associated with β-lactam resistance: mecA, mecR, and blaZ; peptidoglycan biosynthesis: murA, murC, femA, and femB; and biofilm formation: altE and fnbP. Murine implant infection models indicate that diclofenac combined with β-lactams, can substantially alleviate MRSA infections in vivo. In addition, it is investigated that low dose diclofenac can inhibit MRSA antibiotic resistance via the mecA/blaZ pathway and related biofilms in implants. The synergistic effect of diclofenac and β-lactams might have promising applications for preventing perioperative infection, considering its multitarget effects against MRSA.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - You Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| | - Hongtao Yang
- Department of Plastic & Reconstructive SurgeryThe Ohio State UniversityColumbusOH43210USA
- School of Medical Science and EngineeringBeihang UniversityBeijing100191China
| | - Weien Yuan
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of EducationSchool of PharmacyShanghai Jiao Tong UniversityShanghai200240China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of MedicineShanghai Jiaotong UniversityShanghai200127China
| |
Collapse
|
17
|
Successful Development of Bacteriocins into Therapeutic Formulation for Treatment of MRSA Skin Infection in a Murine Model. Antimicrob Agents Chemother 2020; 64:AAC.00829-20. [PMID: 32958719 PMCID: PMC7674055 DOI: 10.1128/aac.00829-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 09/17/2020] [Indexed: 01/08/2023] Open
Abstract
The emergence of antibiotic-resistant pathogens has caused a serious worldwide problem in infection treatment in recent years. One of the pathogens is methicillin-resistant Staphylococcus aureus (MRSA), which is a major cause of skin and soft tissue infections. Alternative strategies and novel sources of antimicrobials to solve antibiotic resistance problems are urgently needed. In this study, we explored the potential of two broad-spectrum bacteriocins, garvicin KS and micrococcin P1, in skin infection treatments. The emergence of antibiotic-resistant pathogens has caused a serious worldwide problem in infection treatment in recent years. One of the pathogens is methicillin-resistant Staphylococcus aureus (MRSA), which is a major cause of skin and soft tissue infections. Alternative strategies and novel sources of antimicrobials to solve antibiotic resistance problems are urgently needed. In this study, we explored the potential of two broad-spectrum bacteriocins, garvicin KS and micrococcin P1, in skin infection treatments. The two bacteriocins acted synergistically with each other and with penicillin G in killing MRSA in vitro. The MICs of the antimicrobials in the three-component mixture were 40 ng/ml for micrococcin P1 and 2 μg/ml for garvicin KS and penicillin G, which were 62, 16, and at least 1,250 times lower than their MICs when assessed individually. To assess its therapeutic potential further, we challenged the three-component formulation in a murine skin infection model with the multidrug-resistant luciferase-tagged MRSA Xen31, a strain derived from the clinical isolate S. aureus ATCC 33591. Using the tagged-luciferase activity as a reporter for the presence of Xen31 in wounds, we demonstrated that the three-component formulation was efficient in eradicating the pathogen from treated wounds. Furthermore, compared to Fucidin cream, which is an antibiotic commonly used in skin infection treatments, our formulation was also superior in terms of preventing resistance development.
Collapse
|
18
|
Zhang S, Tang H, Wang Y, Nie B, Yang H, Yuan W, Qu X, Yue B. Antibacterial and antibiofilm effects of flufenamic acid against methicillin-resistant Staphylococcus aureus. Pharmacol Res 2020; 160:105067. [PMID: 32650057 DOI: 10.1016/j.phrs.2020.105067] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infections are one of the most serious surgery complications, and their prevention is of utmost importance. Flufenamic acid is a non-steroid anti-inflammatory drug approved for clinical use to relieve inflammation and pain in rheumatoid arthritis patients. In this study, we explored the antibacterial efficacy of flufenamic acid and the mechanisms underlying this effect. By using minimal inhibitory concentration (MIC), time-kill, resistance induction assays, and the antibiotic synergy test, we demonstrated that flufenamic acid inhibited the growth of methicillin-resistant staphylococci and did not induce resistance when it was used at the MIC. Furthermore, flufenamic acid acted synergistically with the beta-lactam antibiotic oxacillin and did not show significant toxicity toward mammalian cells. The biofilm inhibition assay revealed that flufenamic acid could prevent biofilm formation on medical implants and destroy the ultrastructure of the bacterial cell wall. RNA sequencing and quantitative RT-PCR indicated that flufenamic acid inhibited the expression of genes associated with peptidoglycan biosynthesis, beta-lactam resistance, quorum sensing, and biofilm formation. Furthermore, flufenamic acid efficiently ameliorated a local infection caused by MRSA in mice. In conclusion, flufenamic acid may be a potent therapeutic compound against MRSA infections and a promising candidate for antimicrobial coating of implants and surgical devices.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin'en Nie
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongtao Yang
- Department of Plastic & Reconstructive Surgery, The Ohio State University, Columbus, OH 43210, United States
| | - Weien Yuan
- Ministry of Education Engineering Research Center of Cell & Therapeutic Antibody, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Bing Yue
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
19
|
Antimicrobial Peptides with Enhanced Salt Resistance and Antiendotoxin Properties. Int J Mol Sci 2020; 21:ijms21186810. [PMID: 32948086 PMCID: PMC7554977 DOI: 10.3390/ijms21186810] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/08/2020] [Accepted: 09/12/2020] [Indexed: 12/14/2022] Open
Abstract
A strategy was described to design antimicrobial peptides (AMPs) with enhanced salt resistance and antiendotoxin activities by linking two helical AMPs with the Ala-Gly-Pro (AGP) hinge. Among the designed peptides, KR12AGPWR6 demonstrated the best antimicrobial activities even in high salt conditions (NaCl ~300 mM) and possessed the strongest antiendotoxin activities. These activities may be related to hydrophobicity, membrane-permeability, and α-helical content of the peptide. Amino acids of the C-terminal helices were found to affect the peptide-induced permeabilization of LUVs, the α-helicity of the designed peptides under various LUVs, and the LPS aggregation and size alternation. A possible model was proposed to explain the mechanism of LPS neutralization by the designed peptides. These findings could provide a new approach for designing AMPs with enhanced salt resistance and antiendotoxin activities for potential therapeutic applications.
Collapse
|
20
|
Isayenko OY, Knysh OV, Kotsar OV, Ryzhkova TN, Dyukareva GI. Simultaneous and sequential influence of metabolite complexes of Lactobacillus rhamnosus and Saccharomyces boulardii and antibiotics against poly-resistant Gram-negative bacteria. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
For the first time the poly-resistant strains of Gram-negative microorganisms were studied for the sensitivity to combined simultaneous and sequential influence of metabolic complexes of Lactobacillus rhamnosus GG and Saccharomyces boulardii, obtained by the author’s method without using the growth media, with antibiotics. The synergic activity of antibacterial preparations and metabolic complexes of L. rhamnosus GG and S. boulardii were studied using modified disk-diffusive method of Kirby-Bauer. During the sequential method of testing (at first the microorganisms were incubated with structural components and metabolites, then their sensitivity to the antibacterial preparations was determined), we observed increase in the diameters of the zones of growth inhibition of Pseudomonas aeruginosa PR to the typical antibiotics (gentamicin, amіcyl, ciprofloxacin, сefotaxime) and non-typical (lincomycin, levomycetin) depending on the tested combinations. Acinetobacter baumannii PR exhibited lower susceptibility: growth inhibition was seen for the combination with ciprofloxacin, сefotaxime, levomycetin. Susceptibility of Lelliottia amnigena (Enterobacter amnigenus) PR increased to levofloxacin, lincomycin. The zones of growth inhibition of Klebsiella pneumoniae PR increased to gentamicin, amіcyl, tetracycline, сeftriaxone. Maximum efficiency was determined during sequential combination of antibiotics with separate metabolic complexes of L. rhamnosus and S. boulardii, and also their combination (to 15.2, 20.2 and 15.4 mm respectively) compared with their simultaneous use (to 12.2, 15.2 and 13.0 mm respectively) for all the tested poly-resistant pathogens, regardless of the mechanism of action of antibacterial preparation. Metabolic complexes of L. rhamnosus GG and S. boulardii, due to increase in the susceptibility of microorganisms, can decrease the therapeutic concentration of antibiotic, slow the probability of the development of resistance of microorganisms, and are therefore promising candidates for developing “accompanying medications” to antibiotics and antimicrobial preparations of new generation.
