1
|
Patra S, Saha S, Singh R, Tomar N, Gulati P. Biofilm battleground: Unveiling the hidden challenges, current approaches and future perspectives in combating biofilm associated bacterial infections. Microb Pathog 2025; 198:107155. [PMID: 39586337 DOI: 10.1016/j.micpath.2024.107155] [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: 07/31/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
A biofilm is a complex aggregation of microorganisms, either of the same or different species, that adhere to a surface and are encased in an extracellular polymeric substances (EPS) matrix. Quorum sensing (QS) and biofilm formation are closely linked, as QS genes regulate the development, maturation, and breakdown of biofilms. Inhibiting QS can be utilized as an effective approach to combat the impacts of biofilm infection. The impact of biofilms includes chronic infections, industrial biofouling, infrastructure corrosion, and environmental contamination as well. Therefore, a deep understanding of biofilms is crucial for enhancing public health, advancing industrial processes, safeguarding the environment, and deepening our knowledge of microbial life as well. This review aims to offer a comprehensive examination of challenges posed by bacterial biofilms, contemporary approaches and strategies for effectively eliminating biofilms, including the inhibition of quorum sensing pathways, while also focusing on emerging technologies and techniques for biofilm treatment. In addition, future research is projected to target the challenges associated with the bacterial biofilms, striving to develop new approaches and improve existing strategies for their effective control and eradication.
Collapse
Affiliation(s)
- Sandeep Patra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sumana Saha
- Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Randhir Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Nandini Tomar
- Department of Biotechnology, South Asian University, New Delhi, India
| | - Pallavi Gulati
- Ram Lal Anand College, University of Delhi, New Delhi, India.
| |
Collapse
|
2
|
Li M, Sun X, Zhao L, Du W, Shang D. The antibacterial activity and mechanisms of Trp-containing peptides against multidrug-resistant Pseudomonas aeruginosa persisters. Biochimie 2024; 225:133-145. [PMID: 38815647 DOI: 10.1016/j.biochi.2024.05.019] [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: 03/03/2024] [Revised: 04/27/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Bacterial persisters avoid antibiotic-mediated death by entering a dormant state and are considered a major cause of antibiotic treatment failure. Antimicrobial peptides (AMPs) with membrane-disrupting activity are promising drugs to eradicate persister cells. In this study, carbonyl cyanide m-chlorophenylhydrazone (CCCP), ciprofloxacin (CIP), and rifampicin (RFP) were applied to induce the formation of multidrug-resistant Pseudomonas aeruginosa (MRPA0108) persisters, and the antibacterial activity and mechanisms of I1W and L12W (two Trp-containing peptides designed in our lab) against MRPA0108 persisters were investigated. The results showed that I1W and L12W displayed potent antibacterial activity against MRPA0108 persisters. Both Trp-containing peptides disturbed the inner and outer membrane of MRPA0108 persisters. In addition, I1W and L12W revealed novel antibacterial mechanisms by decreasing the enzymatic activities of superoxide dismutase (SOD) and catalase (CAT), increasing reactive oxygen species (ROS) and malondialdehyde (MDA) levels, consequently leading to oxidative stress. The transcriptome profile of I1W-treated MRPA0108 persisters revealed that the genes involved in carbon metabolism, biosynthesis of amino acids, and the TCA cycle were downregulated, indicating that I1W interfered with metabolism and energy synthesis processes. Furthermore, both Trp-containing peptides displayed synergistic activities with antibiotic tobramycin and showed additive activities with cefepime or ciprofloxacin, which revealed a potential therapeutic strategy for the eradication of MRPA0108 persisters.
Collapse
Affiliation(s)
- Mengmiao Li
- School of Life Sciences, Liaoning Normal University, Dalian, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Xiaomi Sun
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Lei Zhao
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Wanying Du
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Dejing Shang
- School of Life Sciences, Liaoning Normal University, Dalian, China; Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China.
| |
Collapse
|
3
|
Farzi N, Oloomi M, Bahramali G, Siadat SD, Bouzari S. Antibacterial Properties and Efficacy of LL-37 Fragment GF-17D3 and Scolopendin A2 Peptides Against Resistant Clinical Strains of Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter baumannii In Vitro and In Vivo Model Studies. Probiotics Antimicrob Proteins 2024; 16:796-814. [PMID: 37148452 DOI: 10.1007/s12602-023-10070-w] [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] [Accepted: 03/21/2023] [Indexed: 05/08/2023]
Abstract
Pseudomonas aeruginosa, Staphylococcus aureus, and Acinetobacter baumannii have emerged as major clinical threats owing to the increasing prevalence of ventilator-associated pneumonia caused by multidrug-resistant or extensively drug-resistant strains. The present study aimed to assess the antibacterial effects and efficacy of LL-37 fragment GF-17D3 and synthetic Scolopendin A2 peptides against resistant clinical strains in vitro and in vivo models. P. aeruginosa, S. aureus, and A. baumannii were isolated from clinical infections. Their antibiotic resistance and minimum inhibitory concentration were assessed. LL-37 fragment GF-17D3 peptide was selected from available databases. Scolopendin A2 peptide's 6th amino acid (proline) was substituted with lysine and peptides and MICs were determined. The biofilm inhibitory activity was quantified at sub MIC concentrations. Synergetic effects of Scolopendin A2 and imipenem were assessed by checkerboard. After mice nasal infection with P. aeruginosa, peptides LD50 was determined. Isolates harbored complete resistance toward the majority of antibiotics and MIC values ranged between 1 and > 512 µg/ml. The majority of isolates exhibited strong biofilm activity. Synthetic peptides showed lower MIC values than antibiotic agents and the lowest MIC values were obtained for synthetic peptides in combination with antibiotics. The Synergisms effect of Scolopendin A2 with imipenem was also determined. Scolopendin A2 was found to have antibacterial efficacy against P. aeruginosa, S. aureus, and A. baumannii with MIC 64 µg/ml, 8 µg/ml, and 16 µg/ml, respectively, and LL37 showed antibacterial efficacy against P. aeruginosa, S. aureus, and A. baumannii with MIC 128 µg/ml, 32 µg/ml, and 32 µg/ml, respectively. Both AMPs decreased biofilms by ≥ 96% at 1 × MIC. The biofilm inhibitory activity was measured at sub MIC concentrations of the peptides and the results demonstrated that Scolopendin A2 exhibited anti-biofilm activity at 1/4 × MIC and 1/2 × MIC concentrations was 47.9 to 63.8%, although LL37 among 1/4 × MIC and 1/2 × MIC concentrations was 21.3 to 49.6% against three pathogens. The combination of Scolopendin A2 and antibiotics demonstrated synergistic activity-resistant strains with FIC values ≤ 0.5 for three pathogens, while LL37 and antibiotics showed synergistic activity FIC values ≤ 0.5 for only P. aeruginosa. Infection model Scolopendin A2 with Imipenem (2 × MIC) was efficacious in vivo, with a 100% survival rate following treatment at 2 × MIC after 120 h. The mRNA expression of biofilm-related genes was decreased for both peptides. Synthesis Scolopendin A2 decreased the expression of biofilm formation genes compared to the control group. Synthetic Scolopendin A2 exhibits antimicrobial activity without causing toxicity on the human epithelial cell line. Based on our findings, it seems that synthetic Scolopendin A2 is an appropriate antimicrobial source. That could be a promising option in combination with antibiotics for a topical medication and in the prevention of acute and chronic infections caused by multidrug-resistant bacteria. Nevertheless, additional experiments are required to assess another potential of this novel AMP.
