1
|
Girdhar M, Sen A, Nigam A, Oswalia J, Kumar S, Gupta R. Antimicrobial peptide-based strategies to overcome antimicrobial resistance. Arch Microbiol 2024; 206:411. [PMID: 39311963 DOI: 10.1007/s00203-024-04133-x] [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: 07/13/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
Antibiotic resistance has emerged as a global threat, rendering the existing conventional treatment strategies ineffective. In view of this, antimicrobial peptides (AMPs) have proven to be potent alternative therapeutic interventions with a wide range of applications in clinical health. AMPs are small peptides produced naturally as a part of the innate immune responses against a broad range of bacterial, fungal and viral pathogens. AMPs present a myriad of advantages over traditional antibiotics, including their ability to target multiple sites, reduced susceptibility to resistance development, and high efficacy at low doses. These peptides have demonstrated notable potential in inhibiting microbes resistant to traditional antibiotics, including the notorious ESKAPE pathogens, recognized as the primary culprits behind nosocomial infections. AMPs, with their multifaceted benefits, emerge as promising candidates in the ongoing efforts to combat the escalating challenges posed by antibiotic resistance. This in-depth review provides a detailed discussion on AMPs, encompassing their classification, mechanism of action, and diverse clinical applications. Focus has been laid on combating newly emerging drug-resistant organisms, emphasizing the significance of AMPs in mitigating this pressing challenge. The review also illuminates potential future strategies that may be implemented to improve AMP efficacy, such as structural modifications and using AMPs in combination with antibiotics and matrix-inhibiting compounds.
Collapse
Affiliation(s)
| | - Aparajita Sen
- Department of Genetics, University of Delhi, South Campus, New Delhi, 110021, India
| | - Arti Nigam
- Department of Microbiology, Institute of Home Economics, University of Delhi, New Delhi, 110016, India
| | - Jyoti Oswalia
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sachin Kumar
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India
| | - Rashi Gupta
- Department of Medical Laboratory Technology, School of Allied Health Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India.
| |
Collapse
|
2
|
Elafify M, Liao X, Feng J, Ahn J, Ding T. Biofilm formation in food industries: Challenges and control strategies for food safety. Food Res Int 2024; 190:114650. [PMID: 38945629 DOI: 10.1016/j.foodres.2024.114650] [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: 04/18/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024]
Abstract
Various pathogens have the ability to grow on food matrices and instruments. This grow may reach to form biofilms. Bacterial biofilms are community of microorganisms embedded in extracellular polymeric substances (EPSs) containing lipids, DNA, proteins, and polysaccharides. These EPSs provide a tolerance and favorable living condition for microorganisms. Biofilm formations could not only contribute a risk for food safety but also have negative impacts on healthcare sector. Once biofilms form, they reveal resistances to traditional detergents and disinfectants, leading to cross-contamination. Inhibition of biofilms formation and abolition of mature biofilms is the main target for controlling of biofilm hazards in the food industry. Some novel eco-friendly technologies such as ultrasound, ultraviolet, cold plasma, magnetic nanoparticles, different chemicals additives as vitamins, D-amino acids, enzymes, antimicrobial peptides, and many other inhibitors provide a significant value on biofilm inhibition. These anti-biofilm agents represent promising tools for food industries and researchers to interfere with different phases of biofilms including adherence, quorum sensing molecules, and cell-to-cell communication. This perspective review highlights the biofilm formation mechanisms, issues associated with biofilms, environmental factors influencing bacterial biofilm development, and recent strategies employed to control biofilm-forming bacteria in the food industry. Further studies are still needed to explore the effects of biofilm regulation in food industries and exploit more regulation strategies for improving the quality and decreasing economic losses.
Collapse
Affiliation(s)
- Mahmoud Elafify
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; Department of Food Hygiene and Control, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Xinyu Liao
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
| | - Jinsong Feng
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Juhee Ahn
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; Department of Biomedical Science, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea.
| | - Tian Ding
- Future Food Laboratory, Innovative Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China; College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
3
|
Pereira ACC, Aguiar APS, Barbosa VL, Régis JR, Miyazima EM, Araujo LMP, Dantas LO, Mayer MPA, Andrade FB, Karygianni L, Pinheiro ET. Enhancing Antibiotic Efficacy in Regenerative Endodontics by Improving Biofilm Susceptibility. J Endod 2024; 50:962-965. [PMID: 38615826 DOI: 10.1016/j.joen.2024.04.004] [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: 11/23/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Various strategies have been researched to enhance the susceptibility of biofilms, given their tolerance to antibiotics. This study evaluated the effect of the anti-microbial peptide nisin in association with antibiotics used in regenerative endodontics, exploring different treatment times and biofilm growth conditions. METHODS A mixture of 10 bacterial species was cultivated on dentin specimens anaerobically for 21 days. Biofilms were treated with 1 mL of high-purity nisin Z (nisin ZP, 200 μg/mL) and a triple antibiotic mixture (TAP: ciprofloxacin + metronidazole + minocycline, 5 mg/mL), alone or in combination. The effectiveness of antimicrobial agents was assessed after 1 and 7 days. During the 7-day period, biofilms were treated under 2 conditions: a single dose in a nutrient-depleted setting (ie, no replenishment of growth medium) and multiple doses in a nutrient-rich environment (ie, renewal of medium and antimicrobial agents every 48 h). After treatments, biofilm cells were dispersed, and total colony-forming units were counted. RESULTS After 1 d-treatment, nisin ZP + TAP resulted in 2-log cell reduction compared to TAP alone (P < .05). After 7 d-treatment with a single dose, nisin ZP + TAP and TAP reduced bacteria to nonculturable levels (P < .05), whereas repeated antimicrobial doses did not eliminate bacteria in a nutrient-rich environment. No bacterial reduction was observed with nisin ZP alone in any treatment time. CONCLUSIONS The additional use of nisin improved the TAP activity only after a short exposure time. Longer exposure to TAP or nisin + TAP in a nutrient-deprived environment effectively eliminated biofilms.
Collapse
Affiliation(s)
- Ana C C Pereira
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Alana P S Aguiar
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Vinícius L Barbosa
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Júlia R Régis
- Department of Dentistry, Federal University of Santa Catarina, Florianopolis, Santa Catarina, Brazil
| | - Erica Mina Miyazima
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Leticia M P Araujo
- Department of Biomaterials and Oral Biology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Larissa O Dantas
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Marcia P A Mayer
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Flaviana B Andrade
- Department of Operative Dentistry, Endodontics and Dental Materials, Bauru School of Dentistry, University of São Paulo, Bauru, São Paulo, Brazil
| | - Lamprini Karygianni
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine, University of Zurich, Zürich, Switzerland
| | - Ericka T Pinheiro
- Department of Dentistry, School of Dentistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
4
|
Zhang M, Yang B, Shi J, Wang Z, Liu Y. Host defense peptides mitigate the spread of antibiotic resistance in physiologically relevant condition. Antimicrob Agents Chemother 2024; 68:e0126123. [PMID: 38415983 PMCID: PMC10994823 DOI: 10.1128/aac.01261-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Antibiotic resistance represents a significant challenge to public health and human safety. The primary driver behind the dissemination of antibiotic resistance is the horizontal transfer of plasmids. Current conjugative transfer assay is generally performed in a standardized manner, ignoring the effect of the host environment. Host defense peptides (HDPs) possess a wide range of biological targets and play an essential role in the innate immune system. Herein, we reveal that sub-minimum inhibitory concentrations of HDPs facilitate the conjugative transfer of RP4-7 plasmid in the Luria Broth medium, and this observation is reversed in the RPMI medium, designed to simulate the host environment. Out of these HDPs, indolicidin (Ind), a cationic tridecapeptide from bovine neutrophils, significantly inhibits the conjugation of multidrug resistance plasmids in a dose-dependent manner, including blaNDM- and tet(X4)-bearing plasmids. We demonstrate that the addition of Ind to RPMI medium as the incubation substrate downregulates the expression of conjugation-related genes. In addition, Ind weakens the tricarboxylic acid cycle, impedes the electron transport chain, and disrupts the proton motive force, consequently diminishing the synthesis of adenosine triphosphate and limiting the energy supply. Our findings highlight the importance of the host-like environments for the development of horizontal transfer inhibitors and demonstrate the potential of HDPs in preventing the spread of resistance plasmids.
Collapse
Affiliation(s)
- Miao Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Bingqing Yang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jingru Shi
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
5
|
Bilgin M, Dosler S, Otuk G. Antibiotic adjuvant activities of quorum sensing signal molecules DSF and BDSF against mature biofilms of Staphylococci. J Chemother 2024; 36:11-23. [PMID: 37873740 DOI: 10.1080/1120009x.2023.2270743] [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: 01/21/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023]
Abstract
Among promising antibiofilm compounds, quorum-sensing (QS) molecules that regulate biological processes such as biofilm formation and intra- or interspecies communication appear to be good candidates. The invitro antibiotic-adjuvant effects of QS molecules diffusible signal factor (DSF) and B. cenocepacia producing-DSF (BDSF) were investigated against mature Staphylococcal biofilms. Broth microdilution methods were used for the determinations of MIC, MBC, MBIC, and MBEC, and bactericidal activities were determined by TKC method. The lowest MICs were obtained with ciprofloxacin and gentamicin, and MBECs with ciprofloxacin. DSF and BDSF at 0.5 µM decreased the MICs as 2-8, and 2-32 fold, respectively. In TKC studies, -cidal activities were achieved by BDSF + gentamycin, or ciprofloxacin, and DSF + daptomycin, vancomycin, meropenem or gentamycin combinations. Synergistic effects were generally obtained with BDSF + gentamicin combinations, followed by DSF + daptomycin against most S. aureus; while BDSF + gentamicin or ciprofloxacin, and DSF + vancomycin or meropenem were synergist against some S. epidermidis biofilms. Also, the antagonist effects were observed with BDSF + meropenem or ciprofloxacin against each MSSE and MSSA. It is estimated that these QS molecules, although it was strain dependent, generally enhanced the antibiotic activity, and would be a new and effective treatment strategy for biofilm control, either alone or as an antibiotic adjuvant.
Collapse
Affiliation(s)
- Merve Bilgin
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Institute of Graduate Studies in Health Sciences, Istanbul, Turkiye
- Department of Pharmaceutical Microbiology, Istanbul Health & Technology University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Sibel Dosler
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| | - Gulten Otuk
- Department of Pharmaceutical Microbiology, Istanbul, Istanbul University, Faculty of Pharmacy, Istanbul, Turkiye
| |
Collapse
|
6
|
Akhash N, Farajzadeh Sheikh A, Farshadzadeh Z. Design of a novel analogue peptide with potent antibiofilm activities against Staphylococcus aureus based upon a sapecin B-derived peptide. Sci Rep 2024; 14:2256. [PMID: 38278972 PMCID: PMC10817945 DOI: 10.1038/s41598-024-52721-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/23/2024] [Indexed: 01/28/2024] Open
Abstract
Nowadays, antimicrobial peptides are promising to confront the existing global crisis of antibiotic resistance. Here, a novel analogue peptide (mKLK) was designed based upon a D-form amidated sapecin B-derived peptide (KLK) by replacing two lysine residues with two tryptophan and one leucine by lysine, and inserting one alanine. The mKLK displayed superior amphipathic helixes in which the most of hydrophobic residues are confined to one face of the helix and had a higher hydrophobic moment compared with KLK. The mKLK retained its antibacterial activity and structure in human serum, suggesting its stability to proteolytic degradation. The values of MIC and MBC for mKLK were equal to those of KLK against clinical strains of methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-susceptible Staphylococcus aureus (MSSA). However, mKLK showed more capability of in vitro inhibiting, eradicating, and dispersing MRSA and MSSA biofilms compared with KLK. Furthermore, a remarkable inhibitory activity of mKLK against MRSA and MSSA biofilms was seen in the murine model of catheter-associated biofilm infection. Results of this study show that mKLK not only exhibits antibacterial activity and serum stability but also a potent biofilm inhibitory activity at sub-MIC concentrations, confirming its potential therapeutic advantage for preventing biofilm-associated MRSA and MSSA infections.