Collapse
|
21
|
Ellis JC, Ross RP, Hill C. Nisin Z and lacticin 3147 improve efficacy of antibiotics against clinically significant bacteria. Future Microbiol 2020; 14:1573-1587. [PMID: 32019322 DOI: 10.2217/fmb-2019-0153] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: To determine if bacteriocins improve antibiotic efficacy. Materials & methods: Deferred antagonism assays identified bacteriocins with activity. Growth curves and time kill assays demonstrated bactericidal activity of antimicrobial combinations, and checkerboard assays confirmed synergy. Methicillin-resistant Staphylococcus aureus-infected porcine skin model determined ex vivo efficacy. Results: Subinhibitory concentrations of lacticin with penicillin or vancomycin resulted in complete growth inhibition of strains and the improved inhibitory effect was apparent after 1 h. Nisin with methicillin proved more effective against methicillin-resistant Staphylococcus aureus than either antimicrobial alone, revealing partial synergy and significantly reduced pathogen numbers on porcine skin after 3 h compared with minimal inhibition for either antimicrobial alone. Conclusion: Nisin Z and lacticin 3147 may support the use of certain antibiotics and revive ineffective antibiotics.
Collapse
Affiliation(s)
| | - Reynolds P Ross
- School of Microbiology, University College Cork, T12 K8AF, Ireland.,APC Microbiome Ireland, University College Cork, T12 YT20, Ireland
| | - Colin Hill
- School of Microbiology, University College Cork, T12 K8AF, Ireland.,APC Microbiome Ireland, University College Cork, T12 YT20, Ireland
| |
Collapse
|
22
|
Ruden S, Rieder A, Chis Ster I, Schwartz T, Mikut R, Hilpert K. Synergy Pattern of Short Cationic Antimicrobial Peptides Against Multidrug-Resistant Pseudomonas aeruginosa. Front Microbiol 2019; 10:2740. [PMID: 31849888 PMCID: PMC6901909 DOI: 10.3389/fmicb.2019.02740] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
With the rise of various multidrug-resistant (MDR) pathogenic bacteria, worldwide health care is under pressure to respond. Conventional antibiotics are failing and the development of novel classes and alternative strategies is a major priority. Antimicrobial peptides (AMPs) cannot only kill MDR bacteria, but also can be used synergistically with conventional antibiotics. We selected 30 short AMPs from different origins and measured their synergy in combination with polymyxin B, piperacillin, ceftazidime, cefepime, meropenem, imipenem, tetracycline, erythromycin, kanamycin, tobramycin, amikacin, gentamycin, and ciprofloxacin. In total, 403 unique combinations were tested against an MDR Pseudomonas aeruginosa isolate (PA910). As a measure of the synergistic effects, fractional inhibitory concentrations (FICs) were determined using microdilution assays with FICs ranges between 0.25 and 2. A high number of combinations between peptides and polymyxin B, erythromycin, and tetracycline were found to be synergistic. Novel variants of indolicidin also showed a high frequency in synergist interaction. Single amino acid substitutions within the peptides can have a very strong effect on the ability to synergize, making it possible to optimize future drugs toward synergistic interaction.
Collapse
Affiliation(s)
- Serge Ruden
- Institute of Biological Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Annika Rieder
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Irina Chis Ster
- Institute of Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Thomas Schwartz
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Ralf Mikut
- Institute for Automation and Applied Informatics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Kai Hilpert
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Institute of Infection and Immunity, St George's, University of London, London, United Kingdom.,Institute of Microstructure Technology, Karlsruhe Institute of Technology, Karlsruhe, Germany
| |
Collapse
|
23
|
Lewis R, Bolocan AS, Draper LA, Ross RP, Hill C. The Effect of a Commercially Available Bacteriophage and Bacteriocin on Listeria monocytogenes in Coleslaw. Viruses 2019; 11:E977. [PMID: 31652871 PMCID: PMC6893746 DOI: 10.3390/v11110977] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 10/21/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022] Open
Abstract
Changing consumer attitudes show an increased interest in non-chemical antimicrobials in food preservation and safety. This greater interest of consumers in more 'natural' or 'clean-label' food interventions is complicated by concurrent demands for minimally processed, ready-to-eat (RTE) foods with long shelf lives. Two viable interventions are bacteriophage (phage) and bacteriocins, a number of which have already been approved for use in food safety. Listeriosis is a serious foodborne infection which affects at-risk members of the population. Listeriosis incidence has increased between 2008 and 2015 and has a case fatality rate of up to 20% with antibiotic intervention. Here, we tested an intervention to attempt to control a pathogenic Listeria monocytogenes strain in a food model using two of these alternative antimicrobials. Phage P100 on its own had a significant effect on L. monocytogenes ScottA numbers in coleslaw over a 10-day period at 4 °C (p ≤ 0.001). A combination of P100 and Nisaplin® (a commercial formulation of the lantibiotic bacteriocin, nisin) had a significant effect on the pathogen (p ≤ 0.001). P100 and Nisaplin® in combination were more effective than Nisaplin® alone, but not P100 alone.
Collapse
Affiliation(s)
- Rhea Lewis
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - Andrei Sorin Bolocan
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - Lorraine A Draper
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland.
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland.
| |
Collapse
|
24
|
Mei J, Ma X, Xie J. Review on Natural Preservatives for Extending Fish Shelf Life. Foods 2019; 8:E490. [PMID: 31614926 PMCID: PMC6835557 DOI: 10.3390/foods8100490] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/22/2022] Open
Abstract
Fish is extremely perishable as a result of rapid microbial growth naturally present in fish or from contamination. Synthetic preservatives are widely used in fish storage to extend shelf life and maintain quality and safety. However, consumer preferences for natural preservatives and concerns about the safety of synthetic preservatives have prompted the food industry to search natural preservatives. Natural preservatives from microorganisms, plants, and animals have been shown potential in replacing the chemical antimicrobials. Bacteriocins and organic acids from bacteria showed good antimicrobial activities against spoilage bacteria. Plant-derived antimicrobials could prolong fish shelf life and decrease lipid oxidation. Animal-derived antimicrobials also have good antimicrobial activities; however, their allergen risk should be paid attention. Moreover, some algae and mushroom species can also provide a potential source of new natural preservatives. Obviously, the natural preservatives could perform better in fish storage by combining with other hurdles such as non-thermal sterilization processing, modified atmosphere packaging, edible films and coatings.
Collapse
Affiliation(s)
- Jun Mei
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- National Experimental Teaching Demonstration Center for Food Science and Engineering Shanghai Ocean University, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic Product Processing and Preservation, Shanghai 201306, China
- Shanghai Professional Technology Service Platform on Cold Chain Equipment Performance and Energy Saving Evaluation, Shanghai 201306, China
| | - Xuan Ma
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Jing Xie
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
- National Experimental Teaching Demonstration Center for Food Science and Engineering Shanghai Ocean University, Shanghai 201306, China.
- Shanghai Engineering Research Center of Aquatic Product Processing and Preservation, Shanghai 201306, China.
- Shanghai Professional Technology Service Platform on Cold Chain Equipment Performance and Energy Saving Evaluation, Shanghai 201306, China.
| |
Collapse
|
25
|
Bypassing lantibiotic resistance by an effective nisin derivative. Bioorg Med Chem 2019; 27:3454-3462. [PMID: 31253534 DOI: 10.1016/j.bmc.2019.06.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/29/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022]
Abstract
The need for new antibiotic compounds is rising and antimicrobial peptides are excellent candidates to fulfill this object. The bacteriocin subgroup lantibiotics, for example, are active in the nanomolar range and target the membranes of mainly Gram-positive bacteria. They bind to lipid II, inhibit cell growth and in some cases form pores within the bacterial membrane, inducing rapid cell death. Pharmaceutical usage of lantibiotics is however hampered by the presence of gene clusters in human pathogenic strains which, when expressed, confer resistance. The human pathogen Streptococcus agalactiae COH1, expresses several lantibiotic resistance proteins resulting in resistance against for example nisin. This study presents a highly potent, pore forming nisin variant as an alternative lantibiotic which bypasses the SaNSR protein. It is shown that this nisin derivate nisinC28P keeps its nanomolar antibacterial activity against L. lactis NZ9000 cells but is not recognized by the nisin resistance protein SaNSR. NisinC28P is cleaved by SaNSR in vitro with a highly decreased efficiency, as shown by an cleavage assay. Furthermore, we show that nisinC28P is still able to form pores in the membranes of L. lactis and is three times more efficient against SaNSR-expressing L. lactis cells than wildtype nisin.