Collapse
Affiliation(s)
- Nastaran Farzi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mana Oloomi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Golnaz Bahramali
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran.
- Department of Hepatitis and AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran.
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Hale SJM, Cameron AJ, Lux CA, Biswas K, Kim R, O'Carroll M, Harris PWR, Douglas RG, Wagner Mackenzie B. Polymyxin B and ethylenediaminetetraacetic acid act synergistically against Pseudomonas aeruginosa and Staphylococcus aureus. Microbiol Spectr 2024; 12:e0170923. [PMID: 38168683 PMCID: PMC10845947 DOI: 10.1128/spectrum.01709-23] [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: 04/25/2023] [Accepted: 10/05/2023] [Indexed: 01/05/2024] Open
Abstract
Polymyxin B and ethylenediaminetetraacetic acid are antimicrobials possessing antibiofilm activity. They act by displacement and chelation, respectively, of divalent cations in bacterial membranes and may therefore act synergistically when applied in combination. If so, this combination of agents may be useful for the treatment of diseases like cystic fibrosis (CF), in which biofilms are present on the respiratory epithelium. We used checkerboard assays to investigate the synergy between these agents using reference strains Pseudomonas aeruginosa ATCC 27853 and Staphylococcus aureus ATCC 6538 in planktonic form. We then determined the efficacy of each agent against biofilms of both species grown on 96-pin lids and proceeded to combination testing against the P. aeruginosa reference strain and 10 clinical isolates from patients with CF. Synergism was observed for planktonic forms of both species and for biofilms of P. aeruginosa. The susceptibility of biofilms of P. aeruginosa clinical isolates to these agents was variable compared to the laboratory reference strain. This combination of agents may be useful in the management of biofilm-associated conditions, particularly those amenable to topical therapies. These results provide a basis upon which the antimicrobial and antibiofilm efficacy of preparations containing these agents may be enhanced.IMPORTANCEBacteria living in biofilms produce a protective matrix which makes them difficult to kill. Patients with severe respiratory disease often have biofilms. Polymyxin B is an antibiotic commonly used in topical medications, such as eye drops and nasal sprays. Ethylenediaminetetraacetic acid (EDTA) is used widely as a preservative in medication but also has antimicrobial properties. It has been hypothesized that Polymyxin B and EDTA could have a synergistic relationship: when used in combination their antimicrobial effect is enhanced. Here, we evaluated the levels at which Polymyxin B and EDTA work together to kill common pathogens Pseudomonas aeruginosa and Staphylococcus aureus. We found that Polymyxin B and EDTA were synergistic. This synergy may be useful in the management of planktonic infection with P. aeruginosa and S. aureus, or biofilm infection with P. aeruginosa. This synergy may be beneficial in the treatment of respiratory biofilms, in which P. aeruginosa biofilms are common.
Collapse
Affiliation(s)
- Samuel J M Hale
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Alan J Cameron
- School of Chemical Sciences and School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Christian A Lux
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Raymond Kim
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Mark O'Carroll
- Respiratory Services, Auckland City Hospital, Te Toka Tumai, Te Whatu Ora, Auckland, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences and School of Biological Sciences, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| | - Brett Wagner Mackenzie
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
5
|
Mone NS, Syed S, Ravichandiran P, Kamble EE, Pardesi KR, Salunke-Gawali S, Rai M, Vikram Singh A, Prasad Dakua S, Park BH, Yoo DJ, Satpute SK. Synergistic and Additive Effects of Menadione in Combination with Antibiotics on Multidrug-Resistant Staphylococcus aureus: Insights from Structure-Function Analysis of Naphthoquinones. ChemMedChem 2023; 18:e202300328. [PMID: 37874976 DOI: 10.1002/cmdc.202300328] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 10/26/2023]
Abstract
Antimicrobial resistance (AMR) interferes with the effective treatment of infections and increases the risk of microbial spread and infection-related illness and death. The synergistic activities of combinations of antimicrobial compounds offer satisfactory approaches to some extent. Structurally diverse naphthoquinones (NQs) including menadione (-CH3 group at C2) exhibit substantial antimicrobial activities against multidrug-resistant (MDR) pathogens. We explored the combinations of menadione with antibiotic ciprofloxacin or ampicillin against Staphylococcus aureus and its biofilms. We found an additive (0.590 %) were also observed. However, preformed biofilms were not affected. Dent formation was also evident in S. aureus treated with the test compounds. The structure-function relationship (SFR) of NQs was used to determine and predict their activity pattern against pathogens. Analysis of 10 structurally distinct NQs revealed that the compounds with -Cl, -Br, -CH3 , or -OH groups displayed the lowest MICs (32-256 μg/mL). Furthermore, 1,4-NQs possessing a halogen or -CH3 moiety showed elevated ROS activity, whereas molecules with an -OH group affected cell integrity. Improved activity of antimicrobial combinations and SFR approaches are significant in antimicrobial therapies.