Collapse
Affiliation(s)
- Nasim Akhash
- Health Research Institute, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Farajzadeh Sheikh
- Health Research Institute, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Farshadzadeh
- Health Research Institute, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
7
|
Sharafi T, Ghaemi EA, Rafiee M, Ardebili A. Combination antimicrobial therapy: in vitro synergistic effect of anti-staphylococcal drug oxacillin with antimicrobial peptide nisin against Staphylococcus epidermidis clinical isolates and Staphylococcus aureus biofilms. Ann Clin Microbiol Antimicrob 2024; 23:7. [PMID: 38245727 PMCID: PMC10800071 DOI: 10.1186/s12941-024-00667-6] [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: 09/27/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
The ability of Staphylococcus epidermidis and S. aureus to form strong biofilm on plastic devices makes them the major pathogens associated with device-related infections (DRIs). Biofilm-embedded bacteria are more resistant to antibiotics, making biofilm infections very difficult to effectively treat. Here, we evaluate the in vitro activities of anti-staphylococcal drug oxacillin and antimicrobial peptide nisin, alone and in combination, against methicillin-resistant S. epidermidis (MRSE) clinical isolates and the methicillin-resistant S. aureus ATCC 43,300. The minimum inhibitory concentrations (MIC) and minimum biofilm eradication concentrations (MBEC) of oxacillin and nisin were determined using the microbroth dilution method. The anti-biofilm activities of oxacillin and nisin, alone or in combination, were evaluated. In addition, the effects of antimicrobial agents on the expression of icaA gene were examined by quantitative real-time PCR. MIC values for oxacillin and nisin ranged 4-8 µg/mL and 64-128 µg/mL, respectively. Oxacillin and nisin reduced biofilm biomass in all bacteria in a dose-dependent manner and this inhibitory effect was enhanced with combinatorial treatment. MBEC ranges for oxacillin and nisin were 2048-8192 µg/mL and 2048-4096 µg/mL, respectively. The addition of nisin significantly decreased the oxacillin MBECs from 8- to 32-fold in all bacteria. At the 1× MIC and 1/2× MIC, both oxacillin and nisin decreased significantly the expression of icaA gene in comparison with untreated control. When two antimicrobial agents were combined at 1/2× MIC concentration, the expression of icaA were significantly lower than when were used alone. Nisin/conventional oxacillin combination showed considerable anti-biofilm effects, including inhibition of biofilm formation, eradication of mature biofilm, and down-regulation of biofilm-related genes, proposing its applications for treating or preventing staphylococcal biofilm-associated infections, including device-related infections.
Collapse
Affiliation(s)
- Toktam Sharafi
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ezzat Allah Ghaemi
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Rafiee
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Abdollah Ardebili
- Infectious Disease Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
8
|
Alfei S. Shifting from Ammonium to Phosphonium Salts: A Promising Strategy to Develop Next-Generation Weapons against Biofilms. Pharmaceutics 2024; 16:80. [PMID: 38258091 PMCID: PMC10819902 DOI: 10.3390/pharmaceutics16010080] [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: 11/24/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Since they are difficult and sometimes impossible to treat, infections sustained by multidrug-resistant (MDR) pathogens, emerging especially in nosocomial environments, are an increasing global public health concern, translating into high mortality and healthcare costs. In addition to having acquired intrinsic abilities to resist available antibiotic treatments, MDR bacteria can transmit genetic material encoding for resistance to non-mutated bacteria, thus strongly decreasing the number of available effective antibiotics. Moreover, several pathogens develop resistance by forming biofilms (BFs), a safe and antibiotic-resistant home for microorganisms. BFs are made of well-organized bacterial communities, encased and protected in a self-produced extracellular polymeric matrix, which impedes antibiotics' ability to reach bacteria, thus causing them to lose efficacy. By adhering to living or abiotic surfaces in healthcare settings, especially in intensive care units where immunocompromised older patients with several comorbidities are hospitalized BFs cause the onset of difficult-to-eradicate infections. In this context, recent studies have demonstrated that quaternary ammonium compounds (QACs), acting as membrane disruptors and initially with a low tendency to develop resistance, have demonstrated anti-BF potentialities. However, a paucity of innovation in this space has driven the emergence of QAC resistance. More recently, quaternary phosphonium salts (QPSs), including tri-phenyl alkyl phosphonium derivatives, achievable by easy one-step reactions and well known as intermediates of the Wittig reaction, have shown promising anti-BF effects in vitro. Here, after an overview of pathogen resistance, BFs, and QACs, we have reviewed the QPSs developed and assayed to this end, so far. Finally, the synthetic strategies used to prepare QPSs have also been provided and discussed to spur the synthesis of novel compounds of this class. We think that the extension of the knowledge about these materials by this review could be a successful approach to finding effective weapons for treating chronic infections and device-associated diseases sustained by BF-producing MDR bacteria.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy, University of Genoa, Viale Cembrano, 4, 16148 Genova, Italy
| |
Collapse
|
9
|
Soares RS, Gomes D, Serrano I, Cunha E, Tavares L, Oliveira M. Absence of Synergism between a Dual-AMP Biogel and Antibiotics Used as Therapeutic Agents for Diabetic Foot Infections. Int J Mol Sci 2023; 25:407. [PMID: 38203579 PMCID: PMC10779294 DOI: 10.3390/ijms25010407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetic foot infections (DFIs) are frequently linked to diabetic-related morbidity and death because of the ineffectiveness of conventional antibiotics against multidrug-resistant bacteria. Pexiganan and nisin A are antimicrobial peptides (AMPs), and their application may complement conventional antibiotics in DFI treatment. A collagen 3D model, previously established to mimic a soft-tissue collagen matrix, was used to evaluate the antibacterial efficacy of a guar gum gel containing pexiganan and nisin alone and combined with three antimicrobials toward the biofilms of Staphylococcus aureus and Pseudomonas aeruginosa isolated from infected foot ulcers. Antimicrobials and bacterial diffusion were confirmed by spot-on-lawn and bacterial growth by bacterial count (cfu/mL). Our main conclusion was that the dual-AMP biogel combined with gentamicin, clindamycin, or vancomycin was not able to significantly reduce bacterial growth or eradicate S. aureus and P. aeruginosa DFI isolates. We further reported an antagonism between dual-AMP and dual-AMP combined with antibiotics against S. aureus.
Collapse
Affiliation(s)
- Rui Silva Soares
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; (R.S.S.); (D.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Diana Gomes
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; (R.S.S.); (D.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Isa Serrano
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; (R.S.S.); (D.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Eva Cunha
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; (R.S.S.); (D.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Luís Tavares
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; (R.S.S.); (D.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Manuela Oliveira
- CIISA—Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; (R.S.S.); (D.G.); (E.C.); (L.T.); (M.O.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
- cE3c—Centre for Ecology, Evolution and Environmental Changes & CHANGE—Global Change and Sustainability Institute, Campo Grande 016, 1749-016 Lisbon, Portugal
| |
Collapse
|
10
|
Kim N, Sengupta S, Lee J, Dash U, Kim S, Kim HJ, Song C, Sim T. Synthesis and antibacterial activities of baulamycin A inspired derivatives. Eur J Med Chem 2023; 259:115592. [PMID: 37478559 DOI: 10.1016/j.ejmech.2023.115592] [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: 04/04/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/23/2023]
Abstract
SbnE is an essential enzyme for staphyloferrin B biosynthesis in Staphylococcus aureus. An earlier study showed that natural product baulamycin A has in vitro inhibitory activity against SbnE and antibacterial potency. A SAR study with analogues of baulamycin A was conducted to identify potent inhibitors of SbnE and/or effective antibiotics against MRSA. The results show that selected analogues, including 11, 18, 21, 24a, 24c, 24m and 24n, exhibit single-digit micromolar inhibitory potencies against SbnE (IC50s = 1.81-8.94 μM) and 11, 24m, 24n possess significant activities against both SbnE (IC50s = 4.12-6.12 μM) and bacteria (MICs = 4-32 μg/mL). Biological investigations revealed that these substances possess potent cell wall disruptive activities and that they inhibit siderophore production in MRSA. Among the selected analogues, 7 has excellent antibiotic activities both gram-positive and -negative bacteria (0.5-4 μg/mL). Moreover, these analogues significantly impede biofilm formation in a concentration-dependent manner. Taken together, the results of the investigation provide valuable insight into the nature of novel baulamycin A analogues that have potential efficacy against MRSA owing to their membrane damaging activity and/or inhibitory efficacy against siderophore production.
Collapse
Affiliation(s)
- Namkyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sandip Sengupta
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jiwon Lee
- Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Uttam Dash
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Soojeung Kim
- Department of Chemistry, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hak Joong Kim
- Department of Chemistry, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Chiman Song
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea; Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), 5 Hwarangro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medicinal Science, Brain Korea 21 Project, Yonsei University College of Medicine, 50, Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
11
|
Suchi SA, Lee DY, Kim YK, Kang SS, Bilkis T, Yoo JC. Synergistic Effect, Improved Cell Selectivity, and Elucidating the Action Mechanism of Antimicrobial Peptide YS12. Int J Mol Sci 2023; 24:13522. [PMID: 37686328 PMCID: PMC10487915 DOI: 10.3390/ijms241713522] [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: 06/06/2023] [Revised: 08/15/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Antimicrobial peptides (AMPs) have attracted considerable attention as potential substitutes for traditional antibiotics. In our previous research, a novel antimicrobial peptide YS12 derived from the Bacillus velezensis strain showed broad-spectrum antimicrobial activity against Gram-positive and Gram-negative bacteria. In this study, the fractional inhibitory concentration index (FICI) indicated that combining YS12 with commercial antibiotics produced a synergistic effect. Following these findings, the combination of YS12 with an antibiotic resulted in a faster killing effect against bacterial strains compared to the treatment with the peptide YS12 or antibiotic alone. The peptide YS12 maintained its antimicrobial activity under different physiological salts (Na+, Mg2+, and Fe3+). Most importantly, YS12 exhibited no cytotoxicity towards Raw 264.7 cells and showed low hemolytic activity, whereas positive control melittin indicated extremely high toxicity. In terms of mode of action, we found that peptide YS12 was able to bind with LPS through electrostatic interaction. The results from fluorescent measurement revealed that peptide YS12 damaged the integrity of the bacterial membrane. Confocal laser microscopy further confirmed that the localization of peptide YS12 was almost in the cytoplasm of the cells. Peptide YS12 also exhibited anti-inflammatory activity by reducing the release of LPS-induced pro-inflammatory mediators such as TNF-α, IL-1β, and NO. Collectively, these properties strongly suggest that the antimicrobial peptide YS12 may be a promising candidate for treating microbial infections and inflammation.
Collapse
Affiliation(s)
- Suzia Aktar Suchi
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong 27709, Republic of Korea
| | - Young Kyun Kim
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Seong Soo Kang
- Department of Veterinary Medicine and BK21 Four Program, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Tahmina Bilkis
- Department of Biomedical Sciences, Chosun University, Gwangju 61452, Republic of Korea
| | - Jin Cheol Yoo
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
12
|
Ioannou P, Baliou S, Kofteridis DP. Antimicrobial Peptides in Infectious Diseases and Beyond-A Narrative Review. Life (Basel) 2023; 13:1651. [PMID: 37629508 PMCID: PMC10455936 DOI: 10.3390/life13081651] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Despite recent medical research and clinical practice developments, the development of antimicrobial resistance (AMR) significantly limits therapeutics for infectious diseases. Thus, novel treatments for infectious diseases, especially in this era of increasing AMR, are urgently needed. There is ongoing research on non-classical therapies for infectious diseases utilizing alternative antimicrobial mechanisms to fight pathogens, such as bacteriophages or antimicrobial peptides (AMPs). AMPs are evolutionarily conserved molecules naturally produced by several organisms, such as plants, insects, marine organisms, and mammals, aiming to protect the host by fighting pathogenic microorganisms. There is ongoing research regarding developing AMPs for clinical use in infectious diseases. Moreover, AMPs have several other non-medical applications in the food industry, such as preservatives, animal husbandry, plant protection, and aquaculture. This review focuses on AMPs, their origins, biology, structure, mechanisms of action, non-medical applications, and clinical applications in infectious diseases.