Collapse
|
26
|
Tenea GN, Hurtado P, Ortega C. Inhibitory Effect of Substances Produced by Native Lactococcus lactis Strains of Tropical Fruits towards Food Pathogens. Prev Nutr Food Sci 2018; 23:260-268. [PMID: 30386755 PMCID: PMC6195891 DOI: 10.3746/pnf.2018.23.3.260] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/25/2018] [Indexed: 12/22/2022] Open
Abstract
The use of peptides produced by lactic acid bacteria (LAB) as antimicrobial agents in food emerged from the increasing need of replacing chemicals with natural substances to ensure their safety and quality. A total of 30 LAB belonging to the genus Lactococcus sp. (10) and Enterococcus sp. (20) were isolated from native fruits of Ecuador subtropical rainforest. Among Lactococcus species, the isolates assigned Gt28, Gt29, and Ella8, identified as Lactococcus lactis subsp. lactis with 99% identity, showing highly inhibitory potential against four food pathogens were further characterized. The treatment of cell-free supernatant with proteolytic enzymes indicated the protein nature of released components, which displayed a broad antimicrobial activity against Gram-positive and -negative bacteria. Polymerase chain reaction analysis indicated the presence of lacticin 3147 gene in all isolates, lactococcin M gene in Gt28 and Gt29 but not in Ella8 and lactococcin A gene in Gt28 only. The antimicrobial activity was not linked to the presence of structural nisin gene as no amplification product was obtained. Treatment of Salmonella enterica ATCC 51741 and Escherichia coli ATCC 25922 at both vegetative and exponential phase of growth with the cell-free supernatant of Gt28 resulted in complete inactivation upon 3 h suggesting its bactericidal mode of action. An increment on inhibitory activity occurred when partial purified bacteriocin Gt28 was combined with ethylenediaminetetraacetic acid rather than bacteriocin only, indicating that the cells were sensitized in vitro by the chelator agent acting synergistically to induce the killing of pathogenic cells.
Collapse
Affiliation(s)
- Gabriela Nicoleta Tenea
- Faculty of Engineering in Agricultural and Environmental Sciences, The Technical University of the North, Ibarra, Imbabura 199, Ecuador
| | - Pamela Hurtado
- Faculty of Engineering in Agricultural and Environmental Sciences, The Technical University of the North, Ibarra, Imbabura 199, Ecuador
| | - Clara Ortega
- Faculty of Engineering in Agricultural and Environmental Sciences, The Technical University of the North, Ibarra, Imbabura 199, Ecuador
| |
Collapse
|
27
|
Chih YH, Wang SY, Yip BS, Cheng KT, Hsu SY, Wu CL, Yu HY, Cheng JW. Dependence on size and shape of non-nature amino acids in the enhancement of lipopolysaccharide (LPS) neutralizing activities of antimicrobial peptides. J Colloid Interface Sci 2018; 533:492-502. [PMID: 30176540 DOI: 10.1016/j.jcis.2018.08.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/11/2022]
Abstract
HYPOTHESIS Release of lipopolysaccharides (LPS) from bacteria into bloodstream may cause serious unwanted stimulation of the host immune system. P-113 is a clinically active histidine-rich antimicrobial peptide. Nal-P-113, a β-naphthylalanine-substituted P-113, is salt-resistant but has limited LPS neutralizing activity. We suspected the size and shape of the non-natural bulky amino acid may affect its LPS neutralizing activity. Herein, antimicrobial, LPS neutralizing, and antiproteolytic effects of phenylalanine- (Phe-P-113), β-naphthylalanine- (Nal-P-113), β-diphenylalanine- (Dip-P-113), and β-(4,4'-biphenyl)alanine- (Bip-P-113) substituted P-113 were studied. EXPERIMENTS Structure-activity relationships of P-113, Phe-P-113, Nal-P-113, Dip-P-113, and Bip-P-113 were evaluated using antimicrobial activity assays, serum proteolytic assays, peptide-induced permeabilization of large unilamellar vesicles, zeta potential measurements, dynamic light scattering measurement of LPS aggregation, and Limulus amebocyte lysate assays for measuring LPS neutralization. In vitro and in vivo LPS neutralizing activities were further confirmed by LPS-induced inflammation inhibition in an endotoxemia mouse model. FINDINGS Bip-P-113 and Dip-P-113 had the longest and widest non-nature amino acids, respectively. Bip-P-113 enhanced salt resistance, serum proteolytic stability, peptide-induced permeabilization, zeta potential measurements, LPS aggregation, and in vitro and in vivo LPS neutralizing activities. These results could help design novel antimicrobial peptides that have enhanced stability in vivo and that can have potential therapeutic applications.
Collapse
Affiliation(s)
- Ya-Han Chih
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Siou-Ying Wang
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Bak-Sau Yip
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu 300, Taiwan
| | - Kuang-Ting Cheng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Su-Ya Hsu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chih-Lung Wu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hui-Yuan Yu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jya-Wei Cheng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
28
|
Wang YM, Kong LC, Liu J, Ma HX. Synergistic effect of eugenol with Colistin against clinical isolated Colistin-resistant Escherichia coli strains. Antimicrob Resist Infect Control 2018; 7:17. [PMID: 29423192 PMCID: PMC5789579 DOI: 10.1186/s13756-018-0303-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/12/2018] [Indexed: 11/22/2022] Open
Abstract
Background Bacterial infections have become more challenging to treat due to the emergence of multidrug-resistant pathogenic bacteria. Combined antibiotics prove to be a relatively effective method to control such resistant strains. This study aim to investigate synergistic activity of eugenol combined with colistin against a collection of clinical isolated Escherichia coli (E.coli) strains, and to evaluate potential interaction. Methods Antimicrobial susceptibility, minimum inhibitory concentration (MIC) and fractional inhibitory concentration index (FICI) of the bacteria were determined by disk diffusion assay, broth microdilution method and checkerboard assay, respectively. The mcr-1 mRNA expression was measured by Real-time PCR. To predict possible interactions between eugenol and MCR-1, molecular docking assay was taken. Results For total fourteen strains including eight colistin-resistant strains, eugenol was determined with MIC values of 4 to 8 μg/mL. Checkerboard dilution test suggested that eugenol exhibited synergistic activity when combined with colistin (FICI ranging from 0.375 to 0.625). Comparison analysis of Real-time PCR showed that synergy could significantly down-regulate expression of mcr-1 gene. A metal ion coordination bond with catalytic zinc atom and a hydrogen bond with crucial amino acid residue Ser284 of MCR-1 were observed after molecular docking, indicating antibacterial activity and direct molecular interactions of eugenol with MCR-1 protein. Conclusions Our results demonstrated that eugenol exhibited synergistic effect with colistin and enhanced its antimicrobial activity. This might further contribute to the antibacterial actions against colistin-resistant E.coli strains. Graphical abstract Synergistic effect of eugenol with colistin against colistin-resistant Escherichia coli isolates.![]()
Collapse
Affiliation(s)
- Yi-Ming Wang
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No.2888, Changchun, 130118 China
| | - Ling-Cong Kong
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No.2888, Changchun, 130118 China
| | - Jie Liu
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No.2888, Changchun, 130118 China
| | - Hong-Xia Ma
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street No.2888, Changchun, 130118 China
| |
Collapse
|
29
|
Wang R, Hou S, Dong X, Chen D, Shao L, Qian L, Li Z, Xu X. Synergism of fused bicyclic 2-aminothiazolyl compounds with polymyxin B against Klebsiella pneumoniae. MEDCHEMCOMM 2017; 8:2060-2066. [PMID: 30108723 PMCID: PMC6071964 DOI: 10.1039/c7md00354d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/14/2017] [Indexed: 12/16/2022]
Abstract
A series of fused bicyclic 2-aminothiazolyl compounds were synthesized and evaluated for their synergistic effects with polymyxin B (PB) against Klebsiella pneumoniae (SIPI-KPN-1712). Some of the synthesized compounds exhibited synergistic activity. When 4 μg ml-1 compound B1 was combined with PB, it showed potent antibacterial activity, achieving 64-fold reduction of the MIC of PB. Furthermore, compound B1 showed prominent synergistic efficacy in both concentration gradient and time-kill curves in vitro. In addition, B1 combined with PB also exhibited synergistic and partial synergistic effect against E. coli (ATCC25922 and its clinical isolates), Acinetobacter baumannii (ATCC19606 and its clinical isolates), and Pseudomonas aeruginosa (Pae-1399).