Collapse
Affiliation(s)
- Nishigandha S Mone
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, 411007, India
| | - Sahil Syed
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, 411007, India
| | - Palanisamy Ravichandiran
- Department of Energy Storage/Conversion Engineering (BK21 FOUR) of Graduate School, Hydrogen and Fuel Cell Research Center, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Department of Life Science, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| | - Ekta E Kamble
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, 411007, India
| | - Karishma R Pardesi
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, 411007, India
| | - Sunita Salunke-Gawali
- Department of Chemistry, Savitribai Phule Pune University, Pune, Maharashtra m, 411007, India
| | - Mansi Rai
- Department of Microbiology, Central University of Rajasthan Ajmer, Rajasthan, 305817, India
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute (BfR), Max-Dohrn-Str. 8-10, 10589, Berlin, Germany
| | | | - Byung-Hyun Park
- Department of Biochemistry, Jeonbuk National University Medical School, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| | - Dong Jin Yoo
- Department of Energy Storage/Conversion Engineering (BK21 FOUR) of Graduate School, Hydrogen and Fuel Cell Research Center, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Department of Life Science, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
| | - Surekha K Satpute
- Department of Microbiology, Savitribai Phule Pune University, Pune, Maharashtra, 411007, India
| |
Collapse
|
6
|
Zhang W, An Z, Bai Y, Zhou Y, Chen F, Wang KJ. A novel antimicrobial peptide Scyreptin 1-30 from Scylla paramamosain exhibiting potential therapy of Pseudomonas aeruginosa early infection in a mouse burn wound model. Biochem Pharmacol 2023; 218:115917. [PMID: 37952897 DOI: 10.1016/j.bcp.2023.115917] [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: 09/14/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
Antimicrobial resistance (AMR) constitutes a significant global threat to human health. In recent years, there has been a concerning surge in infections caused by multidrug-resistant bacteria, highlighting the pressing need to urgently explore novel and effective alternatives to conventional antibiotics. Antimicrobial peptides (AMPs) have emerged as a focal point of research, capturing significant attention as promising antimicrobial agents. In this study, we have identified a novel cationic antimicrobial peptide (AMP) named Scyreptin1-30, derived from the marine invertebrate Scylla paramamosain. The results showed that Scyreptin1-30 exhibits a broad-spectrum antimicrobial activity, demonstrating significant potency against both bacteria and fungi, and even against the clinically isolated multidrug-resistant bacteria Pseudomonas aeruginosa. Moreover, Scyreptin1-30 exhibited rapid bactericidal kinetic. The results of antibacterial mechanism showed that Scyreptin1-30 destroyed the integrity of bacterial membranes, leading to bacterial death and exhibited potent anti-biofilm activity against P. aeruginosa. The activity of Scyreptin1-30 against bacteria had a favorable thermal stability, displayed a certain ion tolerance, and showed no discernible cytotoxicity when assessed against both the mammalian cell line HEK293T and the fish cell lines ZF4. In an In vivo study, Scyreptin1-30 exhibited a remarkably reduction in the bacterial load caused by multidrug-resistant P. aeruginosa at the site of infection, and promoted wound healing in a mouse model of burn infection. This study indicated that Scyreptin1-30 holds promise as an effective antibacterial agent, potentially serving as a topical skin treatment against multidrug-resistant bacterial infections, including those caused by P. aeruginosa.
Collapse
Affiliation(s)
- Weibin Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhe An
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yuqi Bai
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Ying Zhou
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China; State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
7
|
Chiloeches A, Zágora J, Plachá D, Torres MDT, de la Fuente-Nunez C, López-Fabal F, Gil-Romero Y, Fernández-García R, Fernández-García M, Echeverría C, Muñoz-Bonilla A. Synergistic Combination of Antimicrobial Peptides and Cationic Polyitaconates in Multifunctional PLA Fibers. ACS APPLIED BIO MATERIALS 2023; 6:4805-4813. [PMID: 37862451 PMCID: PMC10852355 DOI: 10.1021/acsabm.3c00576] [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: 07/25/2023] [Accepted: 10/05/2023] [Indexed: 10/22/2023]
Abstract
Combining different antimicrobial agents has emerged as a promising strategy to enhance efficacy and address resistance evolution. In this study, we investigated the synergistic antimicrobial effect of a cationic biobased polymer and the antimicrobial peptide (AMP) temporin L, with the goal of developing multifunctional electrospun fibers for potential biomedical applications, particularly in wound dressing. A clickable polymer with pendent alkyne groups was synthesized by using a biobased itaconic acid building block. Subsequently, the polymer was functionalized through click chemistry with thiazolium groups derived from vitamin B1 (PTTIQ), as well as a combination of thiazolium and AMP temporin L, resulting in a conjugate polymer-peptide (PTTIQ-AMP). The individual and combined effects of the cationic PTTIQ, Temporin L, and PTTIQ-AMP were evaluated against Gram-positive and Gram-negative bacteria as well as Candida species. The results demonstrated that most combinations exhibited an indifferent effect, whereas the covalently conjugated PTTIQ-AMP displayed an antagonistic effect, potentially attributed to the aggregation process. Both antimicrobial compounds, PTTIQ and temporin L, were incorporated into poly(lactic acid) electrospun fibers using the supercritical solvent impregnation method. This approach yielded fibers with improved antibacterial performance, as a result of the potent activity exerted by the AMP and the nonleaching nature of the cationic polymer, thereby enhancing long-term effectiveness.