Collapse
Affiliation(s)
- Petros Ioannou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
- Internal Medicine, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Stella Baliou
- Internal Medicine, University Hospital of Heraklion, 71110 Heraklion, Greece
| | - Diamantis P. Kofteridis
- School of Medicine, University of Crete, 71003 Heraklion, Greece
- Internal Medicine, University Hospital of Heraklion, 71110 Heraklion, Greece
| |
Collapse
|
13
|
Gatti M, Tedeschi S, Zamparini E, Pea F, Viale P. Pharmacokinetic and pharmacodynamic considerations for optimizing antimicrobial therapy used to treat bone and joint infections: an evidence-based algorithmic approach. Expert Opin Drug Metab Toxicol 2023; 19:511-535. [PMID: 37671793 DOI: 10.1080/17425255.2023.2255525] [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: 06/23/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/07/2023]
Abstract
INTRODUCTION Bone and joint infections (BJIs) are a major health concern causing remarkable morbidity and mortality. However, which antimicrobial treatment could be the best according to specific clinical scenarios and/or to the pharmacokinetic/pharmacodynamic (PK/PD) features remains an unmet clinical need. This multidisciplinary opinion article aims to develop evidence-based algorithms for empirical and targeted antibiotic therapy of patients affected by BJIs. AREAS COVERED A multidisciplinary team of four experts had several rounds of assessment for developing algorithms devoted to empirical and targeted antimicrobial therapy of BJIs. A literature search was performed on PubMed-MEDLINE (until April 2023) to provide evidence for supporting therapeutic choices. Four different clinical scenarios were structured according to specific infection types (i.e. vertebral osteomyelitis, prosthetic joint infections, infected non-unions and other chronic osteomyelitis, and infectious arthritis), need or not of surgical intervention or revision, isolation or not of clinically relevant bacterial pathogens from blood and/or tissue cultures, and PK/PD features of antibiotics. EXPERT OPINION The proposed therapeutic algorithms were based on a multifaceted approach considering the peculiar features of each antibiotic (spectrum of activity, PK/PD properties, bone penetration rate, and anti-biofilm activity), and could be hopefully helpful in improving clinical outcome of BJIs.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Sara Tedeschi
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Eleonora Zamparini
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Federico Pea
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Clinical Pharmacology Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
- Infectious Diseases Unit, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
14
|
Ran W, Yue Y, Long F, Zhong K, Bai J, Xiao Y, Bu Q, Huang Y, Wu Y, Gao H. Antibacterial Mechanism of 2R,3R-Dihydromyricetin Against Staphylococcus aureus: Deciphering Inhibitory Effect on Biofilm and Virulence Based on Transcriptomic and Proteomic Analyses. Foodborne Pathog Dis 2023; 20:90-99. [PMID: 36862127 DOI: 10.1089/fpd.2022.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Staphylococcus aureus is a major foodborne pathogen that leads to various diseases due to its biofilm and virulence factors. This study aimed to investigate the inhibitory effect of 2R,3R-dihydromyricetin (DMY), a natural flavonoid compound, on the biofilm formation and virulence of S. aureus, and to explore the mode of action using transcriptomic and proteomic analyses. Microscopic observation revealed that DMY could remarkably inhibit the biofilm formation by S. aureus, leading to a collapse on the biofilm architecture and a decrease in viability of biofilm cell. Moreover, the hemolytic activity of S. aureus was reduced to 32.7% after treatment with subinhibitory concentration of DMY (p < 0.01). Bioinformation analysis based on RNA-sequencing and proteomic profiling revealed that DMY induced 262 differentially expressed genes and 669 differentially expressed proteins (p < 0.05). Many downregulated genes and proteins related to surface proteins were involved in biofilm formation, including clumping factor A (ClfA), iron-regulated surface determinants (IsdA, IsdB, and IsdC), fibrinogen-binding proteins (FnbA, FnbB), and serine protease. Meanwhile, DMY regulated a wide range of genes and proteins enriched in bacterial pathogenesis, cell envelope, amino acid metabolism, purine and pyrimidine metabolism, and pyruvate metabolism. These findings suggest that DMY targets S. aureus through multifarious mechanisms, and especially prompt that interference of surface proteins in cell envelope would lead to attenuation of biofilm and virulence.
Collapse
Affiliation(s)
- Wenyi Ran
- Department of Food Engineering, College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Yuxi Yue
- Department of Food Engineering, College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Feiwu Long
- Department of Hygienic Toxicology and Pathology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Research Center for Nutrition, Metabolism and Food Safety, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Kai Zhong
- Department of Food Engineering, College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Jinrong Bai
- Department of Hygienic Toxicology and Pathology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Research Center for Nutrition, Metabolism and Food Safety, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Yue Xiao
- Department of Hygienic Toxicology and Pathology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Research Center for Nutrition, Metabolism and Food Safety, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Qian Bu
- Department of Hygienic Toxicology and Pathology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Research Center for Nutrition, Metabolism and Food Safety, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Yina Huang
- Department of Hygienic Toxicology and Pathology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Research Center for Nutrition, Metabolism and Food Safety, West China-PUMC C.C. Chen Institute of Health, Sichuan University, Chengdu, China
| | - Yanping Wu
- Department of Food Engineering, College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Hong Gao
- Department of Food Engineering, College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Sharma P, Kaur J, Sharma G, Kashyap P. Plant derived antimicrobial peptides: Mechanism of target, isolation techniques, sources and pharmaceutical applications. J Food Biochem 2022; 46:e14348. [PMID: 35945701 DOI: 10.1111/jfbc.14348] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 12/29/2022]
Abstract
Antimicrobial resistance is a global health and development threat which is caused by the excess and prolonged usage of antimicrobial compounds in agriculture and pharmaceutical industries. Resistance of pathogenic microorganisms to the already existing drugs represent a serious risk to public health. Plant sources such as cereals, legumes, fruits and vegetables are potential substrates for the isolation of antimicrobial peptides (AMP) with broad spectrum antimicrobial activity against bacteria, fungi and viruses with novel immunomodulatory activities. Thus, in the quest of new antimicrobial agents, AMPs have recently gained interest. Therefore, AMP can be used in agriculture, pharmaceutical and food industries. This review focuses on various explored and unexplored plant based food sources of AMPs, their isolation techniques and antimicrobial mechanism of peptides. Therefore, the literature discussed in this review paper will prove beneficial the research purposes for agriculture, pharmaceutical and food industries. PRACTICAL APPLICATIONS: Isolation of antimicrobial peptides (AMPs) can be done on industrial scale. AMP isolated from food sources can be used in pharmaceutical and agriculture industries. AMP from natural sources mitigate the problem of antimicrobial resistance. AMP isolated from food products can be used as nutraceutical.
Collapse
Affiliation(s)
- Poorva Sharma
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, India
| | - Jasleen Kaur
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, India
| | - Geetika Sharma
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, India
| | - Piyush Kashyap
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, India
| |
Collapse
|
16
|
Dutta B, Lahiri D, Nag M, Abukhader R, Sarkar T, Pati S, Upadhye V, Pandit S, Amin MFM, Al Tawaha ARMS, Kumar M, Ray RR. Multi-Omics Approach in Amelioration of Food Products. Front Microbiol 2022; 13:955683. [PMID: 35903478 PMCID: PMC9315205 DOI: 10.3389/fmicb.2022.955683] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Determination of the quality of food products is an essential key factor needed for safe-guarding the quality of food for the interest of the consumers, along with the nutritional and sensory improvements that are necessary for delivering better quality products. Bacteriocins are a group of ribosomally synthesized antimicrobial peptides that help in maintaining the quality of food. The implementation of multi-omics approach has been important for the overall enhancement of the quality of the food. This review uses various recent technologies like proteomics, transcriptomics, and metabolomics for the overall enhancement of the quality of food products. The matrix associated with the food products requires the use of sophisticated technologies that help in the extraction of a large amount of information necessary for the amelioration of the food products. This review would provide a wholesome view of how various recent technologies can be used for improving the quality food products and for enhancing their shelf-life.
Collapse
Affiliation(s)
- Bandita Dutta
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| | - Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Rose Abukhader
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda, India
| | - Siddhartha Pati
- NatNov Bioscience Private Limited, Balasore, India
- Skills Innovation & Academic Network (SIAN) Institute, Association for Biodiversity Conservation & Research (ABC), Balasore, India
| | - Vijay Upadhye
- Center of Research for Development (CR4D), Parul Institute of Applied Sciences (PIAS), Parul University, Vadodara, India
| | - Soumya Pandit
- Department of Life Sciences, Sharda University, Noida, India
| | | | | | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR-Central Institute for Research on Cotton Technology, Mumbai, India
| | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| |
Collapse
|
17
|
Masadeh M, Ayyad A, Haddad R, Alsagar M, Alzoubi K, Alrabadi N. Functional and toxicological evaluation of the MAA-41: a novel rationally designed antimicrobial peptide using hybridization and modification methods from LL-37 and BMAP-28. Curr Pharm Des 2022; 28:2177-2188. [DOI: 10.2174/1381612828666220705150817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/30/2022] [Indexed: 11/22/2022]
Abstract
Background:
Managing bacterial infections caused by multidrug-resistant (MDR) and biofilm-forming bacteria is a global health concern. Therefore, enormous efforts were directed toward finding potential alternative antimicrobial agents such as antimicrobial peptides (AMPs).
Aim:
We aimed to synthesize a novel modified hybrid peptide designed from natural parents’ peptides with enhanced activity and reduced toxicity profile.
Method:
Rational design was used to hybridize the two antimicrobial peptides, in which the alpha-helical parts of BMAP-28 and LL-37 were combined. Then, several amino acid modifications were applied to generate a modified hybrid peptide named MAA-41. The physicochemical properties were checked using in silico methods. The MAA-41 was evaluated for its antimicrobial and anti-biofilm activities. Synergistic studies were performed with five conventional antibiotics. Finally, the cytotoxicity on mammalian cells and the hemolytic activity were assessed.
Results:
The MAA-41 revealed a broad-spectrum activity against both Gram-positive and Gram-negative bacteria including standard and MDR bacterial strains. The concentration against planktonic cells ranged between 10 and 20 μM with higher potency against Gram-negative bacteria. Additionally, the MAA-41 displayed potent activity in eradicating biofilm-forming cells, and the reported MBECs were equal to the MIC values reported for planktonic cells. This new peptide exhibited reduced toxicity profiles against erythrocyte cells but not against Vero cells. Combining MAA-41 peptides with conventional antibiotics improved the antimicrobial activity of the combined agents. Either synergistic or additive effects were shown as a significant decrease in MIC to 0.25 μM.
Conclusion:
This study proposes the validity of a novel peptide (MAA-41) with enhanced antimicrobial activity and reduced toxicity, especially when used as conventional antibiotic combinations.
Collapse
Affiliation(s)
- Majed Masadeh
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan, 22110
| | - Afnan Ayyad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan, 22110
| | - Razan Haddad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan, 22110
| | - Mohammad Alsagar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan, 22110
| | - Karem Alzoubi
- Department of Pharmacy Practice and Pharmacotherapeutics, University of Sharjah, Sharjah, UAE.
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan, 22110
| | - Nasr Alrabadi
- Department of Pharmacology, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan, 22110
| |
Collapse
|
18
|
Yaacob SN, Wahab RA, Misson M, Sabullah MK, Huyop F, Zin NM. Lactic acid bacteria and their bacteriocins: new potential weapons in the fight against methicillin-resistant Staphylococcus aureus. Future Microbiol 2022; 17:683-699. [PMID: 35414206 DOI: 10.2217/fmb-2021-0256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Alternative solutions are eminently needed to combat the escalating number of infections caused by methicillin-resistant Staphylococcus aureus (MRSA). Bacteriocins produced by lactic acid bacteria are promising candidates for next-generation antibiotics. Studies have found that these stable and nontoxic ribosomally synthesized antimicrobial peptides exhibit significant potency against other bacteria, including antibiotic-resistant strains. Here the authors review previous studies on bacteriocins that have been effectively employed to manage MRSA infections. The authors' review focuses on the beneficial traits of bacteriocins for further application as templates for the design of novel drugs. Treatments that combine bacteriocins with other antimicrobials to combat pervasive MRSA infections are also highlighted. In short, future studies should focus on the pharmacodynamics and pharmacokinetics of bacteriocins-antimicrobials to understand their interactions, as this aspect would likely determine their efficacy in MRSA inhibition.