Collapse
Affiliation(s)
- Rong Wang
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64252945
| | - Shuang Hou
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64252945
| | - Xiaojing Dong
- School of Pharmacy , Shanghai Jiaotong University , China
| | - Daijie Chen
- School of Pharmacy , Shanghai Jiaotong University , China
| | - Lei Shao
- Shanghai Institute of Pharmaceutical Industry , China
| | - Liujia Qian
- Shanghai Institute of Pharmaceutical Industry , China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64252945
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology , Shanghai 200237 , China
| | - Xiaoyong Xu
- Shanghai Key Laboratory of Chemical Biology , School of Pharmacy , East China University of Science and Technology , Shanghai 200237 , China . ; ; Tel: +86 21 64252945
- Shanghai Collaborative Innovation Center for Biomanufacturing Technology , Shanghai 200237 , China
| |
Collapse
|
30
|
Characterization of a Francisella tularensis-Caenorhabditis elegans Pathosystem for the Evaluation of Therapeutic Compounds. Antimicrob Agents Chemother 2017; 61:AAC.00310-17. [PMID: 28652232 DOI: 10.1128/aac.00310-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/15/2017] [Indexed: 12/18/2022] Open
Abstract
Francisella tularensis is a highly infectious Gram-negative intracellular pathogen that causes tularemia. Because of its potential as a bioterrorism agent, there is a need for new therapeutic agents. We therefore developed a whole-animal Caenorhabditis elegans-F. tularensis pathosystem for high-throughput screening to identify and characterize potential therapeutic compounds. We found that the C. elegans p38 mitogen-activate protein (MAP) kinase cascade is involved in the immune response to F. tularensis, and we developed a robust F. tularensis-mediated C. elegans killing assay with a Z' factor consistently of >0.5, which was then utilized to screen a library of FDA-approved compounds that included 1,760 small molecules. In addition to clinically used antibiotics, five FDA-approved drugs were also identified as potential hits, including the anti-inflammatory drug diflunisal that showed anti-F. tularensis activity in vitro Moreover, the nonsteroidal anti-inflammatory drug (NSAID) diflunisal, at 4× MIC, blocked the replication of an F. tularensis live vaccine strain (LVS) in primary human macrophages and nonphagocytic cells. Diflunisal was nontoxic to human erythrocytes and HepG2 human liver cells at concentrations of ≥32 μg/ml. Finally, diflunisal exhibited synergetic activity with the antibiotic ciprofloxacin in both a checkerboard assay and a macrophage infection assay. In conclusion, the liquid C. elegans-F. tularensis LVS assay described here allows screening for anti-F. tularensis compounds and suggests that diflunisal could potentially be repurposed for the management of tularemia.
Collapse
|
31
|
Kitazaki K, Koga S, Nagatoshi K, Kuwano K, Zendo T, Nakayama J, Sonomoto K, Ano H, Katamoto H. In vitro synergistic activities of cefazolin and nisin A against mastitis pathogens. J Vet Med Sci 2017; 79:1472-1479. [PMID: 28757508 PMCID: PMC5627315 DOI: 10.1292/jvms.17-0180] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
First-generation cephalosporins such as cefazolin (CEZ) have been widely used for mastitis treatment in dairy cattle. However, the use of antibiotics results in the presence of antibiotic residues in milk, which is used for human
consumption. Nisin A, a bacteriocin produced by Lactococcus lactis, has been used as a broad-spectrum food preservative for over 50 years. Therefore, a combination of CEZ and nisin A might provide an extended
activity spectrum against mastitis pathogens and reduce the antibiotic dose for mastitis treatment. This study aimed to evaluate the combined effect of CEZ and nisin A against mastitis pathogens using the checkerboard and
time-kill assays. In the checkerboard assay, the CEZ-nisin A combination exhibited a synergistic effect against Staphylococcus aureus (n=20/20) and Enterococcus faecalis (n=13/18), and meanwhile
exhibited a mostly additive effect against Staphylococcus intermedius (n=12/20), Streptococcus agalactiae (n=10/10), Streptococcus dysgalactiae (n=18/18), and Escherichia
coli (n=14/18). There were no indifferent or antagonistic effects between CEZ and nisin A. In the time-kill assay, the CEZ-nisin A combination at 0.5 × or 1 × minimum inhibitory concentration exhibited synergistic
reduction of bacterial growth by over 3 log10 colony forming units per ml relative to that observed with either antimicrobial substance alone. These results suggest that the CEZ-nisin A combination can
be used for developing an intramammary infusion for mastitis treatment, with lower antibiotic concentrations than normal.
Collapse
Affiliation(s)
- Kohei Kitazaki
- Department of Animal Production, Fukuoka Agriculture and Forestry Research Center, 587 Oaza-Yoshiki, Chikushino, Fukuoka 818-8549, Japan.,Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Shoko Koga
- Research and Development Division, Omu Milk Products Co., Ltd., 1-38-1 Shinkatsudati-machi, Omuta, Fukuoka 836-0895, Japan
| | - Kohei Nagatoshi
- Research and Development Division, Omu Milk Products Co., Ltd., 1-38-1 Shinkatsudati-machi, Omuta, Fukuoka 836-0895, Japan
| | - Koichi Kuwano
- Division of Microbiology, Department of Infectious Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka 830-0011, Japan
| | - Takeshi Zendo
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | - Jiro Nakayama
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | - Kenji Sonomoto
- Laboratory of Microbial Technology, Division of Systems Bioengineering, Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | - Hitoshi Ano
- Laboratory of Veterinary Internal Medicine, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki 889-2192, Japan
| | - Hiromu Katamoto
- Laboratory of Veterinary Internal Medicine, Faculty of Agriculture, University of Miyazaki, 1-1 Gakuen Kibanadai-nishi, Miyazaki 889-2192, Japan
| |
Collapse
|
32
|
Mathur H, Field D, Rea MC, Cotter PD, Hill C, Ross RP. Bacteriocin-Antimicrobial Synergy: A Medical and Food Perspective. Front Microbiol 2017; 8:1205. [PMID: 28706513 PMCID: PMC5489601 DOI: 10.3389/fmicb.2017.01205] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/14/2017] [Indexed: 12/18/2022] Open
Abstract
The continuing emergence of multi-drug resistant pathogens has sparked an interest in seeking alternative therapeutic options. Antimicrobial combinatorial therapy is one such avenue. A number of studies have been conducted, involving combinations of bacteriocins with other antimicrobials, to circumvent the development of antimicrobial resistance and/or increase antimicrobial potency. Such bacteriocin-antimicrobial combinations could have tremendous value, in terms of reducing the likelihood of resistance development due to the involvement of two distinct mechanisms of antimicrobial action. Furthermore, antimicrobial synergistic interactions may also have potential financial implications in terms of decreasing the costs of treatment by reducing the concentration of an expensive antimicrobial and utilizing it in combination with an inexpensive one. In addition, combinatorial therapies with bacteriocins can broaden antimicrobial spectra and/or result in a reduction in the concentration of an antibiotic required for effective treatments to the extent that potentially toxic or adverse side effects can be reduced or eliminated. Here, we review studies in which bacteriocins were found to be effective in combination with other antimicrobials, with a view to targeting clinical and/or food-borne pathogens. Furthermore, we discuss some of the bottlenecks which are currently hindering the development of bacteriocins as viable therapeutic options, as well as addressing the need to exercise caution when attempting to predict clinical outcomes of bacteriocin-antimicrobial combinations.
Collapse
Affiliation(s)
- Harsh Mathur
- Teagasc Food Research Centre, MooreparkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Des Field
- APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| | - Mary C Rea
- Teagasc Food Research Centre, MooreparkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Centre, MooreparkCork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Colin Hill
- APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| | - R Paul Ross
- APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| |
Collapse
|
33
|
Synergistic Efficacy of Aedes aegypti Antimicrobial Peptide Cecropin A2 and Tetracycline against Pseudomonas aeruginosa. Antimicrob Agents Chemother 2017; 61:AAC.00686-17. [PMID: 28483966 DOI: 10.1128/aac.00686-17] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 04/29/2017] [Indexed: 12/21/2022] Open
Abstract
The increasing prevalence of antibiotic resistance has created an urgent need for alternative drugs with new mechanisms of action. Antimicrobial peptides (AMPs) are promising candidates that could address the spread of multidrug-resistant bacteria, either alone or in combination with conventional antibiotics. We studied the antimicrobial efficacy and bactericidal mechanism of cecropin A2, a 36-residue α-helical cationic peptide derived from Aedes aegypti cecropin A, focusing on the common pathogen Pseudomonas aeruginosa The peptide showed little hemolytic activity and toxicity toward mammalian cells, and the MICs against most clinical P. aeruginosa isolates were 32 to 64 μg/ml, and its MICs versus other Gram-negative bacteria were 2 to 32 μg/ml. Importantly, cecropin A2 demonstrated synergistic activity against P. aeruginosa when combined with tetracycline, reducing the MICs of both agents by 8-fold. The combination was also effective in vivo in the P. aeruginosa/Galleria mellonella model (P < 0.001). We found that cecropin A2 bound to P. aeruginosa lipopolysaccharides, permeabilized the membrane, and interacted with the bacterial genomic DNA, thus facilitating the translocation of tetracycline into the cytoplasm. In summary, the combination of cecropin A2 and tetracycline demonstrated synergistic antibacterial activity against P. aeruginosain vitro and in vivo, offering an alternative approach for the treatment of P. aeruginosa infections.