Collapse
Affiliation(s)
- Alberto Chiloeches
- Instituto
de Ciencia y Tecnología de Polímeros (ICTP-CSIC), C/Juan de la Cierva 3, Madrid 28006, Spain
- Universidad
Nacional de Educación a Distancia (UNED), C/Bravo Murillo 38, Madrid 28015, Spain
| | - Jakub Zágora
- Nanotechnology
Centre, CEET, VSB—Technical University
of Ostrava, 17. Listopadu 2172/15, Ostrava-Poruba 708 00, Czech Republic
| | - Daniela Plachá
- Nanotechnology
Centre, CEET, VSB—Technical University
of Ostrava, 17. Listopadu 2172/15, Ostrava-Poruba 708 00, Czech Republic
| | - Marcelo D. T. Torres
- Machine
Biology Group, Departments of Psychiatry and Microbiology, Institute
for Biomedical Informatics, Institute for Translational Medicine and
Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments
of Bioengineering and Chemical and Biomolecular Engineering, School
of Engineering and Applied Science, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute
for Computational Science, University of
Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Cesar de la Fuente-Nunez
- Machine
Biology Group, Departments of Psychiatry and Microbiology, Institute
for Biomedical Informatics, Institute for Translational Medicine and
Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments
of Bioengineering and Chemical and Biomolecular Engineering, School
of Engineering and Applied Science, University
of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute
for Computational Science, University of
Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Fátima López-Fabal
- Hospital
Universitario de Móstoles C/Dr. Luis Montes, s/n, Móstoles 28935, Madrid, Spain
- Facultad
de Ciencias Experimentales, Universidad
Francisco de Vitoria, Carretera Pozuelo a Majadahonda, Km 1.800, Madrid 28223, Spain
| | - Yolanda Gil-Romero
- Hospital
Universitario de Móstoles C/Dr. Luis Montes, s/n, Móstoles 28935, Madrid, Spain
| | | | - Marta Fernández-García
- Instituto
de Ciencia y Tecnología de Polímeros (ICTP-CSIC), C/Juan de la Cierva 3, Madrid 28006, Spain
| | - Coro Echeverría
- Instituto
de Ciencia y Tecnología de Polímeros (ICTP-CSIC), C/Juan de la Cierva 3, Madrid 28006, Spain
| | - Alexandra Muñoz-Bonilla
- Instituto
de Ciencia y Tecnología de Polímeros (ICTP-CSIC), C/Juan de la Cierva 3, Madrid 28006, Spain
| |
Collapse
|
8
|
Abdulkareem AH, Alalwani AK, Ahmed MM, Al-Meani SAL, Al-Janaby MS, Al-Qaysi AMK, Edan AI, Lahij HF. Impact of Solidago virgaurea Extract on Biofilm Formation for ESBL- Pseudomonas aeruginosa: An In Vitro Model Study. Pharmaceuticals (Basel) 2023; 16:1383. [PMID: 37895854 PMCID: PMC10609828 DOI: 10.3390/ph16101383] [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: 07/25/2023] [Revised: 08/27/2023] [Accepted: 09/09/2023] [Indexed: 10/29/2023] Open
Abstract
The increasing disparity between antimicrobial resistance (AMR) and the development of new antimicrobials continues to pose a significant global health concern. However, plant extracts have shown promise in combating this issue either through their inherent antimicrobial activity or by serving as potential reservoirs of effective antimicrobial compounds. These compounds have the ability to target pathogenic biofilms and inhibit the production of extended-spectrum β -lactamases (ESBLs). However, there is limited research available on the antibacterial properties of goldenrod extract. Thus, the objective of this study was to investigate the impact of S. virgaurea (SV) extract on the viability and ability to form biofilms of ESBL-Pseudomonas aeruginosa (P. aeruginosa). A cross-sectional study was conducted from August 2022 to March 2023. The broth microdilution method was employed to determine the minimum inhibitory concentration (MIC) of the (SV) extract. Subsequently, the minimum bactericidal concentration (MBC) was determined based on the MIC values obtained. The antibiotic susceptibility of bacteria was evaluated using the Kirby disk diffusion assay and an Antimicrobial Susceptibility Testing (AST) card in conjunction with the Vitek-2 compact system. Biofilm formation was evaluated using Congo red and a 96-well Elisa plate, while the presence of extended-spectrum β-lactamases (ESBLs) was estimated by measuring the reduction of nitrocefin at a wavelength of 390 nm. In addition, treatment of biofilm and ESBL activity with SV extract using 96-well Elisa plate and nitrocefin hydrolyzing, respectively. The resistance rates of P. aeruginosa isolates to the tested antibiotics were as follows: Levofloxacin 33%, Ciprofloxacin 40%, Amikacin 49%, Meropenem 50%, Cefepime 70%, Ceftazidime 75%, Cefotaxime 85%, Piperacillin-Tazobactam 90%, Amoxiclav 97%, Ampicillin 99%, Ceftriaxone 100%. The prevalence of MDR-P. aeruginosa, XDR-P. aeruginosa, PDR-P. aeruginosa and non-MDR-PA were 40% (n = 40), 7% (n = 7), 3% (n = 3) and 50% (n = 50), respectively. From the GC-MS results, it was observed that the presence of Octadecane, Clioquinol, Glycerol tricaprylate, hexadecanoic acid, cis-13-octadecenoic acid, oleic acid and Propanamide were the major components in the Solidago extract. In the determination of plant crude extracts, the values ranged between 0.25 and 64 mg/mL against bacteria. The resulting activity of the extract showed a significant statistical relationship at a p-value ≤ 0.01 against ESBL production and biofilm formation in P. aeruginosa. The S. virgaurea extract exhibited effectiveness in inhibiting biofilm formation and combating P. aeruginosa strains that produce extended-spectrum β-lactamases (ESBLs).