Collapse
Affiliation(s)
- Syariffah Ns Yaacob
- Department of Bioscience, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Malaysia
| | - Roswanira A Wahab
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Malaysia
| | - Mailin Misson
- Biotechnology Research Institute, Jalan Universiti Malaysia Sabah, Kota Kinabalu, Sabah, 88400, Malaysia
| | - Mohd K Sabullah
- Faculty of Science and Natural Resources, Jalan Universiti Malaysia Sabah, Kota Kinabalu, Sabah, 88400, Malaysia
| | - Fahrul Huyop
- Department of Bioscience, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru, 81310, Malaysia
| | - Noraziah M Zin
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur, 50300, Malaysia
| |
Collapse
|
19
|
Batista Araujo J, Sastre de Souza G, Lorenzon EN. Indolicidin revisited: biological activity, potential applications and perspectives of an antimicrobial peptide not yet fully explored. World J Microbiol Biotechnol 2022; 38:39. [PMID: 35018535 DOI: 10.1007/s11274-022-03227-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/02/2022] [Indexed: 11/29/2022]
Abstract
The emergence of multidrug-resistant bacteria, viruses and tumors is a serious threat to public health. Among natural peptides, indolicidin, a 13-residue peptide belonging to the cathelicidin family, deserves special attention. Indolicidin has a broad spectrum of biological activity and is active against a wide range of targets, such as bacteria (Gram+ and Gram-), fungi and viruses. Here, we review the most important features of the biological activity, potential applications and perspectives of indolicidin and its analogs. Although not yet approved for commercialization, this peptide has great potential to be applied in different areas, including the medical, biomedical, food industry and other unexplored areas. To achieve this goal, a multidisciplinary team of researchers must work together to fine tune peptides that overall lead to novel analogs and formulations to combat existing and possibly future diseases.
Collapse
Affiliation(s)
| | - Guilherme Sastre de Souza
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, USP, Ribeirão Prêto, São Paulo, Brazil
| | | |
Collapse
|
20
|
Shaban TF, Alkawareek MY. Prediction of qualitative antibiofilm activity of antibiotics using supervised machine learning techniques. Comput Biol Med 2022; 140:105065. [PMID: 34839184 DOI: 10.1016/j.compbiomed.2021.105065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/21/2021] [Accepted: 11/21/2021] [Indexed: 11/18/2022]
Abstract
Although biofilm-specific antibiotic susceptibility assays are available, they are time-consuming and resource-intensive, and hence they are not usually performed in clinical settings. Herein, we introduce a machine learning-based predictive modeling approach that uses routinely available and easily accessible data to qualitatively predict in vitro antibiofilm activity of antibiotics with relatively high accuracy. Three optimized models based on logistic regression, decision tree, and random forest algorithms were successfully developed in this study using data manually collected from published literature. In these models, independent variables that serve as significant predictors of antibiofilm activity are minimum inhibitory concentration, bacterial Gram type, biofilm formation method, in addition to antibiotic's mechanism of action, molecular weight, and pKa. The cross-validation method showed that the optimized models exhibit prediction accuracy of 67% ± 6.1% for the logistic regression model, 73% ± 5.8% for the decision tree model, and 74% ± 5% for the random forest model. However, the one-way ANOVA test revealed that the difference in prediction accuracy between the 3 models is not statistically significant, and hence they can be considered to have comparable performance. The presented modeling approach can serve as an alternative to the resource-intensive biofilm assays to rapidly and properly manage biofilm-associated infections, especially in resource-limited clinical settings.
Collapse
Affiliation(s)
- Taqwa F Shaban
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | | |
Collapse
|
21
|
Geitani R, Moubareck CA, Costes F, Marti L, Dupuis G, Sarkis DK, Touqui L. Bactericidal effects and stability of LL-37 and CAMA in the presence of human lung epithelial cells. Microbes Infect 2021; 24:104928. [PMID: 34954126 DOI: 10.1016/j.micinf.2021.104928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022]
Abstract
Cationic antimicrobial peptides (CAMPs) are important actors in host innate immunity and represent a promising alternative to combat antibiotic resistance. Here, the bactericidal activity of two CAMPs (LL-37, and CAMA) was evaluated against Pseudomonas aeruginosa (PA) in the presence of IB3-1 cells, a cell line derived from patients with cystic fibrosis. The two CAMPs exerted different effects on PA survival depending on the timing of their administration. We observed a greater bactericidal effect when IB3-1 cells were pretreated with sub-minimum bactericidal concentrations (Sub-MBCs) of the CAMPs prior to infection. These findings suggest that CAMPs induce the production of factors by IB3-1 cells that improve their bactericidal action. However, we observed no bactericidal effect when supra-minimum bactericidal concentrations (Supra-MBCs) of the CAMPs were added to IB3-1 cells at the same time or after infection. Western-blot analysis showed a large decrease in LL-37 levels in supernatants of infected IB3-1 cells and an increase in LL-37 binding to these cells after LL-37 administration. LL-37 induced a weak inflammatory response in the cells without being toxic. In conclusion, our findings suggest a potential prophylactic action of CAMPs. The bactericidal effects were low when the CAMPs were added after cell infection, likely due to degradation of CAMPs by bacterial or epithelial cell proteases and/or due to adherence of CAMPs to cells becoming less available for direct bacterial killing.
Collapse
Affiliation(s)
- Regina Geitani
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon.
| | - Carole Ayoub Moubareck
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon; College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Floriane Costes
- "Sorbonne Université", INSERM UMR_S 938, "Centre de Recherche Saint-Antoine" (CRSA), Paris, France; "Mucoviscidose and Bronchopathies Chroniques", Department "Santé Globale", Pasteur Institute, Paris, France
| | - Léa Marti
- "Sorbonne Université", INSERM UMR_S 938, "Centre de Recherche Saint-Antoine" (CRSA), Paris, France; "Mucoviscidose and Bronchopathies Chroniques", Department "Santé Globale", Pasteur Institute, Paris, France
| | - Gabrielle Dupuis
- "Sorbonne Université", INSERM UMR_S 938, "Centre de Recherche Saint-Antoine" (CRSA), Paris, France; "Mucoviscidose and Bronchopathies Chroniques", Department "Santé Globale", Pasteur Institute, Paris, France
| | - Dolla Karam Sarkis
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon
| | - Lhousseine Touqui
- "Sorbonne Université", INSERM UMR_S 938, "Centre de Recherche Saint-Antoine" (CRSA), Paris, France; "Mucoviscidose and Bronchopathies Chroniques", Department "Santé Globale", Pasteur Institute, Paris, France.
| |
Collapse
|
22
|
Ude Z, Flothkötter N, Sheehan G, Brennan M, Kavanagh K, Marmion CJ. Multi-targeted metallo-ciprofloxacin derivatives rationally designed and developed to overcome antimicrobial resistance. Int J Antimicrob Agents 2021; 58:106449. [PMID: 34644603 DOI: 10.1016/j.ijantimicag.2021.106449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/28/2021] [Accepted: 10/02/2021] [Indexed: 10/20/2022]
Abstract
Antimicrobial resistance is a major global threat to human health due to the rise, spread and persistence of multi-drug-resistant bacteria or 'superbugs'. There is an urgent need to develop novel chemotherapeutics to overcome this overarching challenge. The authors derivatized a clinically used fluoroquinolone antibiotic ciprofloxacin (Cip), and complexed it to a copper phenanthrene framework. This resulted in the development of two novel metallo-antibiotics of general formula [Cu(N,N)(CipHA)]NO3 where N,N represents a phenanthrene ligand and CipHA represents a hydroxamic acid of Cip derivative. Comprehensive studies, including a detailed proteomic study in which Staphylococcus aureus cells were exposed to the complexes, were undertaken to gain an insight into their mode of action. These new complexes possess potent antibacterial activity against S. aureus and methicillin-resistant S. aureus. In addition, they were found to be well tolerated in vivo in Galleria mellonella larvae, which has both functional and structural similarities to the innate immune system of mammals. These findings suggest that proteins involved in virulence, pathogenesis, and the synthesis of nucleotides and DNA repair mechanisms are most affected. In addition, both complexes affected similar cell pathways when compared with clinically used Cip, including cationic antimicrobial peptide resistance. The Cu-DPPZ-CipHA (DPPZ = dipyrido[3,2-a:2',3'-c]phenazine) analogue also induces cell leakage, which leads to an altered proteome indicative of reduced virulence and increased stress.
Collapse
Affiliation(s)
- Ziga Ude
- Centre for Synthesis and Chemical Biology, Department of Chemistry, RCSI, University of Medicine and Health Sciences, Dublin, Ireland
| | - Nils Flothkötter
- Centre for Synthesis and Chemical Biology, Department of Chemistry, RCSI, University of Medicine and Health Sciences, Dublin, Ireland
| | - Gerard Sheehan
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland
| | - Marian Brennan
- School of Pharmacy and Biomolecular Sciences, RCSI, University of Medicine and Health Sciences, Dublin, Ireland
| | - Kevin Kavanagh
- SSPC Pharma Research Centre, Department of Biology, Maynooth University, Maynooth, Co. Kildare, Ireland.
| | - Celine J Marmion
- Centre for Synthesis and Chemical Biology, Department of Chemistry, RCSI, University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
23
|
Enhancement of Antibiofilm Activity of Ciprofloxacin against Staphylococcus aureus by Administration of Antimicrobial Peptides. Antibiotics (Basel) 2021; 10:antibiotics10101159. [PMID: 34680739 PMCID: PMC8532819 DOI: 10.3390/antibiotics10101159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus aureus can develop resistance by mutation, transfection or biofilm formation. Resistance was induced in S. aureus by growth in sub-inhibitory concentrations of ciprofloxacin for 30 days. The ability of the antimicrobials to disrupt biofilms was determined using crystal violet and live/dead staining. Effects on the cell membranes of biofilm cells were evaluated by measuring release of dyes and ATP, and nucleic acids. None of the strains developed resistance to AMPs while only S. aureus ATCC 25923 developed resistance (128 times) to ciprofloxacin after 30 passages. Only peptides reduced biofilms of ciprofloxacin-resistant cells. The antibiofilm effect of melimine with ciprofloxacin was more (27%) than with melimine alone at 1X MIC (p < 0.001). Similarly, at 1X MIC the combination of Mel4 and ciprofloxacin produced more (48%) biofilm disruption than Mel4 alone (p < 0.001). Combinations of either of the peptides with ciprofloxacin at 2X MIC released ≥ 66 nM ATP, more than either peptide alone (p ≤ 0.005). At 2X MIC, only melimine in combination with ciprofloxacin released DNA/RNA which was three times more than that released by melimine alone (p = 0.043). These results suggest the potential use of melimine and Mel4 with conventional antibiotics for the treatment of S. aureus biofilms.
Collapse
|
24
|
Zhang QY, Yan ZB, Meng YM, Hong XY, Shao G, Ma JJ, Cheng XR, Liu J, Kang J, Fu CY. Antimicrobial peptides: mechanism of action, activity and clinical potential. Mil Med Res 2021; 8:48. [PMID: 34496967 PMCID: PMC8425997 DOI: 10.1186/s40779-021-00343-2] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
The management of bacterial infections is becoming a major clinical challenge due to the rapid evolution of antibiotic resistant bacteria. As an excellent candidate to overcome antibiotic resistance, antimicrobial peptides (AMPs) that are produced from the synthetic and natural sources demonstrate a broad-spectrum antimicrobial activity with the high specificity and low toxicity. These peptides possess distinctive structures and functions by employing sophisticated mechanisms of action. This comprehensive review provides a broad overview of AMPs from the origin, structural characteristics, mechanisms of action, biological activities to clinical applications. We finally discuss the strategies to optimize and develop AMP-based treatment as the potential antimicrobial and anticancer therapeutics.
Collapse
Affiliation(s)
- Qi-Yu Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Zhi-Bin Yan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Yue-Ming Meng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Xiang-Yu Hong
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Gang Shao
- Department of Oncology, The 903rd Hospital of PLA, Hangzhou, 310013, Zhejiang, China
| | - Jun-Jie Ma
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Xu-Rui Cheng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China
| | - Jun Liu
- Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California San Francisco, 555 Mission Bay Blvd. South, San Francisco, CA, 94158, USA
| | - Jian Kang
- Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, VIC, 3000, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cai-Yun Fu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, No. 928, Street 2, Xiasha Higher Education Zone, Hangzhou, 310018, Zhejiang, China.
| |
Collapse
|
25
|
Kranjec C, Kristensen SS, Bartkiewicz KT, Brønner M, Cavanagh JP, Srikantam A, Mathiesen G, Diep DB. A bacteriocin-based treatment option for Staphylococcus haemolyticus biofilms. Sci Rep 2021; 11:13909. [PMID: 34230527 PMCID: PMC8260761 DOI: 10.1038/s41598-021-93158-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/15/2021] [Indexed: 01/19/2023] Open
Abstract
Bacteriocins are ribosomally-synthesized antimicrobial peptides, showing great potential as novel treatment options for multidrug-resistant pathogens. In this study, we designed a novel hybrid bacteriocin, Hybrid 1 (H1), by combing the N-terminal part and the C-terminal part of the related bacteriocins enterocin K1 (K1) and enterocin EJ97 (EJ97), respectively. Like the parental bacteriocins, H1 used the membrane-bound protease RseP as receptor, however, it differed from the others in the inhibition spectrum. Most notably, H1 showed a superior antimicrobial effect towards Staphylococcus haemolyticus—an important nosocomial pathogen. To avoid strain-dependency, we further evaluated H1 against 27 clinical and commensal S. haemolyticus strains, with H1 indeed showing high activity towards all strains. To curtail the rise of resistant mutants and further explore the potential of H1 as a therapeutic agent, we designed a bacteriocin-based formulation where H1 was used in combination with the broad-spectrum bacteriocins micrococcin P1 and garvicin KS. Unlike the individual bacteriocins, the three-component combination was highly effective against planktonic cells and completely eradicated biofilm-associated S. haemolyticus cells in vitro. Most importantly, the formulation efficiently prevented development of resistant mutants as well. These findings indicate the potential of a bacteriocins-based formulation as a treatment option for S. haemolyticus.