Collapse
|
34
|
García-Cayuela T, Requena T, Martínez-Cuesta MC, Peláez C. Rapid detection of Lactococcuslactis isolates producing the lantibiotics nisin, lacticin 481 and lacticin 3147 using MALDI-TOF MS. J Microbiol Methods 2017; 139:138-142. [PMID: 28583849 DOI: 10.1016/j.mimet.2017.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 05/29/2017] [Accepted: 06/01/2017] [Indexed: 01/17/2023]
Abstract
The aim of the study was to evaluate the potential use of Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry (MALDI-TOF MS) for fast and reliable detection of strains producing the lantibiotics nisin, lacticin 481 and lacticin 3147 in a large collection of lactococci. A total of one hundred lactococcal isolates from traditional ewe's and goat's raw milk cheeses were identified to the species level as Lactococcuslactis by MALDI-TOF MS based on comparison with lactococcal entries in the BioTyper database. Mass spectra in the range 2000-4000Da of the identified isolates were compared to reference spectra of three lactococcal strains producing lacticin 481 (IFPL 330), lacticin 3147 (IFPL 105) and nisin (IFPL 503). Only eight isolates had mass spectra with peaks that could be unequivocally identified as lacticin 481 (2900.47Da) or nisin (3330.31Da). None of the assayed isolates matched the mass spectra corresponding to the two-peptide lacticin 3147 (2847.97 and 3306.29Da). The results obtained by MALDI-TOF MS were genetically validated by amplification of the corresponding structural gene coding for lacticin 481, nisin and lacticin 3147. MALDI-TOF MS can be used as a fast and reliable technique to screen a large number of lactococcal isolates for the ability to produce the lantibiotics nisin, lacticin 481 and lacticin 3147.
Collapse
Affiliation(s)
- Tomás García-Cayuela
- Departamento de Biotecnología y Microbiología de Alimentos, Instituto de Investigación en Ciencias de la Alimentación CIAL (CSIC), Madrid, Spain.
| | - Teresa Requena
- Departamento de Biotecnología y Microbiología de Alimentos, Instituto de Investigación en Ciencias de la Alimentación CIAL (CSIC), Madrid, Spain
| | - M Carmen Martínez-Cuesta
- Departamento de Biotecnología y Microbiología de Alimentos, Instituto de Investigación en Ciencias de la Alimentación CIAL (CSIC), Madrid, Spain
| | - Carmen Peláez
- Departamento de Biotecnología y Microbiología de Alimentos, Instituto de Investigación en Ciencias de la Alimentación CIAL (CSIC), Madrid, Spain
| |
Collapse
|
35
|
Mathur H, Fallico V, O'Connor PM, Rea MC, Cotter PD, Hill C, Ross RP. Insights into the Mode of Action of the Sactibiotic Thuricin CD. Front Microbiol 2017; 8:696. [PMID: 28473822 PMCID: PMC5397516 DOI: 10.3389/fmicb.2017.00696] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/05/2017] [Indexed: 12/28/2022] Open
Abstract
Thuricin CD is a two-component bacteriocin, consisting of the peptides Trnα and Trnβ, and belongs to the newly designated sactibiotic subclass of bacteriocins. While it is clear from studies conducted thus far that it is a narrow-spectrum bacteriocin, requiring the synergistic activity of the two peptides, the precise mechanism of action of thuricin CD has not been elucidated. This study used a combination of flow cytometry and traditional culture-dependent assays to ascertain the effects of the thuricin CD peptides on the morphology, physiology and viability of sensitive Bacillus firmus DPC6349 cells. We show that both Trnα and Trnβ are membrane-acting and cause a collapse of the membrane potential, which could not be reversed even under membrane-repolarizing conditions. Furthermore, the depolarizing action of thuricin CD is accompanied by reductions in cell size and granularity, producing a pattern of physiological alterations in DPC6349 cells similar to those triggered by the pore-forming single-component bacteriocin Nisin A, and two-component lacticin 3147. Taken together, these results lead us to postulate that the lytic activity of thuricin CD involves the insertion of thuricin CD peptides into the membrane of target cells leading to permeabilization due to pore formation and consequent flux of ions across the membrane, resulting in membrane depolarization and eventual cell death.
Collapse
Affiliation(s)
- Harsh Mathur
- Moorepark Food Research CentreCounty Cork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | | | | | - Mary C Rea
- Moorepark Food Research CentreCounty Cork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Paul D Cotter
- Moorepark Food Research CentreCounty Cork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland
| | - Colin Hill
- APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| | - R Paul Ross
- Moorepark Food Research CentreCounty Cork, Ireland.,APC Microbiome Institute, University College CorkCork, Ireland.,School of Microbiology, University College CorkCork, Ireland
| |
Collapse
|
36
|
Bacteriocins: antibiotics in the age of the microbiome. Emerg Top Life Sci 2017; 1:55-63. [PMID: 33525813 DOI: 10.1042/etls20160015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/27/2017] [Accepted: 03/01/2017] [Indexed: 12/20/2022]
Abstract
Antibiotics have revolutionised the treatment of infectious disease and improved the lives of billions of people worldwide over many decades. With the rise in antimicrobial resistance (AMR) and corresponding lack of antibiotic development, we find ourselves in dire need of alternative treatments. Bacteriocins are a class of bacterially produced, ribosomally synthesised, antimicrobial peptides that may be narrow or broad in their spectra of activity. Animal models have demonstrated the safety and efficacy of bacteriocins in treating a broad range of infections; however, one of the principal drawbacks has been their relatively narrow spectra when compared with small-molecule antibiotics. In an era where we are beginning to appreciate the role of the microbiota in human and animal health, the fact that bacteriocins cause much less collateral damage to the host microbiome makes them a highly desirable therapeutic. This review makes a case for the implementation of bacteriocins as therapeutic antimicrobials, either alone or in combination with existing antibiotics to alleviate the AMR crisis and to lessen the impact of antibiotics on the host microbiome.
Collapse
|
37
|
Yu HY, Chen YA, Yip BS, Wang SY, Wei HJ, Chih YH, Chen KH, Cheng JW. Role of β-naphthylalanine end-tags in the enhancement of antiendotoxin activities: Solution structure of the antimicrobial peptide S1-Nal-Nal in complex with lipopolysaccharide. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1114-1123. [PMID: 28288781 DOI: 10.1016/j.bbamem.2017.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/08/2017] [Accepted: 03/09/2017] [Indexed: 12/18/2022]
Abstract
Lipopolysaccharide (LPS, endotoxin) is the major component of Gram-negative bacterial outer surface membrane. LPS released from bacteria into bloodstream during infection may cause serious unwanted stimulation of host's immune system and lead to septic shock of the patient. Recently, we have developed a strategy to increase salt resistance and LPS neutralization of short antimicrobial peptides by adding β-naphthylalanine end-tags to their termini. Herein, correlations between membrane immersion depth, orientation, and antiendotoxin activities of the antimicrobial peptides S1 and S1-Nal-Nal have been investigated via solution structure, paramagnetic resonance enhancement, and saturation transfer difference NMR studies. Unlike the parent peptide S1, S1-Nal-Nal rotated its two terminal β-naphthylalanine residues into the hydrophobic lipid A motif of LPS micelles. The LPS-induced inflammation may then be prohibited by the blocked lipid A motif.
Collapse
Affiliation(s)
- Hui-Yuan Yu
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Yi-An Chen
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Bak-Sau Yip
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan; Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu 300, Taiwan
| | - Siou-Ying Wang
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Hsiu-Ju Wei
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ya-Han Chih
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Kuan-Hao Chen
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jya-Wei Cheng
- Institute of Biotechnology and Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan.
| |
Collapse
|
38
|
Hanchi H, Hammami R, Gingras H, Kourda R, Bergeron MG, Ben Hamida J, Ouellette M, Fliss I. Inhibition of MRSA and of Clostridium difficile by durancin 61A: synergy with bacteriocins and antibiotics. Future Microbiol 2017; 12:205-212. [PMID: 28262046 DOI: 10.2217/fmb-2016-0113] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The aim of this study was to evaluate the efficacy of durancin 61A alone or in combination with nisin, pediocin PA-1, reuterin, microcin J25, vancomycin or tetracycline as an inhibitor of resistant clinical pathogens and to shed light on its mode of action. RESULTS Durancin and reuterin were effective inhibitors of Clostridium difficile, vancomycin-resistant Enterococcus faecium and methicillin-resistant Staphylococcus aureus. The combination of durancin and reuterin was highly synergistic against C. difficile (fractional inhibitory concentration index = 0.2). Durancin/vancomycin combination was synergistic against S. aureus ATCC® 700699 (fractional inhibitory concentration index = 0.3). Conclusion & future perspective: Durancin 61A alone or combined with other bacteriocins or antibiotics may therefore provide a possible therapeutic option for the treatment of infections by these pathogens.