Collapse
Affiliation(s)
- Ali Hazim Abdulkareem
- Department of Biotechnology, College of Science, University of Anbar, Ramadi 31001, Iraq; (A.H.A.); (A.K.A.); (S.A.L.A.-M.); (M.S.A.-J.); (A.-M.K.A.-Q.)
| | - Anmar Kamil Alalwani
- Department of Biotechnology, College of Science, University of Anbar, Ramadi 31001, Iraq; (A.H.A.); (A.K.A.); (S.A.L.A.-M.); (M.S.A.-J.); (A.-M.K.A.-Q.)
| | - Mohammed Mukhles Ahmed
- Department of Biotechnology, College of Science, University of Anbar, Ramadi 31001, Iraq; (A.H.A.); (A.K.A.); (S.A.L.A.-M.); (M.S.A.-J.); (A.-M.K.A.-Q.)
| | - Safaa Abed Latef Al-Meani
- Department of Biotechnology, College of Science, University of Anbar, Ramadi 31001, Iraq; (A.H.A.); (A.K.A.); (S.A.L.A.-M.); (M.S.A.-J.); (A.-M.K.A.-Q.)
| | - Mohammed Salih Al-Janaby
- Department of Biotechnology, College of Science, University of Anbar, Ramadi 31001, Iraq; (A.H.A.); (A.K.A.); (S.A.L.A.-M.); (M.S.A.-J.); (A.-M.K.A.-Q.)
| | - Al-Moghira Khairi Al-Qaysi
- Department of Biotechnology, College of Science, University of Anbar, Ramadi 31001, Iraq; (A.H.A.); (A.K.A.); (S.A.L.A.-M.); (M.S.A.-J.); (A.-M.K.A.-Q.)
| | - Ali Ibrahim Edan
- Medical Laboratory Technology, Al-Huda University College, Ramadi 31001, Iraq;
| | - Hasan Falah Lahij
- Medical Laboratory Technology, Almaarif University College, Ramadi 31001, Iraq;
| |
Collapse
|
9
|
Lv J, Liu G, Ju Y, Huang H, Sun Y. AADB: A Manually Collected Database for Combinations of Antibiotics With Adjuvants. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:2827-2836. [PMID: 37279138 DOI: 10.1109/tcbb.2023.3283221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Antimicrobial resistance is a global public health concern. The lack of innovations in antibiotic development has led to renewed interest in antibiotic adjuvants. However, there is no database to collect antibiotic adjuvants. Herein, we build a comprehensive database named Antibiotic Adjuvant DataBase (AADB) by manually collecting relevant literature. Specifically, AADB includes 3,035 combinations of antibiotics with adjuvants, covering 83 antibiotics, 226 adjuvants, and 325 bacterial strains. AADB provides user-friendly interfaces for searching and downloading. Users can easily obtain these datasets for further analysis. In addition, we also collected related datasets (e.g., chemogenomic and metabolomic data) and proposed a computational strategy to dissect these datasets. As a test case, we identified 10 candidates for minocycline, and 6 of 10 candidates are the known adjuvants that synergize with minocycline to inhibit the growth of E. coli BW25113. We hope that AADB can help users to identify effective antibiotic adjuvants. AADB is freely available at http://www.acdb.plus/AADB.
Collapse
|
10
|
Synergy between Human Peptide LL-37 and Polymyxin B against Planktonic and Biofilm Cells of Escherichia coli and Pseudomonas aeruginosa. Antibiotics (Basel) 2023; 12:antibiotics12020389. [PMID: 36830299 PMCID: PMC9952724 DOI: 10.3390/antibiotics12020389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 02/01/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The rise in antimicrobial resistant bacteria is limiting the number of effective treatments for bacterial infections. Escherichia coli and Pseudomonas aeruginosa are two of the pathogens with the highest prevalence of resistance, and with the greatest need for new antimicrobial agents. Combinations of antimicrobial peptides (AMPs) and antibiotics that display synergistic effects have been shown to be an effective strategy in the development of novel therapeutic agents. In this study, we investigated the synergy between the AMP LL-37 and various classes of antibiotics against E. coli and P. aeruginosa strains. Of the six antibiotics tested (ampicillin, tetracycline, ciprofloxacin, gentamicin, aztreonam, and polymyxin B (PMB)), LL-37 displayed the strongest synergy against E. coli MG1655 and P. aeruginosa PAO1 laboratory strains when combined with PMB. Given the strong synergy, the PMB + LL-37 combination was chosen for further examination where it demonstrated synergy against multidrug-resistant and clinical E. coli isolates. Synergy of PMB + LL-37 towards clinical isolates of P. aeruginosa varied and showed synergistic, additive, or indifferent effects. The PMB + LL-37 combination treatment showed significant prevention of biofilm formation as well as eradication of pre-grown E. coli and P. aeruginosa biofilms. Using the Galleria mellonella wax worm model, we showed that the PMB + LL-37 combination treatment retained its antibacterial capacities in vivo. Flow analyses were performed to characterize the mode of action. The results of the present study provide proof of principle for the synergistic response between LL-37 and PMB and give novel insights into a promising new antimicrobial combination against gram-negative planktonic and biofilm cells.