Collapse
Affiliation(s)
- Christian Kranjec
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Sofie S Kristensen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Karolina T Bartkiewicz
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Mikkel Brønner
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Jorunn P Cavanagh
- Pediatric Infections Group, Department of Pediatrics, University Hospital of North Norway, Tromsö, Norway.,Pediatric Infections Group, Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsö, Norway
| | - Aparna Srikantam
- Blue Peter Public Health and Research Centre, LEPRA Society, Hyderabad, India
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Dzung B Diep
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway.
| |
Collapse
|
26
|
Moretta A, Scieuzo C, Petrone AM, Salvia R, Manniello MD, Franco A, Lucchetti D, Vassallo A, Vogel H, Sgambato A, Falabella P. Antimicrobial Peptides: A New Hope in Biomedical and Pharmaceutical Fields. Front Cell Infect Microbiol 2021; 11:668632. [PMID: 34195099 PMCID: PMC8238046 DOI: 10.3389/fcimb.2021.668632] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
Antibiotics are essential drugs used to treat pathogenic bacteria, but their prolonged use contributes to the development and spread of drug-resistant microorganisms. Antibiotic resistance is a serious challenge and has led to the need for new alternative molecules less prone to bacterial resistance. Antimicrobial peptides (AMPs) have aroused great interest as potential next-generation antibiotics, since they are bioactive small proteins, naturally produced by all living organisms, and representing the first line of defense against fungi, viruses and bacteria. AMPs are commonly classified according to their sources, which are represented by microorganisms, plants and animals, as well as to their secondary structure, their biosynthesis and their mechanism of action. They find application in different fields such as agriculture, food industry and medicine, on which we focused our attention in this review. Particularly, we examined AMP potential applicability in wound healing, skin infections and metabolic syndrome, considering their ability to act as potential Angiotensin-Converting Enzyme I and pancreatic lipase inhibitory peptides as well as antioxidant peptides. Moreover, we argued about the pharmacokinetic and pharmacodynamic approaches to develop new antibiotics, the drug development strategies and the formulation approaches which need to be taken into account in developing clinically suitable AMP applications.
Collapse
Affiliation(s)
- Antonio Moretta
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Carmen Scieuzo
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| | | | - Rosanna Salvia
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| | | | - Antonio Franco
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| | - Donatella Lucchetti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Vassallo
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Heiko Vogel
- Department of Entomology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Patrizia Falabella
- Department of Sciences, University of Basilicata, Potenza, Italy
- Spinoff XFlies s.r.l, University of Basilicata, Potenza, Italy
| |
Collapse
|
27
|
Sahoo A, Swain SS, Behera A, Sahoo G, Mahapatra PK, Panda SK. Antimicrobial Peptides Derived From Insects Offer a Novel Therapeutic Option to Combat Biofilm: A Review. Front Microbiol 2021; 12:661195. [PMID: 34248873 PMCID: PMC8265172 DOI: 10.3389/fmicb.2021.661195] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022] Open
Abstract
Biofilms form a complex layer with defined structures, that attach on biotic or abiotic surfaces, are tough to eradicate and tend to cause some resistance against most antibiotics. Several studies confirmed that biofilm-producing bacteria exhibit higher resistance compared to the planktonic form of the same species. Antibiotic resistance factors are well understood in planktonic bacteria which is not so in case of biofilm producing forms. This may be due to the lack of available drugs with known resistance mechanisms for biofilms. Existing antibiotics cannot eradicate most biofilms, especially of ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species). Insects produce complex and diverse set of chemicals for survival and defense. Antimicrobial peptides (AMPs), produced by most insects, generally have a broad spectrum of activity and the potential to bypass the resistance mechanisms of classical antibiotics. Besides, AMPs may well act synergistically with classical antibiotics for a double-pronged attack on infections. Thus, AMPs could be promising alternatives to overcome medically important biofilms, decrease the possibility of acquired resistance and treatment of multidrug-resistant pathogens including ESKAPE. The present review focuses on insect-derived AMPs with special reference to anti-biofilm-based strategies. It covers the AMP composition, pathways and mechanisms of action, the formation of biofilms, impact of biofilms on human diseases, current strategies as well as therapeutic options to combat biofilm with antimicrobial peptides from insects. In addition, the review also illustrates the importance of bioinformatics tools and molecular docking studies to boost the importance of select bioactive peptides those can be developed as drugs, as well as suggestions for further basic and clinical research.
Collapse
Affiliation(s)
- Alaka Sahoo
- Department of Skin & VD, Institute of Medical Sciences, SUM Hospital, Siksha O Anusandhan University, Bhubaneswar, India
| | - Shasank Sekhar Swain
- Division of Microbiology & NCDs, ICMR-Regional Medical Research Centre, Bhubaneswar, India
| | - Ayusman Behera
- Department of Zoology, Maharaja Sriram Chandra Bhanja Deo University, Baripada, India
| | - Gunanidhi Sahoo
- Department of Zoology, Utkal University, Vani Vihar, Bhubaneswar, India
| | | | - Sujogya Kumar Panda
- Centre of Environment, Climate Change and Public Health, RUSA 2.0, Utkal University, Vani Vihar, Bhubaneswar, India
| |
Collapse
|
28
|
Kranjec C, Morales Angeles D, Torrissen Mårli M, Fernández L, García P, Kjos M, Diep DB. Staphylococcal Biofilms: Challenges and Novel Therapeutic Perspectives. Antibiotics (Basel) 2021; 10:131. [PMID: 33573022 PMCID: PMC7911828 DOI: 10.3390/antibiotics10020131] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Staphylococci, like Staphylococcus aureus and S. epidermidis, are common colonizers of the human microbiota. While being harmless in many cases, many virulence factors result in them being opportunistic pathogens and one of the major causes of hospital-acquired infections worldwide. One of these virulence factors is the ability to form biofilms-three-dimensional communities of microorganisms embedded in an extracellular polymeric matrix (EPS). The EPS is composed of polysaccharides, proteins and extracellular DNA, and is finely regulated in response to environmental conditions. This structured environment protects the embedded bacteria from the human immune system and decreases their susceptibility to antimicrobials, making infections caused by staphylococci particularly difficult to treat. With the rise of antibiotic-resistant staphylococci, together with difficulty in removing biofilms, there is a great need for new treatment strategies. The purpose of this review is to provide an overview of our current knowledge of the stages of biofilm development and what difficulties may arise when trying to eradicate staphylococcal biofilms. Furthermore, we look into promising targets and therapeutic methods, including bacteriocins and phage-derived antibiofilm approaches.
Collapse
Affiliation(s)
- Christian Kranjec
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Danae Morales Angeles
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Marita Torrissen Mårli
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Lucía Fernández
- Department of Technology and Biotechnology of Dairy Products, Dairy Research Institute of Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (L.F.); (P.G.)
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Pilar García
- Department of Technology and Biotechnology of Dairy Products, Dairy Research Institute of Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (L.F.); (P.G.)
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Morten Kjos
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| | - Dzung B. Diep
- Faculty of Chemistry, Biotechnology and Food Science, The Norwegian University of Life Sciences, 1432 Ås, Norway; (C.K.); (D.M.A.); (M.T.M.)
| |
Collapse
|
29
|
Duraisamy S, Balakrishnan S, Ranjith S, Husain F, Sathyan A, Peter AS, Prahalathan C, Kumarasamy A. Bacteriocin-a potential antimicrobial peptide towards disrupting and preventing biofilm formation in the clinical and environmental locales. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:44922-44936. [PMID: 33006097 DOI: 10.1007/s11356-020-10989-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/23/2020] [Indexed: 06/11/2023]
Abstract
Biofilm, a consortium of microbial cells, protected by extracellular polymeric matrix, is considered a global challenge due to the inherent antibiotic resistance conferred by its lifestyle. Besides, it poses environmental threats causing huge damage in food industries, fisheries, refineries, water systems, pharmaceutical industries, medical industries, etc. Living in a community of microbial populations is most critical in the clinical field, making it responsible for about 80% of severe and chronic microbial diseases. The necessity to find an alternative approach is the need of the hour to solve these crises. So far, many approaches have been attempted to disrupt the initial stage of biofilm formation, including adherence and maturation. Bacteriocins are a group of antimicrobial peptides, produced by bacteria having the potential to disrupt biofilm either by itself or in combination with other drugs than antibiotic counterparts. A clear understanding on mechanisms of bacterial biofilm formation, progression, and resistance will surely lead to the development of innovative, effective biofilm control strategies in pharmaceutical, health care industries and environmental locales.
Collapse
Affiliation(s)
- Senbagam Duraisamy
- Microbial Biotechnology Laboratory, Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Senthilkumar Balakrishnan
- Department of Medical Microbiology, College of Health and Medical Sciences, Haramaya University, P.O. Box 235, Harar, Ethiopia
| | - Sukumar Ranjith
- Microbial Biotechnology Laboratory, Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Fazal Husain
- Microbial Biotechnology Laboratory, Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Aswathy Sathyan
- Microbial Biotechnology Laboratory, Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Ansu Susan Peter
- Microbial Biotechnology Laboratory, Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Chidambaram Prahalathan
- Department of Biochemistry, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Anbarasu Kumarasamy
- Microbial Biotechnology Laboratory, Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
30
|
Activity of Antimicrobial Peptides and Ciprofloxacin against Pseudomonas aeruginosa Biofilms. Molecules 2020; 25:molecules25173843. [PMID: 32847059 PMCID: PMC7503749 DOI: 10.3390/molecules25173843] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 12/31/2022] Open
Abstract
Pseudomonas aeruginosa is increasingly resistant to conventional antibiotics, which can be compounded by the formation of biofilms on surfaces conferring additional resistance. P. aeruginosa was grown in sub-inhibitory concentrations of the antimicrobial peptides (AMPs) melimine and Mel4 or ciprofloxacin for 30 consecutive days to induce the development of resistance. Antibiofilm effect of AMPs and ciprofloxacin was evaluated using crystal violet and live/dead staining with confocal microscopy. Effect on the cell membrane of biofilm cells was evaluated using DiSC(3)-5 dye and release of intracellular ATP and DNA/RNA. The minimum inhibitory concentration (MIC) of ciprofloxacin increased 64-fold after 30 passages, but did not increase for melimine or Mel4. Ciprofloxacin could not inhibit biofilm formation of resistant cells at 4× MIC, but both AMPs reduced biofilms by >75% at 1× MIC. At 1× MIC, only the combination of either AMP with ciprofloxacin was able to significantly disrupt pre-formed biofilms (≥61%; p < 0.001). Only AMPs depolarized the cell membranes of biofilm cells at 1× MIC. At 1× MIC either AMP with ciprofloxacin released a significant amount of ATP (p < 0.04), but did not release DNA/RNA. AMPs do not easily induce resistance in P. aeruginosa and can be used in combination with ciprofloxacin to treat biofilm.
Collapse
|
31
|
Schilcher K, Horswill AR. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol Mol Biol Rev 2020; 84:e00026-19. [PMID: 32792334 PMCID: PMC7430342 DOI: 10.1128/mmbr.00026-19] [Citation(s) in RCA: 321] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In many natural and clinical settings, bacteria are associated with some type of biotic or abiotic surface that enables them to form biofilms, a multicellular lifestyle with bacteria embedded in an extracellular matrix. Staphylococcus aureus and Staphylococcus epidermidis, the most frequent causes of biofilm-associated infections on indwelling medical devices, can switch between an existence as single free-floating cells and multicellular biofilms. During biofilm formation, cells first attach to a surface and then multiply to form microcolonies. They subsequently produce the extracellular matrix, a hallmark of biofilm formation, which consists of polysaccharides, proteins, and extracellular DNA. After biofilm maturation into three-dimensional structures, the biofilm community undergoes a disassembly process that leads to the dissemination of staphylococcal cells. As biofilms are dynamic and complex biological systems, staphylococci have evolved a vast network of regulatory mechanisms to modify and fine-tune biofilm development upon changes in environmental conditions. Thus, biofilm formation is used as a strategy for survival and persistence in the human host and can serve as a reservoir for spreading to new infection sites. Moreover, staphylococcal biofilms provide enhanced resilience toward antibiotics and the immune response and impose remarkable therapeutic challenges in clinics worldwide. This review provides an overview and an updated perspective on staphylococcal biofilms, describing the characteristic features of biofilm formation, the structural and functional properties of the biofilm matrix, and the most important mechanisms involved in the regulation of staphylococcal biofilm formation. Finally, we highlight promising strategies and technologies, including multitargeted or combinational therapies, to eradicate staphylococcal biofilms.