Collapse
Affiliation(s)
- Hasna Hanchi
- STELA Dairy Research Centre, Institute of Nutrition & Functional Foods, Université Laval, G1K 7P4 Québec, QC, Canada.,Unité Protéomie Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El Manar, Tunisie
| | - Riadh Hammami
- STELA Dairy Research Centre, Institute of Nutrition & Functional Foods, Université Laval, G1K 7P4 Québec, QC, Canada.,Present address: Department of Soil Sciences and Agri-Food Engineering, Université Laval, Quebec, QC G1V 0A6, Canada
| | - Hélène Gingras
- Centre de Recherche en Infectiologie de l'Université Laval, CHUQ, Pavillon CHUL 2705 boul. Laurier, Ste-Foy, Quebec G1V 4G2, Canada
| | - Rim Kourda
- Unité Protéomie Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El Manar, Tunisie
| | - Michel G Bergeron
- Centre de Recherche en Infectiologie de l'Université Laval, CHUQ, Pavillon CHUL 2705 boul. Laurier, Ste-Foy, Quebec G1V 4G2, Canada
| | - Jeannette Ben Hamida
- Unité Protéomie Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El Manar, Tunisie
| | - Marc Ouellette
- Centre de Recherche en Infectiologie de l'Université Laval, CHUQ, Pavillon CHUL 2705 boul. Laurier, Ste-Foy, Quebec G1V 4G2, Canada
| | - Ismail Fliss
- STELA Dairy Research Centre, Institute of Nutrition & Functional Foods, Université Laval, G1K 7P4 Québec, QC, Canada
| |
Collapse
|
39
|
Smith MK, Draper LA, Hazelhoff PJ, Cotter PD, Ross RP, Hill C. A Bioengineered Nisin Derivative, M21A, in Combination with Food Grade Additives Eradicates Biofilms of Listeria monocytogenes. Front Microbiol 2016; 7:1939. [PMID: 27965658 PMCID: PMC5127793 DOI: 10.3389/fmicb.2016.01939] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 11/18/2016] [Indexed: 12/22/2022] Open
Abstract
The burden of foodborne disease has large economic and social consequences worldwide. Despite strict regulations, a number of pathogens persist within the food environment, which is greatly contributed to by a build-up of resistance mechanisms and also through the formation of biofilms. Biofilms have been shown to be highly resistant to a number of antimicrobials and can be extremely difficult to remove once they are established. In parallel, the growing concern of consumers regarding the use of chemically derived antimicrobials within food has led to a drive toward more natural products. As a consequence, the use of naturally derived antimicrobials has become of particular interest. In this study we investigated the efficacy of nisin A and its bioengineered derivative M21A in combination with food grade additives to treat biofilms of a representative foodborne disease isolate of Listeria monocytogenes. Investigations revealed the enhanced antimicrobial effects, in liquid culture, of M21A in combination with citric acid or cinnamaldehyde over its wild type nisin A counterpart. Subsequently, an investigation was conducted into the effects of these combinations on an established biofilm of the same strain. Nisin M21A (0.1 μg/ml) alone or in combination with cinnamaldehyde (35 μg/ml) or citric acid (175 μg/ml) performed significantly better than combinations involving nisin A. All combinations of M21A with either citric acid or cinnamaldehyde eradicated the L. monocytogenes biofilm (in relation to a non-biofilm control). We conclude that M21A in combination with available food additives could further enhance the antimicrobial treatment of biofilms within the food industry, simply by substituting nisin A with M21A in current commercial products such as Nisaplin® (Danisco, DuPont).
Collapse
Affiliation(s)
- Muireann K Smith
- APC Microbiome Institute, School of Microbiology, University College Cork Cork, Ireland
| | - Lorraine A Draper
- APC Microbiome Institute, School of Microbiology, University College Cork Cork, Ireland
| | | | - Paul D Cotter
- APC Microbiome Institute, School of Microbiology, University College CorkCork, Ireland; Teagasc Food Research CentreCork, Ireland
| | - R P Ross
- APC Microbiome Institute, School of Microbiology, University College Cork Cork, Ireland
| | - Colin Hill
- APC Microbiome Institute, School of Microbiology, University College Cork Cork, Ireland
| |
Collapse
|
40
|
Tillotson GS. Trojan Horse Antibiotics-A Novel Way to Circumvent Gram-Negative Bacterial Resistance? Infect Dis (Lond) 2016; 9:45-52. [PMID: 27773991 PMCID: PMC5063921 DOI: 10.4137/idrt.s31567] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 09/13/2016] [Accepted: 09/18/2016] [Indexed: 11/16/2022] Open
Abstract
Antibiotic resistance has been emerged as a major global health problem. In particular, gram-negative species pose a significant clinical challenge as bacteria develop or acquire more resistance mechanisms. Often, these bacteria possess multiple resistance mechanisms, thus nullifying most of the major classes of drugs. Novel approaches to this issue are urgently required. However, the challenges of developing new agents are immense. Introducing novel agents is fraught with hurdles, thus adapting known antibiotic classes by altering their chemical structure could be a way forward. A chemical addition to existing antibiotics known as a siderophore could be a solution to the gram-negative resistance issue. Siderophore molecules rely on the bacterial innate need for iron ions and thus can utilize a Trojan Horse approach to gain access to the bacterial cell. The current approaches to using this potential method are reviewed.
Collapse
|
41
|
Kaur S, Sharma P. Protease-Sensitive Inhibitory Activity of Cell-free Supernatant of Lactobacillus crispatus 156 Synergizes with Ciprofloxacin, Moxifloxacin and Streptomycin Against Pseudomonas aeruginosa: An In Vitro Study. Probiotics Antimicrob Proteins 2016; 7:172-80. [PMID: 25693845 DOI: 10.1007/s12602-015-9188-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Ciprofloxacin and streptomycin are frequently prescribed for the treatment of medical conditions originating due to infection by Pseudomonas aeruginosa. However, fluoroquinolone administration has been linked to the outgrowth of Clostridium difficile pathogen, especially in immunocompromised patients. Secondly, frequent administration of antibiotics may lead to development of resistance in the pathogens. Thus, there is a need to explore innovative adjunct therapies to lower the therapeutic doses of the antibiotics. Herein, we evaluated the synergism, if any, between conventional antibiotics and the cell-free supernatant (CFS) of vaginal Lactobacillus crispatus 156 against P. aeruginosa MTCC 741. L. crispatus 156 was isolated from the human vaginal tract, and its CFS had broad-spectrum antimicrobial activity against various Gram-positive and Gram-negative pathogens, including P. aeruginosa. The inhibitory substance present in the CFS completely lost its activity after treatment with proteinases and was resistant to temperatures up to 80 °C and pH ranging from 2 to 6. The cumulative production of the inhibitory substance in CFS was studied, and it showed that the secretion of the inhibitory substance was initiated in middle log phase of growth and peaked in late log phase. Further, CFS synergized the activities of ciprofloxacin, moxifloxacin, and streptomycin as evaluated in terms of checkerboard titrations. It lowered the minimum inhibitory concentration (MIC) of ciprofloxacin by almost 30 times and MIC of both moxifloxacin and streptomycin by 8 times. Interestingly, pepsin treatment of CFS caused the complete abrogation of its synergistic effect with all the three antibiotics. Thus, from the study, it can be concluded that probiotic-based alternative therapeutic regimen can be designed for the treatment of P. aeruginosa infections.
Collapse
Affiliation(s)
- Sukhraj Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, 143005, India,
| | | |
Collapse
|
42
|
Cai W, Fu Y, Zhang W, Chen X, Zhao J, Song W, Li Y, Huang Y, Wu Z, Sun R, Dong C, Zhang F. Synergistic effects of baicalein with cefotaxime against Klebsiella pneumoniae through inhibiting CTX-M-1 gene expression. BMC Microbiol 2016; 16:181. [PMID: 27502110 PMCID: PMC4977660 DOI: 10.1186/s12866-016-0797-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/02/2016] [Indexed: 02/28/2023] Open
Abstract
Background Generation of extended- spectrum β- lactamases is one of the major mechanisms by which clinical Klebsiella pneumoniae develop resistance to antibiotics. Combined antibiotics prove to be a relatively effective method of controlling such resistant strains. Some of Chinese herbal active ingredients are known to have synergistic antibacterial effects. This study is aimed to investigate synergistic effects of Chinese herbal active ingredients with cefotaxime on the extended- spectrum β- lactamase positive strains of Klebsiella pneumoniae, and to analyze mechanism of synergistic action, providing experimental evidence for clinical application of antimicrobial drugs. Results For total sixteen strains including fifteen strains of cefotaxime resistant K. pneumoniae and one extended- spectrum β- lactamase positive standard strain, the synergy rates of cefotaxime with baicalein, matrine, and clavulanic acid were 56.3 %, 0 %, and 100 %, respectively. The fractional inhibitory concentration index of combined baicalein and cefotaxime was correlated with the percentage decrease of cefotaxime MIC of all the strains (r = −0.78, p <0.01). In the group of synergy baicalein and cefotaxime, the transcribed mRNA level of CTX-M-1 after treatment of baicalein was decreased significantly (p <0.05). Moreover, the CTX-M-1 mRNA expression percentage inhibition (100 %, 5/5) was significantly higher than non- synergy group (25 %, 1/4) (p <0.05). Conclusions Our study demonstrated that baicalein exhibited synergistic activity when combined with cefotaxime against some of extended- spectrum β- lactamases positive K. pneumoniae strains by inhibiting CTX-M-1 mRNA expression. However, no direct bactericidal or bacteriostatic activity was involved in the synergistic action. Baicalein seems to be a promising novel effective synergistic antimicrobial agent. Electronic supplementary material The online version of this article (doi:10.1186/s12866-016-0797-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenhui Cai
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China.,Department of Microbiology and Immunology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yingmei Fu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Wenli Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China
| | - Xiaobei Chen
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Jizi Zhao
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Wuqi Song
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Yujun Li
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Ying Huang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China
| | - Zheng Wu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China
| | - Rui Sun
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China
| | - Chunping Dong
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China
| | - Fengmin Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, 157, Baojian Road, Nangang District, Harbin, 150081, China. .,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China.