Collapse
|
11
|
Bari AK, Belalekar TS, Poojary A, Rohra S. Combination drug strategies for biofilm eradication using synthetic and natural agents in KAPE pathogens. Front Cell Infect Microbiol 2023; 13:1155699. [PMID: 37139491 PMCID: PMC10149696 DOI: 10.3389/fcimb.2023.1155699] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/29/2023] [Indexed: 05/05/2023] Open
Abstract
Antibiotic resistance is a global threat caused by factors such as overuse of antibiotics, lack of awareness, development of biofilms etc. World Health Organization released a list of global priority pathogens which consisted of 12 species of bacteria categorized as expressing critical, high and medium resistance. Several Gram-negative and Gram-positive species are known to cause wide varieties of infections and have become multidrug or extremely drug resistant. Pathogens causing infections associated with invasive medical devices are biofilm producers and hence their treatment becomes difficult due to a structurally stable matrix which prevents antibiotics from penetrating the biofilm and thereby showing its effects. Factors contributing to tolerance are inhibition of penetration, restricted growth and activation of biofilm genes. Combination drug therapies has also shown potential to eradicate biofilm infections. A combination of inhaled Fosfomycin/tobramycin antibiotic strategy has been effective against Gram-negative as well as Gram positive organisms. Along with antibiotics, use of natural or synthetic adjuvants shows promising effects to treat biofilm infections. Fluroquinolone activity on biofilms is disrupted by low oxygen tension in the matrix, a strategy known as hyperbaric oxygen treatment that can enhance efficacy of antibiotics if well optimized. Adjuvants such as Ethylenediaminetetraacetic acid (EDTA), Sodium Dodecyl Sulphate (SDS) and chlorhexidine act by killing non-growing microbial cells aggregated on the inner layer of the biofilm. This review aims to list down current combination therapies used against Gram-negative and Gram-positive biofilm forming pathogens and brief about comparison of combination drugs and their efficacies.
Collapse
|
12
|
Momenzadeh M, Soltani R, Shafiee F, Hakamifard A, Pourahmad M, Abbasi S. The effectiveness of colistin/levofloxacin compared to colistin/meropenem in the treatment of ventilator-associated pneumonia (VAP) caused by carbapenem-resistant Acinetobacter baumannii: a randomized controlled clinical trial. Res Pharm Sci 2022; 18:39-48. [PMID: 36846731 PMCID: PMC9951781 DOI: 10.4103/1735-5362.363594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Accepted: 10/28/2022] [Indexed: 12/25/2022] Open
Abstract
Background and purpose The treatment of ventilator-associated pneumonia (VAP) caused by carbapenem-resistant Acinetobacter baumannii (CRAB) is still a great challenge. This study evaluated the effectiveness of the colistin/levofloxacin regimen compared to the usual colistin/meropenem regimen in the treatment of patients with VAP caused by CRAB. Experimental approach The patients with VAP were randomly assigned to experimental (n = 26) and control (n = 29) groups. The first group received IV colistin 4.5 MIU every 12 h + levofloxacin 750 mg IV daily, and the second group received IV colistin with the same dose + meropenem 1 g IV every 8 h for 10 days. The clinical (complete response, partial response, or treatment failure) and microbiological responses at the end of the intervention were recorded and compared between the two groups. Findings/Results The complete response rate was higher (n = 7; 35%) and the failure rate was lower (n = 4; 20%) in the experimental group than in the control group (n = 2; 8%, and n = 11; 44%, respectively), but the differences were not statistically significant. Even though the microbiological response rate was higher in the experimental group (n = 14; 70%) than in the control group (n = 12; 48%), the difference was not statistically significant. The mortality rate was 6 (23.10%) and 4 patients (13.8%) in the experimental and control groups, respectively (P = 0.490). Conclusion and implication The levofloxacin/colistin combination can be considered an alternative regimen to meropenem/colistin in the treatment of VAP caused by CRAB.
Collapse
Affiliation(s)
- Mahnaz Momenzadeh
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan, I.R. Iran
| | - Rasool Soltani
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Isfahan, I.R. Iran,Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, I.R. Iran,Corresponding author: R. Soltani Tel: +98-3137927067, Fax: +98-3136680011
| | - Fatemeh Shafiee
- Deparment of Pharmaceutical Biotechnology, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Atousa Hakamifard
- Department of Infectious Diseases, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran,Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, I.R. Iran
| | - Morteza Pourahmad
- Department of Infectious Diseases, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Saeed Abbasi
- Anaesthesiology Department, School of Medicine, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
13
|
Chen YC, Qiu W, Zhang W, Zhang J, Chen R, Chen F, Wang KJ. A Novel Antimicrobial Peptide Sp-LECin with Broad-Spectrum Antimicrobial Activity and Anti- Pseudomonas aeruginosa Infection in Zebrafish. Int J Mol Sci 2022; 24:ijms24010267. [PMID: 36613722 PMCID: PMC9820466 DOI: 10.3390/ijms24010267] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
New antimicrobial agents are urgently needed to address the increasing emergence and dissemination of multidrug-resistant bacteria. In the study, a chemically synthesized truncated peptide containing 22-amino acids derived from a C-type lectin homolog SpCTL6 of Scylla paramamosain was screened and found to exhibit broad-spectrum antimicrobial activity, indicating that it is an antimicrobial peptide (AMP), named Sp-LECin. Sp-LECin possessed the basic characteristics of most cationic AMPs, such as positive charge (+4) and a relatively high hydrophobicity (45%). After treatment with Sp-LECin, the disruption of microbial membrane integrity and even leakage of cellular contents was observed by scanning electron microscopy (SEM). In addition, Sp-LECin could bind lipopolysaccharide (LPS), increase the outer and inner membrane permeability and induce reactive oxygen species (ROS) production, ultimately leading to the death of Pseudomonas aeruginosa. Furthermore, Sp-LECin exhibited potent anti-biofilm activity against P. aeruginosa during both biofilm formation and maturation. Notably, Sp-LECin had no obvious cytotoxicity and could greatly improve the survival of P. aeruginosa-infected zebrafish, by approximately 40% over the control group after 72 h of treatment. This study indicated that Sp-LECin is a promising antibacterial agent with the potential to be used against devastating global pathogen infections such as P. aeruginosa.