Collapse
Affiliation(s)
- Katrin Schilcher
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
32
|
Schilcher K, Horswill AR. Staphylococcal Biofilm Development: Structure, Regulation, and Treatment Strategies. Microbiol Mol Biol Rev 2020. [PMID: 32792334 DOI: 10.1128/mmbr.00026-19/asset/e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023] Open
Abstract
In many natural and clinical settings, bacteria are associated with some type of biotic or abiotic surface that enables them to form biofilms, a multicellular lifestyle with bacteria embedded in an extracellular matrix. Staphylococcus aureus and Staphylococcus epidermidis, the most frequent causes of biofilm-associated infections on indwelling medical devices, can switch between an existence as single free-floating cells and multicellular biofilms. During biofilm formation, cells first attach to a surface and then multiply to form microcolonies. They subsequently produce the extracellular matrix, a hallmark of biofilm formation, which consists of polysaccharides, proteins, and extracellular DNA. After biofilm maturation into three-dimensional structures, the biofilm community undergoes a disassembly process that leads to the dissemination of staphylococcal cells. As biofilms are dynamic and complex biological systems, staphylococci have evolved a vast network of regulatory mechanisms to modify and fine-tune biofilm development upon changes in environmental conditions. Thus, biofilm formation is used as a strategy for survival and persistence in the human host and can serve as a reservoir for spreading to new infection sites. Moreover, staphylococcal biofilms provide enhanced resilience toward antibiotics and the immune response and impose remarkable therapeutic challenges in clinics worldwide. This review provides an overview and an updated perspective on staphylococcal biofilms, describing the characteristic features of biofilm formation, the structural and functional properties of the biofilm matrix, and the most important mechanisms involved in the regulation of staphylococcal biofilm formation. Finally, we highlight promising strategies and technologies, including multitargeted or combinational therapies, to eradicate staphylococcal biofilms.
Collapse
Affiliation(s)
- Katrin Schilcher
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Veterans Affairs Eastern Colorado Health Care System, Denver, Colorado, USA
| |
Collapse
|
33
|
Schofs L, Sparo MD, Sánchez Bruni SF. Gram-positive bacteriocins: usage as antimicrobial agents in veterinary medicine. Vet Res Commun 2020; 44:89-100. [PMID: 32656740 DOI: 10.1007/s11259-020-09776-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Antimicrobial resistance is a worldwide spread phenomenon that affects both human and veterinary medicine. This issue has led to a "One Health" approach in order to coordinate efforts and set back the development of drug-resistant microbes. In the search for alternatives therapies, bacteriocins or antimicrobial peptides have proven to be effective both in vitro and in vivo for multiples pathogens, even those resistant to many classic antibiotics. Gram-positive bacteriocins have been the most studied to the present. The use of bacteriocins as therapeutically active molecules is limited mainly due to difficulties in production, purification, delivery systems and regulatory approvals. To overcome some of these limitations, biotechnological and nanotechnological approaches are evaluated. Bacteriocins proved to be a good complement for conventional antibiotics therapy. Antimicrobial peptides are nowadays included in the veterinary products such as udder disinfectant for dairy cattle and dermatological medicated wipe for topical use on dogs, cats, and horses. But there are other potential uses to explore in the veterinary field for both companion and production animals.
Collapse
Affiliation(s)
- Laureano Schofs
- Laboratory of Pharmacology, Faculty of Veterinary Medicine, Universidad Nacional del Centro de la Provincia de Buenos Aires, CIVETAN- CONICET, B7000, Tandil, Argentina. .,Tandil Veterinary Research Center (CIVETAN) Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Comisión de investigaciones científicas de la Provincia de Buenos Aires (CICPBA), Tandil, B7000, Argentina.
| | - Mónica D Sparo
- Tandil Veterinary Research Center (CIVETAN) Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Comisión de investigaciones científicas de la Provincia de Buenos Aires (CICPBA), Tandil, B7000, Argentina.,Clinical Department, Faculty of Health Science, Universidad Nacional del Centro de la Provincia de Buenos Aires, Olavarría, B7400, Argentina
| | - Sergio F Sánchez Bruni
- Laboratory of Pharmacology, Faculty of Veterinary Medicine, Universidad Nacional del Centro de la Provincia de Buenos Aires, CIVETAN- CONICET, B7000, Tandil, Argentina.,Tandil Veterinary Research Center (CIVETAN) Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Comisión de investigaciones científicas de la Provincia de Buenos Aires (CICPBA), Tandil, B7000, Argentina
| |
Collapse
|
34
|
Combinational therapy with antibiotics and antibiotic-loaded adipose-derived stem cells reduce abscess formation in implant-related infection in rats. Sci Rep 2020; 10:11182. [PMID: 32636453 PMCID: PMC7341734 DOI: 10.1038/s41598-020-68184-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
Implant-related infection is difficult to treat without extended antibiotic courses. However, the long-term use of antibiotics has led to the development of multidrug- and methicillin-resistant Staphylococcusaureus. Thus, alternatives to conventional antibiotic therapy are needed. Recently, mesenchymal stem cells have been shown to have antimicrobial properties. This study aimed to evaluate the antimicrobial activity and therapeutic effect of local treatment with antibiotic-loaded adipose-derived stem cells (ADSCs) plus an antibiotic in a rat implant-associated infection model. Liquid chromatography/tandem mass spectrometry revealed that ADSCs cultured in the presence of ciprofloxacin for 24 h showed time-dependent antibiotic loading. Next, we studied the therapeutic effects of ADSCs and ciprofloxacin alone or in combination in an implant-related infection rat model. The therapeutic effects of ADSCs plus antibiotics, antibiotics, and ADSCs were compared with no treatment as a control. Rats treated with ADSCs plus ciprofloxacin had the lowest modified osteomyelitis scores, abscess formation, and bacterial burden on the implant among all groups (P < 0.05). Thus, local treatment with ADSCs plus an antibiotic has an antimicrobial effect in implant-related infection and decrease abscess formation. Thus, our findings indicate that local administration of ADSCs with antibiotics represents a novel treatment strategy for implant-associated osteomyelitis.
Collapse
|
35
|
Jiang Y, Mei C, Huang X, Gu Q, Song D. Antibacterial Activity and Mechanism of a Bacteriocin Derived from the Valine-Cecropin A(1–8)-Plantaricin ZJ5(1–18) Hybrid Peptide Against Escherichia coli O104. FOOD BIOPHYS 2020. [DOI: 10.1007/s11483-020-09636-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
Sharma G, Dang S, K A, Kalia M, Gabrani R. Synergistic antibacterial and anti-biofilm activity of nisin like bacteriocin with curcumin and cinnamaldehyde against ESBL and MBL producing clinical strains. BIOFOULING 2020; 36:710-724. [PMID: 32772715 DOI: 10.1080/08927014.2020.1804553] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 06/11/2023]
Abstract
Bacteriocins are small peptides that can inhibit the growth of a diverse range of microbes. There is a need to identify bacteriocins that are effective against biofilms of resistant clinical strains. The present study focussed on the efficacy of purified nisin like bacteriocin-GAM217 against extended spectrum β-lactamase (ESBL) and metallo-beta-lactamase (MBL) producing clinical strains. Bacteriocin-GAM217 when combined with curcumin and cinnamaldehyde, synergistically enhanced antibacterial activity against planktonic and biofilm cultures of Staphylococcus epidermidis and Escherichia coli. Bacteriocin-GAM217 and phytochemical combinations inhibited biofilm formation by >80%, and disrupted the biofilm for selected ESBL and MBL producing clinical strains. The anti-adhesion assay showed that these combinatorial compounds significantly lowered the attachment of bacteria to Vero cells and that they elicited membrane permeability and rapid killing as viewed by confocal microscopy. This study demonstrates that bacteriocin-GAM217 in combination with phytochemicals can be a potential anti-biofilm agent and thus has potential for biomedical applications.
Collapse
Affiliation(s)
- Garima Sharma
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Aruna K
- Department of Microbiology, Wilson College, Mumbai, India
| | | | - Reema Gabrani
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
37
|
Geitani R, Moubareck CA, Xu Z, Karam Sarkis D, Touqui L. Expression and Roles of Antimicrobial Peptides in Innate Defense of Airway Mucosa: Potential Implication in Cystic Fibrosis. Front Immunol 2020; 11:1198. [PMID: 32695100 PMCID: PMC7338688 DOI: 10.3389/fimmu.2020.01198] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
The treatment of respiratory infections is associated with the dissemination of antibiotic resistance in the community and clinical settings. Development of new antibiotics is notoriously costly and slow; therefore, alternative strategies are needed. Antimicrobial peptides (AMPs), the central effector molecules of the immune system, are being considered as alternatives to conventional antibiotics. Most AMPs are epithelium-derived and play a key role in host defense at mucosal surfaces. They are classified on the basis of their structure and amino acid motifs. These peptides display a range of activities, including not only direct antimicrobial activity, but also immunomodulation and wound repair. In the lung, airway epithelial cells and neutrophils, in particular, contribute to AMP synthesis. The relevance of AMPs for host defense against infection has been demonstrated in animal models and is supported by observations in patient studies, showing altered expression and/or unfavorable circumstances for their action in a variety of lung diseases. Of note, AMPs are active against bacterial strains that are resistant to conventional antibiotics, including multidrug-resistant bacteria. Several strategies have been proposed to use these peptides in the treatment of infections, including direct administration of AMPs. In this review, we focus on studies related to direct bactericidal effects of AMPs and their potential clinical applications with a particular focus on cystic fibrosis.
Collapse
Affiliation(s)
- Regina Geitani
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon
| | - Carole Ayoub Moubareck
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon
- College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint Antoine (CRSA), Paris, France
- “Mucoviscidose and Bronchopathies Chroniques”, Pasteur Institute, Paris, France
| | - Dolla Karam Sarkis
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon
| | - Lhousseine Touqui
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint Antoine (CRSA), Paris, France
- “Mucoviscidose and Bronchopathies Chroniques”, Pasteur Institute, Paris, France
| |
Collapse
|
38
|
Li J, Fernández-Millán P, Boix E. Synergism between Host Defence Peptides and Antibiotics Against Bacterial Infections. Curr Top Med Chem 2020; 20:1238-1263. [DOI: 10.2174/1568026620666200303122626] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/22/2020] [Accepted: 02/07/2020] [Indexed: 01/10/2023]
Abstract
Background:Antimicrobial resistance (AMR) to conventional antibiotics is becoming one of the main global health threats and novel alternative strategies are urging. Antimicrobial peptides (AMPs), once forgotten, are coming back into the scene as promising tools to overcome bacterial resistance. Recent findings have attracted attention to the potentiality of AMPs to work as antibiotic adjuvants.Methods:In this review, we have tried to collect the currently available information on the mechanism of action of AMPs in synergy with other antimicrobial agents. In particular, we have focused on the mechanisms of action that mediate the inhibition of the emergence of bacterial resistance by AMPs.Results and Conclusion:We find in the literature many examples where AMPs can significantly reduce the antibiotic effective concentration. Mainly, the peptides work at the bacterial cell wall and thereby facilitate the drug access to its intracellular target. Complementarily, AMPs can also contribute to permeate the exopolysaccharide layer of biofilm communities, or even prevent bacterial adhesion and biofilm growth. Secondly, we find other peptides that can directly block the emergence of bacterial resistance mechanisms or interfere with the community quorum-sensing systems. Interestingly, the effective peptide concentrations for adjuvant activity and inhibition of bacterial resistance are much lower than the required for direct antimicrobial action. Finally, many AMPs expressed by innate immune cells are endowed with immunomodulatory properties and can participate in the host response against infection. Recent studies in animal models confirm that AMPs work as adjuvants at non-toxic concentrations and can be safely administrated for novel combined chemotherapies.