| |
Collapse
|
43
|
Pseudomonas aeruginosa: targeting cell-wall metabolism for new antibacterial discovery and development. Future Med Chem 2016; 8:975-92. [PMID: 27228070 DOI: 10.4155/fmc-2016-0017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Pseudomonas aeruginosa is a leading cause of hospital-acquired infections and is resistant to most antibiotics. With therapeutic options against P. aeruginosa dwindling, and the lack of new antibiotics in advanced developmental stages, strategies for preserving the effectiveness of current antibiotics are urgently required. β-Lactam antibiotics are important agents for treating P. aeruginosa infections, thus, adjuvants that potentiate the activity of these compounds are desirable for extending their lifespan while new antibiotics - or antibiotic classes - are discovered and developed. In this review, we discuss recent research that has identified exploitable targets of cell-wall metabolism for the design and development of compounds that hinder resistance and potentiate the activity of antipseudomonal β-lactams.
Collapse
|
44
|
Baindara P, Chaudhry V, Mittal G, Liao LM, Matos CO, Khatri N, Franco OL, Patil PB, Korpole S. Characterization of the Antimicrobial Peptide Penisin, a Class Ia Novel Lantibiotic from Paenibacillus sp. Strain A3. Antimicrob Agents Chemother 2016; 60:580-91. [PMID: 26574006 PMCID: PMC4704198 DOI: 10.1128/aac.01813-15] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/08/2015] [Indexed: 11/20/2022] Open
Abstract
Attempts to isolate novel antimicrobial peptides from microbial sources have been on the rise recently, despite their low efficacy in therapeutic applications. Here, we report identification and characterization of a new efficient antimicrobial peptide from a bacterial strain designated A3 that exhibited highest identity with Paenibacillus ehimensis. Upon purification and subsequent molecular characterization of the antimicrobial peptide, referred to as penisin, we found the peptide to be a bacteriocin-like peptide. Consistent with these results, RAST analysis of the entire genome sequence revealed the presence of a lantibiotic gene cluster containing genes necessary for synthesis and maturation of a lantibiotic. While circular dichroism and one-dimension nuclear magnetic resonance experiments confirmed a random coil structure of the peptide, similar to other known lantibiotics, additional biochemical evidence suggests posttranslational modifications of the core peptide yield six thioether cross-links. The deduced amino acid sequence of the putative biosynthetic gene penA showed approximately 74% similarity with elgicin A and 50% similarity with the lantibiotic paenicidin A. Penisin effectively killed methicillin-resistant Staphylococcus aureus (MRSA) and did not exhibit hemolysis activity. Unlike other lantibiotics, it effectively inhibited the growth of Gram-negative bacteria. Furthermore, 80 mg/kg of body weight of penisin significantly reduced bacterial burden in a mouse thigh infection model and protected BALB/c mice in a bacteremia model entailing infection with Staphylococcus aureus MTCC 96, suggesting that it could be a promising new antimicrobial peptide.
Collapse
Affiliation(s)
| | - Vasvi Chaudhry
- CSIR Institute of Microbial Technology, Chandigarh, India
| | - Garima Mittal
- CSIR Institute of Microbial Technology, Chandigarh, India
| | - Luciano M Liao
- Institute of Chemistry, Federal University of Goiás, Goiânia, Brazil
| | - Carolina O Matos
- Institute of Chemistry, Federal University of Goiás, Goiânia, Brazil
| | - Neeraj Khatri
- CSIR Institute of Microbial Technology, Chandigarh, India
| | - Octavio L Franco
- Centro de Analises Proteomicas e Bioquimicas, Pós-graduacão em Ciências Genomicas e Biotecnologia, Brasília, Brazil S-Inova, Programa de Pós-Graduacão em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Prabhu B Patil
- CSIR Institute of Microbial Technology, Chandigarh, India
| | - Suresh Korpole
- CSIR Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
45
|
Rajamuthiah R, Jayamani E, Majed H, Conery AL, Kim W, Kwon B, Fuchs BB, Kelso MJ, Ausubel FM, Mylonakis E. Antibacterial properties of 3-(phenylsulfonyl)-2-pyrazinecarbonitrile. Bioorg Med Chem Lett 2015; 25:5203-7. [PMID: 26459212 PMCID: PMC4718707 DOI: 10.1016/j.bmcl.2015.09.066] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/23/2015] [Accepted: 09/28/2015] [Indexed: 11/30/2022]
Abstract
The emergence of multidrug-resistant bacterial strains has heightened the need for new antimicrobial agents based on novel chemical scaffolds that are able to circumvent current modes of resistance. We recently developed a whole-animal drug-screening methodology in pursuit of this goal and now report the discovery of 3-(phenylsulfonyl)-2-pyrazinecarbonitrile (PSPC) as a novel antibacterial effective against resistant nosocomial pathogens. The minimum inhibitory concentrations (MIC) of PSPC against Staphylococcus aureus and Enterococcus faecium were 4 μg/mL and 8 μg/mL, respectively, whereas the MICs were higher against the Gram-negative bacteria Klebsiella pneumoniae (64 μg/mL), Acinetobacter baumannii (32 μg/mL), Pseudomonas aeruginosa (>64 μg/mL), and Enterobacter spp. (>64 μg/mL). However, co-treatment of PSPC with the efflux pump inhibitor phenylalanine arginyl β-naphthylamide (PAβN) or with sub-inhibitory concentrations of the lipopeptide antibiotic polymyxin B reduced the MICs of PSPC against the Gram-negative strains by >4-fold. A sulfide analog of PSPC (PSPC-1S) showed no antibacterial activity, whereas the sulfoxide analog (PSPC-6S) showed identical activity as PSPC across all strains, confirming structure-dependent activity for PSPC and suggesting a target-based mechanism of action. PSPC displayed dose dependent toxicity to both Caenorhabditis elegans and HEK-293 mammalian cells, culminating with a survival rate of 16% (100 μg/mL) and 8.5% (64 μg/mL), respectively, at the maximum tested concentration. However, PSPC did not result in hemolysis of erythrocytes, even at a concentration of 64 μg/mL. Together these results support PSPC as a new chemotype suitable for further development of new antibiotics against Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
- Rajmohan Rajamuthiah
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, POB, 3rd Floor, Suite 328/330, 593 Eddy Street, Providence, RI 02903, USA; Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Elamparithi Jayamani
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, POB, 3rd Floor, Suite 328/330, 593 Eddy Street, Providence, RI 02903, USA; Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Hiwa Majed
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; School of Chemistry, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Annie L Conery
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Wooseong Kim
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, POB, 3rd Floor, Suite 328/330, 593 Eddy Street, Providence, RI 02903, USA
| | - Bumsup Kwon
- Division of Neurology, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, POB, 3rd Floor, Suite 328/330, 593 Eddy Street, Providence, RI 02903, USA
| | - Michael J Kelso
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia; School of Chemistry, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Frederick M Ausubel
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, POB, 3rd Floor, Suite 328/330, 593 Eddy Street, Providence, RI 02903, USA; Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
46
|
Cavera VL, Arthur TD, Kashtanov D, Chikindas ML. Bacteriocins and their position in the next wave of conventional antibiotics. Int J Antimicrob Agents 2015; 46:494-501. [PMID: 26341839 DOI: 10.1016/j.ijantimicag.2015.07.011] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/10/2015] [Accepted: 07/15/2015] [Indexed: 12/13/2022]
Abstract
Micro-organisms are capable of producing a range of defence mechanisms, including antibiotics, bacteriocins, lytic agents, protein exotoxins, etc. Such mechanisms have been identified in nearly 99% of studied bacteria. The multiplicity and diversity of bacteriocins and the resultant effects of their interactions with targeted bacteria on microbial ecology has been thoroughly studied and remains an area of investigation attracting many researchers. However, the incorporation of bacteriocins into drug delivery systems used in conjunction with, or as potential alternatives to, conventional antibiotics is only a recent, although rapidly expanding, field. The extensive array of bacteriocins positions them as one of the most promising options in the next wave of antibiotics. The goal of this review was to explore bacteriocins as novel antimicrobials, alone and in combination with established antibiotics, and thus position them as a potential tool for addressing the current antibiotic crisis.