Collapse
Affiliation(s)
- Yan-Chao Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Wanlei Qiu
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Weibin Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Jingrong Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Roushi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| |
Collapse
|
14
|
Schwartz T, Schewe N, Schwotzer M, Heinle M, Mahmood A, Krolla P, Thissen P. Antibacterial Inorganic Coating of Calcium Silicate Hydrate Substrates by Copper Incorporation. ACS APPLIED BIO MATERIALS 2022; 5:5190-5198. [PMID: 36280235 PMCID: PMC9683100 DOI: 10.1021/acsabm.2c00616] [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: 07/12/2022] [Accepted: 10/05/2022] [Indexed: 01/25/2023]
Abstract
Under environmental conditions, biofilms can oftentimes be found on different surfaces, accompanied by the structural degradation of the substrate. Since high-copper-content paints were banned in the EU, a solution for the protection of these surfaces has to be found. In addition to hydrophobation, making the surfaces inherently biofilm-repellent is a valid strategy. We want to accomplish this via the metal exchange in calcium silicate hydrate (CSH) substrates with transition metals. As has been shown with Europium, even small amounts of metal can have a great influence on the material properties. To effectively model CSH surfaces, ultrathin CSH films were grown on silicon wafers using Ca(OH)2 solutions. Subsequently, copper was incorporated as an active component via ion exchange. Biofilm development is quantified using a multiple-resistant Pseudomonas aeruginosa strain described as a strong biofilm former cultivated in the culture medium for 24 h. Comprehensive structural and chemical analyses of the substrates are done by environmental scanning electron microscopy (ESEM), transmission Fourier transform infrared spectroscopy (FT-IR), X-ray photoelectron spectroscopy (XPS), and time-of-flight secondary ion mass spectrometry (ToF-SIMS). Results do not show any structural deformation of the substrates by the incorporation of the Cu combined with three-dimensional (3D) homogeneous distribution. While the copper-free CSH phase shows a completely random distribution of the bacteria in biofilms, the samples with copper incorporation reveal lower bacterial colonization of the modified surfaces with an enhanced cluster formation.
Collapse
Affiliation(s)
- Thomas Schwartz
- Institut
für Funktionelle Grenzflächen (IFG), Karlsruher Institut für Technologie (KIT), Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Deutschland
| | - Nils Schewe
- Institut
für Funktionelle Grenzflächen (IFG), Karlsruher Institut für Technologie (KIT), Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Deutschland
| | - Matthias Schwotzer
- Institut
für Funktionelle Grenzflächen (IFG), Karlsruher Institut für Technologie (KIT), Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Deutschland
| | - Marita Heinle
- Institut
für Funktionelle Grenzflächen (IFG), Karlsruher Institut für Technologie (KIT), Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Deutschland
| | - Ammar Mahmood
- Institut
für Massivbau und Baustofftechnologie (IMB), Karlsruher Institut für Technologie (KIT), Gotthard-Franz-Str. 3, 76131Karlsruhe, Deutschland
| | - Peter Krolla
- Institut
für Funktionelle Grenzflächen (IFG), Karlsruher Institut für Technologie (KIT), Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Deutschland
| | - Peter Thissen
- Institut
für Funktionelle Grenzflächen (IFG), Karlsruher Institut für Technologie (KIT), Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Deutschland
- Institut
für Massivbau und Baustofftechnologie (IMB), Karlsruher Institut für Technologie (KIT), Gotthard-Franz-Str. 3, 76131Karlsruhe, Deutschland
| |
Collapse
|
15
|
Chiu S, Hancock AM, Schofner BW, Sniezek KJ, Soto-Echevarria N, Leon G, Sivaloganathan DM, Wan X, Brynildsen MP. Causes of polymyxin treatment failure and new derivatives to fill the gap. J Antibiot (Tokyo) 2022; 75:593-609. [PMID: 36123537 DOI: 10.1038/s41429-022-00561-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/08/2022]
Abstract
Polymyxins are a class of antibiotics that were discovered in 1947 from programs searching for compounds effective in the treatment of Gram-negative infections. Produced by the Gram-positive bacterium Paenibacillus polymyxa and composed of a cyclic peptide chain with a peptide-fatty acyl tail, polymyxins exert bactericidal effects through membrane disruption. Currently, polymyxin B and colistin (polymyxin E) have been developed for clinical use, where they are reserved as "last-line" therapies for multidrug-resistant (MDR) infections. Unfortunately, the incidences of strains resistant to polymyxins have been increasing globally, and polymyxin heteroresistance has been gaining appreciation as an important clinical challenge. These phenomena, along with bacterial tolerance to this antibiotic class, constitute important contributors to polymyxin treatment failure. Here, we review polymyxins and their mechanism of action, summarize the current understanding of how polymyxin treatment fails, and discuss how the next generation of polymyxins holds promise to invigorate this antibiotic class.
Collapse
Affiliation(s)
- Selena Chiu
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Anna M Hancock
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Bob W Schofner
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Katherine J Sniezek
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | | | - Gabrielle Leon
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | | | - Xuanqing Wan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Mark P Brynildsen
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA.
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
16
|
Jiang M, Chen R, Zhang J, Chen F, Wang KJ. A Novel Antimicrobial Peptide Spampcin 56-86 from Scylla paramamosain Exerting Rapid Bactericidal and Anti-Biofilm Activity In Vitro and Anti-Infection In Vivo. Int J Mol Sci 2022; 23:ijms232113316. [PMID: 36362111 PMCID: PMC9653689 DOI: 10.3390/ijms232113316] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
The abuse of antibiotics leads to the increase of bacterial resistance, which seriously threatens human health. Therefore, there is an urgent need to find effective alternatives to antibiotics, and antimicrobial peptides (AMPs) are the most promising antibacterial agents and have received extensive attention. In this study, a novel potential AMP was identified from the marine invertebrate Scylla paramamosain and named Spampcin. After bioinformatics analysis and AMP database prediction, four truncated peptides (Spa31, Spa22, Spa20 and Spa14) derived from Spampcin were screened, all of which showed potent antimicrobial activity with different antibacterial spectrum. Among them, Spampcin56-86 (Spa31 for short) exhibited strong bactericidal activity against a variety of clinical pathogens and could rapidly kill the tested bacteria within minutes. Further analysis of the antibacterial mechanism revealed that Spa31 disrupted the integrity of the bacterial membrane (as confirmed by scanning electron microscopy observation, NPN, and PI staining assays), leading to bacterial rupture, leakage of cellular contents (such as elevated extracellular ATP), increased ROS production, and ultimately cell death. Furthermore, Spa31 was found to interact with LPS and effectively inhibit bacterial biofilms. The antibacterial activity of Spa31 had good thermal stability, certain ion tolerance, and no obvious cytotoxicity. It is worth noting that Spa31 could significantly improve the survival rate of zebrafish Danio rerio infected with Pseudomonas aeruginosa, indicating that Spa31 played an important role in anti-infection in vivo. This study will enrich the database of marine animal AMPs and provide theoretical reference and scientific basis for the application of marine AMPs in medical fields.