Collapse
Affiliation(s)
- Jiarui Li
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Pablo Fernández-Millán
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| | - Ester Boix
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autonoma de Barcelona, Cerdanyola del Valles, Spain
| |
Collapse
|
39
|
Gilbertie JM, Schaer TP, Schubert AG, Jacob ME, Menegatti S, Lavoie RA, Schnabel LV. Platelet-rich plasma lysate displays antibiofilm properties and restores antimicrobial activity against synovial fluid biofilms in vitro. J Orthop Res 2020; 38:1365-1374. [PMID: 31922274 PMCID: PMC8018705 DOI: 10.1002/jor.24584] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/06/2019] [Accepted: 01/03/2020] [Indexed: 02/04/2023]
Abstract
Infectious arthritis is difficult to treat in both human and veterinary clinical practice. Recent literature reports Staphylococcus aureus as well as other gram-positive and gram-negative isolates forming free-floating biofilms in both human and equine synovial fluid that are tolerant to traditional antimicrobial therapy. Using an in vitro equine model, we investigated the ability of platelet-rich plasma (PRP) formulations to combat synovial fluid biofilm aggregates. Synovial fluid was infected, and biofilm aggregates allowed to form over a 2-hour period. PRP was collected and processed into different formulations by platelet concentration, leukocyte presence, and activation or lysis. Infected synovial fluid was treated with different PRP formulations with or without aminoglycoside cotreatment. Bacterial load (colony-forming unit/mL) was determined by serial dilutions and plate counting at 8 hours posttreatment. All PRP formulations displayed antimicrobial properties; however, formulations containing higher concentrations of platelets without leukocytes had increased antimicrobial activity. Lysis of PRP and pooling of the PRP lysate (PRP-L) from multiple horses as compared to individual horses further increased antimicrobial activity. This activity was lost with the removal of the plasma component or inhibition of the proteolytic activity within the plasma. Fractionation of pooled PRP-L identified the bioactive components to be cationic and low-molecular weight (<10 kDa). Overall, PRP-L exhibited synergism with amikacin against aminoglycoside tolerant biofilm aggregates with greater activity against gram-positive bacteria. In conclusion, the use of PRP-L has the potential to augment current antimicrobial treatment regimens which could lead to a decrease in morbidity and mortality associated with infectious arthritis.
Collapse
Affiliation(s)
- Jessica M. Gilbertie
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina,Department of Clinical Studies New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| | - Thomas P. Schaer
- Department of Clinical Studies New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, Pennsylvania
| | - Alicia G. Schubert
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Megan E. Jacob
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina,Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Stefano Menegatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina,Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - R. Ashton Lavoie
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Lauren V. Schnabel
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
40
|
Khan F, Pham DTN, Oloketuyi SF, Kim YM. Antibiotics Application Strategies to Control Biofilm Formation in Pathogenic Bacteria. Curr Pharm Biotechnol 2020; 21:270-286. [PMID: 31721708 DOI: 10.2174/1389201020666191112155905] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/09/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND The establishment of a biofilm by most pathogenic bacteria has been known as one of the resistance mechanisms against antibiotics. A biofilm is a structural component where the bacterial community adheres to the biotic or abiotic surfaces by the help of Extracellular Polymeric Substances (EPS) produced by bacterial cells. The biofilm matrix possesses the ability to resist several adverse environmental factors, including the effect of antibiotics. Therefore, the resistance of bacterial biofilm-forming cells could be increased up to 1000 times than the planktonic cells, hence requiring a significantly high concentration of antibiotics for treatment. METHODS Up to the present, several methodologies employing antibiotics as an anti-biofilm, antivirulence or quorum quenching agent have been developed for biofilm inhibition and eradication of a pre-formed mature biofilm. RESULTS Among the anti-biofilm strategies being tested, the sub-minimal inhibitory concentration of several antibiotics either alone or in combination has been shown to inhibit biofilm formation and down-regulate the production of virulence factors. The combinatorial strategies include (1) combination of multiple antibiotics, (2) combination of antibiotics with non-antibiotic agents and (3) loading of antibiotics onto a carrier. CONCLUSION The present review paper describes the role of several antibiotics as biofilm inhibitors and also the alternative strategies adopted for applications in eradicating and inhibiting the formation of biofilm by pathogenic bacteria.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea.,Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201306, U.P., India
| | - Dung T N Pham
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| | - Sandra F Oloketuyi
- Laboratory for Environmental and Life Sciences, University of Nova Gorica 5000, Nova Gorica, Slovenia
| | - Young-Mog Kim
- Marine-Integrated Bionics Research Center, Pukyong National University, Busan 48513, Korea.,Department of Food Science and Technology, Pukyong National University, Busan 48513, Korea
| |
Collapse
|
41
|
Synergistic antimicrobial activity of melittin with clindamycin on the expression of encoding exfoliative toxin in Staphylococcus aureus. Toxicon 2020; 183:11-19. [PMID: 32450143 DOI: 10.1016/j.toxicon.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/23/2020] [Accepted: 05/08/2020] [Indexed: 10/24/2022]
Abstract
Staphylococcus aureus is an opportunistic human pathogens, with the ability to produce a series of virulence factors that contribute to the severity of infections. Exfoliative toxins (ETs) are one of the important virulence factors that participating in staphylococcal scalded skin syndrome. Melittin has different biological activities, comprising of antiviral, broad spectrum antibacterial, antiprotozoal, antifungal and anti-inflammatory effects. Twelve clinical isolates of methicillin-resistant S. aureus (MRSA) and methicillin-susceptible S. aureus (MSSA) were obtained from wound infection in the burn patients. The MIC plus three sub-inhibitory concentrations (I, II and III) of clindamycin and melittin were tested. Next, the synergistic effects of melittin and clindamycin were evaluated using the broth microdilution checkerboard assay. The detection of exfoliative toxin A and B genes were examined by PCR method. Then the effects of sub-MIC melittin on the expression levels of eta and etb were assessed by quantitative real-time PCR (qRT-PCR) assay. Melittin MIC values against MRSA and MSSA planktonic cells were 0.25-0.5 and 0.25-1 μg/ml, respectively. The clindamycin MIC values against MRSA and MSSA were between 0.5 and 8 μg/ml and 0.5-2 μg/ml, respectively. The results of the time-kill kinetics assay (3.5log10 and 3log10) against MSSA and MRSA planktonic cells were determined within 24 h using melittin. The mean expression of eta in MRSA and MSSA was significantly downregulated to approximately 3.5 and 4 fold, respectively. Moreover, the mean expression of etb in MRSA and MSSA were significantly downregulated to approximately 2.5 and 3 fold, respectively. Hemolytic assay showed that the extracted melittin indicates a strong hemolytic activity (HD50 = 2 μg/ml). Melittin at 0.5 μg/ml induced cell lysis and stimulated the formation of vesicles in S. aureus strains. Melittin could reduce the expression of eta and etb as encoding exfoliative toxin A and B genes. This component appears to be a good candidate for the treatment of MRSA and MSSA strains. So, melittin in combination with clindamycin can be classified as a complementary treatment of wound infections in burn patients.
Collapse
|
42
|
Pexiganan in Combination with Nisin to Control Polymicrobial Diabetic Foot Infections. Antibiotics (Basel) 2020; 9:antibiotics9030128. [PMID: 32244862 PMCID: PMC7148459 DOI: 10.3390/antibiotics9030128] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/22/2022] Open
Abstract
Diabetic foot ulcers (DFUs) are major complications of Diabetes mellitus being responsible for significant morbidity and mortality. DFUs frequently become chronically infected by a complex community of bacteria, including multidrug-resistant and biofilm-producing strains of Staphylococcus aureus and Pseudomonas aeruginosa. Diabetic foot infections (DFI) are often recalcitrant to conventional antibiotics and alternative treatment strategies are urgently needed. Antimicrobial Peptides (AMPs), such as pexiganan and nisin, have been increasingly investigated and reported as effective antimicrobial agents. Here, we evaluated the antibacterial potential of pexiganan and nisin used in combination (dual-AMP) to control the growth of planktonic and biofilm co-cultures of S. aureus and P. aeruginosa clinical strains, co-isolated from a DFU. A DFU collagen three-dimensional (3D) model was used to evaluate the distribution and efficacy of AMPs locally delivered into the model. The concentration of pexiganan required to inhibit and eradicate both planktonic and biofilm-based bacterial cells was substantially reduced when used in combination with nisin. Moreover, incorporation of both AMPs in a guar gum delivery system (dual-AMP biogel) did not affect the dual-AMP antimicrobial activity. Importantly, the application of the dual-AMP biogel resulted in the eradication of the S. aureus strain from the model. In conclusion, data suggest that the local application of the dual-AMPs biogel constitutes a potential complementary therapy for the treatment of infected DFU.
Collapse
|
43
|
Subinhibitory Concentrations of Mupirocin Stimulate Staphylococcus aureus Biofilm Formation by Upregulating cidA. Antimicrob Agents Chemother 2020; 64:AAC.01912-19. [PMID: 31932378 DOI: 10.1128/aac.01912-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/04/2019] [Indexed: 01/02/2023] Open
Abstract
Previous studies have shown that the administration of antibiotics at subinhibitory concentrations stimulates biofilm formation by the majority of multidrug-resistant Staphylococcus aureus (MRSA) strains. Here, we investigated the effect of subinhibitory concentrations of mupirocin on biofilm formation by the community-associated (CA) mupirocin-sensitive MRSA strain USA300 and the highly mupirocin-resistant clinical S. aureus SA01 to SA05 isolates. We found that mupirocin increased the ability of MRSA cells to attach to surfaces and form biofilms. Confocal laser scanning microscopy (CLSM) demonstrated that mupirocin treatment promoted thicker biofilm formation, which also correlated with the production of extracellular DNA (eDNA). Furthermore, quantitative real-time PCR (RT-qPCR) results revealed that this effect was largely due to the involvement of holin-like and antiholin-like proteins (encoded by the cidA gene), which are responsible for modulating cell death and lysis during biofilm development. We found that cidA expression levels significantly increased by 6.05- to 35.52-fold (P < 0.01) after mupirocin administration. We generated a cidA-deficient mutant of the USA300 S. aureus strain. Exposure of the ΔcidA mutant to mupirocin did not result in thicker biofilm formation than that in the parent strain. We therefore hypothesize that the mupirocin-induced stimulation of S. aureus biofilm formation may involve the upregulation of cidA.
Collapse
|
44
|
Cao Y, Yin H, Wang W, Pei P, Wang Y, Wang X, Jiang J, Luo SZ, Chen L. Killing Streptococcus mutans in mature biofilm with a combination of antimicrobial and antibiofilm peptides. Amino Acids 2019; 52:1-14. [PMID: 31797056 DOI: 10.1007/s00726-019-02804-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/11/2019] [Indexed: 10/25/2022]
Abstract
Biofilm poses a serious challenge for the treatment of bacterial infections, as it endows bacteria a pronounced resistance to traditional antibiotics. Antimicrobial peptides (AMPs) are considered potential substitutes for antibiotics. Combinational use of AMPs with other compounds to exert antibiofilm effects has been proved to be an effective means to reduce their toxicity and maximize their antimicrobial activity. However, the combination of various AMPs with different action mechanisms is rarely investigated. A newly designed lytic AMP ZXR-2.3 combined with antibiofilm peptide IDR-1018 or KT2 was tested for the antibiofilm effect on mature Streptococcus mutans biofilms. In general, the combination of ZXR-2.3 + IDR-1018 displayed synergistic effect on both biofilm eradication and bacterial killing, while ZXR-2.3 + KT2 showed no obvious synergism. The confocal images of preformed S. mutans biofilms confirmed the effective bactericidal activity of ZXR-2.3 + IDR-1018. A tube system was applied to investigate the biofilm infection under a flow of medium and SEM images indicated the biofilm disruption and bacterial killing effects of ZXR-2.3 + IDR-1018. Quantitative RT-PCR analysis showed that IDR-1018 induced dramatic changes in the mRNA expressions of the quorum sensing (QS) related genes comC, comD, vicR, and vicK of S. mutans in mature biofilms, whereas the other peptides and ciprofloxacin did not cause obvious changes in these genes. This might explain the better synergism of ZXR-2.3 and IDR-1018. The results of this study provide a potential application using the combination of different AMPs in the treatment of mature biofilm infection.