Collapse
Affiliation(s)
- Veronica L Cavera
- Department of Biochemistry and Microbiology, Rutgers State University, 76 Lipman Drive, New Brunswick, NJ 08901, USA
| | - Timothy D Arthur
- Department of Biochemistry and Microbiology, Rutgers State University, 76 Lipman Drive, New Brunswick, NJ 08901, USA
| | - Dimitri Kashtanov
- School of Environmental and Biological Sciences, Rutgers State University, 65 Dudley Road, New Brunswick, NJ 08901, USA
| | - Michael L Chikindas
- School of Environmental and Biological Sciences, Rutgers State University, 65 Dudley Road, New Brunswick, NJ 08901, USA.
| |
Collapse
|
47
|
Rajamuthiah R, Jayamani E, Conery AL, Fuchs BB, Kim W, Johnston T, Vilcinskas A, Ausubel FM, Mylonakis E. A Defensin from the Model Beetle Tribolium castaneum Acts Synergistically with Telavancin and Daptomycin against Multidrug Resistant Staphylococcus aureus. PLoS One 2015; 10:e0128576. [PMID: 26062137 PMCID: PMC4465704 DOI: 10.1371/journal.pone.0128576] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 04/28/2015] [Indexed: 01/19/2023] Open
Abstract
The red flour beetle Tribolium castaneum is a common insect pest and has been established as a model beetle to study insect development and immunity. This study demonstrates that defensin 1 from T. castaneum displays in vitro and in vivo antimicrobial activity against drug resistant Staphylococcus aureus strains. The minimum inhibitory concentration (MIC) of defensin 1 against 11 reference and clinical staphylococcal isolates was between 16–64 μg/ml. The putative mode of action of the defensin peptide is disruption of the bacterial cell membrane. The antibacterial activity of defensin 1 was attenuated by salt concentrations of 1.56 mM and 25 mM for NaCl and CaCl2 respectively. Treatment of defensin 1 with the reducing agent dithiothreitol (DTT) at concentrations 1.56 to 3.13 mM abolished the antimicrobial activity of the peptide. In the presence of subinhibitory concentrations of antibiotics that also target the bacterial cell envelope such as telavancin and daptomycin, the MIC of the peptide was as low as 1 μg/ml. Moreover, when tested against an S. aureus strain that was defective in D-alanylation of the cell wall, the MIC of the peptide was 0.5 μg/ml. Defensin 1 exhibited no toxicity against human erythrocytes even at 400 μg/ml. The in vivo activity of the peptide was validated in a Caenorhabditis elegans-MRSA liquid infection assay. These results suggest that defensin 1 behaves similarly to other cationic AMPs in its mode of action against S. aureus and that the activity of the peptide can be enhanced in combination with other antibiotics with similar modes of action or with compounds that have the ability to decrease D-alanylation of the bacterial cell wall.
Collapse
Affiliation(s)
- Rajmohan Rajamuthiah
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Elamparithi Jayamani
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Annie L. Conery
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
| | - Wooseong Kim
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
| | - Tatiana Johnston
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
| | - Andreas Vilcinskas
- Institute of Phytopathology and Applied Zoology, Justus-Liebig University, Heinrich-Buff-Ring 26–32, 35392, Giessen, Germany
| | - Frederick M. Ausubel
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
- * E-mail:
| |
Collapse
|
48
|
Ultrashort Antimicrobial Peptides with Antiendotoxin Properties. Antimicrob Agents Chemother 2015; 59:5052-6. [PMID: 26033727 DOI: 10.1128/aac.00519-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 05/25/2015] [Indexed: 11/20/2022] Open
Abstract
Release of lipopolysaccharide (LPS) (endotoxin) from bacteria into the bloodstream may cause serious unwanted stimulation of the host immune system. Some but not all antimicrobial peptides can neutralize LPS-stimulated proinflammatory responses. Salt resistance and serum stability of short antimicrobial peptides can be boosted by adding β-naphthylalanine to their termini. Herein, significant antiendotoxin effects were observed in vitro and in vivo with the β-naphthylalanine end-tagged variants of the short antimicrobial peptides S1 and KWWK.
Collapse
|
49
|
Rumjuankiat K, Perez RH, Pilasombut K, Keawsompong S, Zendo T, Sonomoto K, Nitisinprasert S. Purification and characterization of a novel plantaricin, KL-1Y, from Lactobacillus plantarum KL-1. World J Microbiol Biotechnol 2015; 31:983-94. [PMID: 25862353 DOI: 10.1007/s11274-015-1851-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 04/03/2015] [Indexed: 11/30/2022]
Abstract
Three bacteriocins from Lactobacillus plantarum KL-1 were successfully purified using ammonium sulfate precipitation, cation-exchange chromatography and reverse-phase HPLC. The bacteriocin peptides KL-1X, -1Y and -1Z had molecular masses of 3053.82, 3498.16 and 3533.16 Da, respectively. All three peptides were stable at pH 2-12 and 25 °C and at high temperatures of 80 and 100 °C for 30 min and 121 °C for 15 min. However, they differed in their susceptibility to proteolytic enzymes and their inhibition spectra. KL-1Y showed broad inhibitory activities against Gram-positive and Gram-negative bacteria, including Salmonella enterica serovar Enteritidis DMST 17368, Pseudomonas aeruginosa ATCC 15442, P. aeruginosa ATCC 9027, Escherichia coli O157:H7 and E. coli ATCC 8739. KL-1X and -1Z inhibited only Gram-positive bacteria. KL-1X, KL-1Y and KL-1Z exhibited synergistic activity. The successful amino acid sequencing of KL-1Y had a hydrophobicity of approximately 30 % and no cysteine residues suggested its novelty, and it was designated "plantaricin KL-1Y". Plantaricin KL-1Y exhibited bactericidal activity against Bacillus cereus JCM 2152(T). Compared to nisin, KL-1Y displayed broad inhibitory activities of 200, 800, 1600, 800, 400 and 400 AU/mL against the growth of Bacillus coagulans JCM 2257(T), B. cereus JCM 2152(T), Listeria innocua ATCC 33090(T), Staphylococcus aureus TISTR 118, E. coli O157:H7 and E. coli ATCC 8739, respectively, whereas nisin had similar activities against only B. coagulans JCM 2257(T) and B. cereus JCM 2152(T). Therefore, the novel plantaricin KL-1Y is a promising antimicrobial substance for food safety uses in the future.
Collapse
Affiliation(s)
- Kittaporn Rumjuankiat
- Specialized Research Unit: Prebiotics and Probiotics for Health, Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Lat Yao, Chatuchak, Bangkok, 10900, Thailand
| | | | | | | | | | | | | |
Collapse
|
50
|
Algburi A, Volski A, Chikindas ML. Natural antimicrobials subtilosin and lauramide arginine ethyl ester synergize with conventional antibiotics clindamycin and metronidazole against biofilms of Gardnerella vaginalis but not against biofilms of healthy vaginal lactobacilli. Pathog Dis 2015; 73:ftv018. [PMID: 25838136 DOI: 10.1093/femspd/ftv018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2015] [Indexed: 11/12/2022] Open
Abstract
The purpose of this study was to evaluate the ability of clindamycin and metronidazole to synergize with natural antimicrobials against biofilms of bacterial vaginosis (BV)-associated Gardnerella vaginalis. Minimum bactericidal concentrations for biofilm cells (MBCs-B) were determined for each antimicrobial. The MBCs-B of lauramide arginine ethyl ester (LAE), subtilosin, clindamycin and metronidazole were 50, 69.5, 20 and 500 μg mL(-1), respectively. A checkerboard assay and isobologram were used to analyze the type of interactions between these antimicrobials. The combination of metronidazole with natural antimicrobials did not inhibit planktonic lactobacilli. Clindamycin with either LAE or with subtilosin was inhibitory for planktonic but not for biofilm-associated lactobacilli. All tested antimicrobial combinations were inhibitory for BV-associated Mobiluncus curtisii and Peptostreptococcus anaerobius. LAE and subtilosin synergized with clindamycin and metronidazole against biofilms of G. vaginalis but not biofilm-associated vaginal lactobacilli. The biofilms of BV-associated pathogens can be controlled by synergistically acting combinations of conventional antibiotics and natural antimicrobials which will help better management of current antibiotics, especially considering robust bacterial resistance. Our findings create a foundation for a new strategy in the effective control of vaginal infections.
Collapse
Affiliation(s)
- Ammar Algburi
- Department of Biochemistry and Microbiology, Rutgers State University, New Brunswick, NJ 08901, USA Department of Microbiology, Veterinary College, Diyala University, Baqubah, Iraq
| | - Anna Volski
- School of Arts and Science, Rutgers State University, New Brunswick, NJ 08901, USA
| | - Michael L Chikindas
- School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ 08901, USA
| |
Collapse
|