Collapse
Affiliation(s)
- Yimeng Cao
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Huiwei Yin
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Wujun Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Pengfei Pei
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Yin Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Xinyu Wang
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Jianhui Jiang
- Engineering Laboratory of Chemical Resources Utilization in South Xinjiang of Xinjiang Production and Construction Corps, College of Life Sciences, Tarim University, Alar, Xinjiang, 843300, China
| | - Shi-Zhong Luo
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Long Chen
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| |
Collapse
|
45
|
Ciandrini E, Morroni G, Cirioni O, Kamysz W, Kamysz E, Brescini L, Baffone W, Campana R. Synergistic combinations of antimicrobial peptides against biofilms of methicillin-resistant Staphylococcus aureus (MRSA) on polystyrene and medical devices. J Glob Antimicrob Resist 2019; 21:203-210. [PMID: 31678322 DOI: 10.1016/j.jgar.2019.10.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/10/2019] [Accepted: 10/21/2019] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Antimicrobial research is being focused to look for more effective therapeutics against antibiotic-resistant infections such as those caused by methicillin-resistant Staphylococcus aureus (MRSA). In this regard, antimicrobial peptides (AMPs) appear to be a promising solution. The aim of the present study was to investigate the potential activity of temporin A, citropin 1.1, CA(1-7)M(2-9)NH2 and Pal-KGK-NH2 in synergistic activity against MRSA biofilms developed on polystyrene surface (PSS) and central venous catheter (CVC). METHODS The study was subdivided into distinct phases to assess the ability of AMPs to inhibit biofilm formation, to identify possible synergy between AMPs, and to eradicate preformed biofilms on PSS and CVC using AMPs alone or in combination. RESULTS Activity of the AMPs was particularly evident in the inhibition of biofilm formation on PSS and CVC, whilst the eradication of preformed biofilms was more difficult and was reached only after 24h of contact. The synergistic activity of AMP combinations, selected by their fractional inhibitory concentration index (FICI), led to an improvement in the performance of all of the molecules in the removal of different biofilms. CONCLUSION Overall, AMPs could represent the next generation of antimicrobial agents for a prophylactic or therapeutic tool to control biofilms of antibiotic-resistant bacteria and/or biofilm-associated infections on different medical devices.
Collapse
Affiliation(s)
- Eleonora Ciandrini
- Department of Biomolecular Science, Division of Toxicological, Hygiene and Environmental Science, University of Urbino Carlo Bo, Via S. Chiara 27, 61029 Urbino, Italy
| | - Gianluca Morroni
- Infectious Diseases Clinic, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Oscar Cirioni
- Infectious Diseases Clinic, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Wojciech Kamysz
- Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | | | - Lucia Brescini
- Infectious Diseases Clinic, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Ancona, Italy
| | - Wally Baffone
- Department of Biomolecular Science, Division of Toxicological, Hygiene and Environmental Science, University of Urbino Carlo Bo, Via S. Chiara 27, 61029 Urbino, Italy
| | - Raffaella Campana
- Department of Biomolecular Science, Division of Toxicological, Hygiene and Environmental Science, University of Urbino Carlo Bo, Via S. Chiara 27, 61029 Urbino, Italy.
| |
Collapse
|
46
|
Non-Lytic Antibacterial Peptides That Translocate Through Bacterial Membranes to Act on Intracellular Targets. Int J Mol Sci 2019; 20:ijms20194877. [PMID: 31581426 PMCID: PMC6801614 DOI: 10.3390/ijms20194877] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/12/2019] [Accepted: 09/14/2019] [Indexed: 11/28/2022] Open
Abstract
The advent of multidrug resistance among pathogenic bacteria has attracted great attention worldwide. As a response to this growing challenge, diverse studies have focused on the development of novel anti-infective therapies, including antimicrobial peptides (AMPs). The biological properties of this class of antimicrobials have been thoroughly investigated, and membranolytic activities are the most reported mechanisms by which AMPs kill bacteria. Nevertheless, an increasing number of works have pointed to a different direction, in which AMPs are seen to be capable of displaying non-lytic modes of action by internalizing bacterial cells. In this context, this review focused on the description of the in vitro and in vivo antibacterial and antibiofilm activities of non-lytic AMPs, including indolicidin, buforin II PR-39, bactenecins, apidaecin, and drosocin, also shedding light on how AMPs interact with and further translocate through bacterial membranes to act on intracellular targets, including DNA, RNA, cell wall and protein synthesis.
Collapse
|
47
|
Maiden MM, Zachos MP, Waters CM. Hydrogels Embedded With Melittin and Tobramycin Are Effective Against Pseudomonas aeruginosa Biofilms in an Animal Wound Model. Front Microbiol 2019; 10:1348. [PMID: 31293530 PMCID: PMC6598697 DOI: 10.3389/fmicb.2019.01348] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022] Open
Abstract
We demonstrate that the antimicrobial peptide, melittin, is effective alone and in combination with the aminoglycosides tobramycin to kill Pseudomonas aeruginosa growing as biofilms both in vitro and in vivo. Melittin and tobramycin show enhanced in vitro activity in combination at micromolar concentrations, resulting in a 2-log10 reduction in the number of cells within mature PAO1 P. aeruginosa biofilms after 6-h of treatment. Alternatively, either agent alone resulted in half-a-log10 reduction. Time-killing assays demonstrated that the combination of melittin and tobramycin was effective at 2-h whereas tobramycin was not effective until after 6-h of treatment. We also found the combination was more effective than tobramycin alone against biofilms of 7 P. aeruginosa cystic fibrosis clinical isolates, resulting in a maximum 1.5-log10 cellular reduction. Additionally, melittin alone was effective at killing biofilms of 4 Staphylococcus aureus isolates, resulting in a maximum 2-log10 cellular reduction. Finally, melittin in combination with tobramycin embedded in an agarose-based hydrogel resulted in a 4-fold reduction in bioluminescent P. aeruginosa colonizing mouse wounds by 4-h. In contrast, tobramycin or melittin treatment alone did not cause a statistically significant reduction in bioluminescence. These data demonstrate that melittin in combination with tobramycin embedded in a hydrogel is a potential treatment for biofilm-associated wound infections.
Collapse
Affiliation(s)
- Michael M. Maiden
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
- The BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI, United States
| | - Mitchell P. Zachos
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| | - Christopher M. Waters
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
- The BEACON Center for the Study of Evolution in Action, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
48
|
Enzyme responsive copolymer micelles enhance the anti-biofilm efficacy of the antiseptic chlorhexidine. Int J Pharm 2019; 566:329-341. [PMID: 31152793 DOI: 10.1016/j.ijpharm.2019.05.069] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/09/2019] [Accepted: 05/27/2019] [Indexed: 12/22/2022]
Abstract
Staphylococcal biofilms cause many infectious diseases and are highly tolerant to the effects of antimicrobials; this is partly due to the biofilm matrix, which acts as a physical barrier retarding the penetration and reducing susceptibility to antimicrobials, thereby decreasing successful treatment outcomes. In this study, both single and mixed micellar systems based on poly vinyl caprolactam (PCL)-polyethylene glycol (PEG) copolymers were optimised for delivery of chlorhexidine (CHX) to S. aureus, MRSA and S. epidermidis biofilms and evaluated for their toxicity using Caenorhabditis elegans. The respective polyethylene glycol (PEG) and poly vinyl caprolactam (PCL) structural components promoted stealth properties and enzymatic responsive release of CHX inside biofilms, leading to significantly enhanced penetration (56%) compared with free CHX and improving the efficacy against Staphylococcus aureus biofilms grown on an artificial dermis (2.4 log reduction of CFU). Mixing Soluplus-based micelles with Solutol further enhanced the CHX penetration (71%) and promoted maximum reduction in biofilm biomass (>60%). Nematodes-based toxicity assay showed micelles with no lethal effects as indicated by their high survival rate (100%) after 72 h exposure. This study thus demonstrated that bio-responsive carriers can be designed to deliver a poorly water-soluble antimicrobial agent and advance the control of biofilm associated infections.
Collapse
|
49
|
Amso Z, Hayouka Z. Antimicrobial random peptide cocktails: a new approach to fight pathogenic bacteria. Chem Commun (Camb) 2019; 55:2007-2014. [PMID: 30688322 DOI: 10.1039/c8cc09961h] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Antibiotic resistance in bacteria has become a serious threat to public health, and therefore there is an urgent need to develop new classes of antimicrobial agents. Nowadays, natural antimicrobial peptides (AMPs) and their synthetic derivatives are considered as promising alternatives to traditional antibiotics. The broad molecular diversity of AMPs, in terms of sequences and structures, suggests that their activity does not depend on specific features of amino acid sequence or peptide conformation. We therefore selected two common properties of AMPs, (high percentage of hydrophobic and cationic amino acids), to develop a novel approach to synthesize random antimicrobial peptide mixtures (RPMs). Instead of incorporating a single amino acid at each coupling step, a mixture of hydrophobic and cationic amino acids in a defined proportion is coupled. This results in a mixture that contains up to 2n sequences, where n is the number of the coupling step, of random peptides with a defined composition, stereochemistry, and controlled chain length. We have discovered that RPMs of hydrophobic and cationic α-amino acids, such as phenylalanine and lysine, display strong and broad antimicrobial activity towards Gram-negative, Gram-positive, clinically isolated antibiotic resistant "superbugs", and several plant pathogenic bacteria. This review summarizes our efforts to explore the mode of action of RPMs and their potential as bioactive agents for multiple applications, including the prevention of biofilm formation and degradation of mature biofilm (related to human health), reduction of disease severity in plant bacterial disease models (related to crop protection), and inhibition of bacterial growth in milk (related to food preservation). All our findings illustrate the effectiveness of RPMs and their great potential for various applications.
Collapse
Affiliation(s)
- Zaid Amso
- The Scripps Research Institute, d/b/a Calibr, a division of Scripps Research, La Jolla, CA 92037, USA
| | | |
Collapse
|
50
|
Geitani R, Ayoub Moubareck C, Touqui L, Karam Sarkis D. Cationic antimicrobial peptides: alternatives and/or adjuvants to antibiotics active against methicillin-resistant Staphylococcus aureus and multidrug-resistant Pseudomonas aeruginosa. BMC Microbiol 2019; 19:54. [PMID: 30849936 PMCID: PMC6408789 DOI: 10.1186/s12866-019-1416-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 02/08/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Methicillin-resistant Staphylococcus aureus and multidrug-resistant Pseudomonas aeruginosa are becoming difficult to treat with antibiotics whereas Cationic Antimicrobial Peptides (CAMPs) represent promising alternatives. The effects of four CAMPs (LL-37: human cathelicidin, CAMA: cecropin(1-7)-melittin A(2-9) amide, magainin-II and nisin) were investigated against clinical and laboratory S. aureus (n = 10) and P. aeruginosa (n = 11) isolates either susceptible or resistant to antibiotics. Minimal Inhibitory Concentrations (MICs), Minimal Bactericidal Concentrations (MBCs), and bacterial survival rates (2 h post-treatment) were determined by microbroth dilution. The antipseudomonal effects of the antibiotics colistin or imipenem combined to LL-37 or CAMA were also studied. The toxicity of CAMPs used alone and in combination with antibiotics was evaluated on two human lung epithelial cell lines by determining the quantity of released cytoplasmic lactate dehydrogenase (LDH). Attempts to induce bacterial resistance to gentamicin, LL-37 or CAMA were also performed. RESULTS The results revealed the rapid antibacterial effect of LL-37 and CAMA against both antibiotic susceptible and resistant strains with almost a total reduction in bacterial count 2 h post-treatment. Magainin-II and nisin were less active against tested strains. When antibiotics were combined with LL-37 or CAMA, MICs of colistin decreased up to eight-fold and MICs of imipenem decreased up to four-fold. Cytotoxicity assays revealed non-significant LDH-release suggesting no cell damage in all experiments. Induction of bacterial resistance to LL-37 was transient, tardive and much lower than that to gentamicin and induction of resistance to CAMA was not observed. CONCLUSION This study showed the potent and rapid antibacterial activity of CAMPs on both laboratory and clinical isolates of S. aureus and P. aeruginosa either susceptible or resistant to antibiotics. Most importantly, CAMPs synergized the efficacy of antibiotics, had non toxic effects on human cells and were associated with transient and low levels of induced resistance.
Collapse
Affiliation(s)
- Regina Geitani
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon
| | - Carole Ayoub Moubareck
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon
- College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
| | - Lhousseine Touqui
- Unité de Mucoviscidose et Bronchopathies Chroniques, Institut Pasteur/Faculté de Médecine Cochin, Paris, France
| | - Dolla Karam Sarkis
- Microbiology Laboratory, School of Pharmacy, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|