Collapse
Affiliation(s)
- Manyu Jiang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Roushi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Jingrong Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
- Correspondence: (F.C.); (K.-J.W.)
| |
Collapse
|
17
|
Hale SJM, Wagner Mackenzie B, Lux CA, Biswas K, Kim R, Douglas RG. Topical Antibiofilm Agents With Potential Utility in the Treatment of Chronic Rhinosinusitis: A Narrative Review. Front Pharmacol 2022; 13:840323. [PMID: 35770097 PMCID: PMC9234399 DOI: 10.3389/fphar.2022.840323] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
The role of bacterial biofilms in chronic and recalcitrant diseases is widely appreciated, and the treatment of biofilm infection is an increasingly important area of research. Chronic rhinosinusitis (CRS) is a complex disease associated with sinonasal dysbiosis and the presence of bacterial biofilms. While most biofilm-related diseases are associated with highly persistent but relatively less severe inflammation, the presence of biofilms in CRS is associated with greater severity of inflammation and recalcitrance despite appropriate treatment. Oral antibiotics are commonly used to treat CRS but they are often ineffective, due to poor penetration of the sinonasal mucosa and the inherently antibiotic resistant nature of bacteria in biofilms. Topical non-antibiotic antibiofilm agents may prove more effective, but few such agents are available for sinonasal application. We review compounds with antibiofilm activity that may be useful for treating biofilm-associated CRS, including halogen-based compounds, quaternary ammonium compounds and derivatives, biguanides, antimicrobial peptides, chelating agents and natural products. These include preparations that are currently available and those still in development. For each compound, antibiofilm efficacy, mechanism of action, and toxicity as it relates to sinonasal application are summarised. We highlight the antibiofilm agents that we believe hold the greatest promise for the treatment of biofilm-associated CRS in order to inform future research on the management of this difficult condition.
Collapse
Affiliation(s)
- Samuel J M Hale
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Brett Wagner Mackenzie
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christian A Lux
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Raymond Kim
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
Häring M, Amann V, Kissmann AK, Herberger T, Synatschke C, Kirsch-Pietz N, Perez-Erviti JA, Otero-Gonzalez AJ, Morales-Vicente F, Andersson J, Weil T, Stenger S, Rodríguez A, Ständker L, Rosenau F. Combination of Six Individual Derivatives of the Pom-1 Antibiofilm Peptide Doubles Their Efficacy against Invasive and Multi-Resistant Clinical Isolates of the Pathogenic Yeast Candida albicans. Pharmaceutics 2022; 14:pharmaceutics14071332. [PMID: 35890228 PMCID: PMC9319270 DOI: 10.3390/pharmaceutics14071332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/14/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
In previous studies, derivatives of the peptide Pom-1, which was originally extracted from the freshwater mollusk Pomacea poeyana, showed an exceptional ability to specifically inhibit biofilm formation of the laboratory strain ATCC 90028 as a model strain of the pathogenic yeast Candida albicans. In follow-up, here, we demonstrate that the derivatives Pom-1A to Pom-1F are also active against biofilms of invasive clinical C. albicans isolates, including strains resistant against fluconazole and/or amphotericin B. However, efficacy varied strongly between the isolates, as indicated by large deviations in the experiments. This lack of robustness could be efficiently bypassed by using mixtures of all peptides. These mixed peptide preparations were active against biofilm formation of all the isolates with uniform efficacies, and the total peptide concentration could be halved compared to the original MIC of the individual peptides (2.5 µg/mL). Moreover, mixing the individual peptides restored the antifungal effect of fluconazole against fluconazole-resistant isolates even at 50% of the standard therapeutic concentration. Without having elucidated the reason for these synergistic effects of the peptides yet, both the gain of efficacy and the considerable increase in efficiency by combining the peptides indicate that Pom-1 and its derivatives in suitable formulations may play an important role as new antibiofilm antimycotics in the fight against invasive clinical infections with (multi-) resistant C. albicans.
Collapse
Affiliation(s)
- Michelle Häring
- Institute of Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (M.H.); (V.A.)
| | - Valerie Amann
- Institute of Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (M.H.); (V.A.)
| | - Ann-Kathrin Kissmann
- Institute of Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (M.H.); (V.A.)
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany; (T.H.); (C.S.); (N.K.-P.); (T.W.)
- Correspondence: (A.-K.K.); (F.R.)
| | - Tilmann Herberger
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany; (T.H.); (C.S.); (N.K.-P.); (T.W.)
| | - Christopher Synatschke
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany; (T.H.); (C.S.); (N.K.-P.); (T.W.)
| | - Nicole Kirsch-Pietz
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany; (T.H.); (C.S.); (N.K.-P.); (T.W.)
| | - Julio A. Perez-Erviti
- Center for Protein Studies, Faculty of Biology, University of Havana, 25 Street, Havana 10400, Cuba; (J.A.P.-E.); (A.J.O.-G.)
| | - Anselmo J. Otero-Gonzalez
- Center for Protein Studies, Faculty of Biology, University of Havana, 25 Street, Havana 10400, Cuba; (J.A.P.-E.); (A.J.O.-G.)
| | - Fidel Morales-Vicente
- Synthetic Peptides Group, Center for Genetic Engineering and Biotechnology, Havana 10600, Cuba;
| | - Jakob Andersson
- AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria;
| | - Tanja Weil
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany; (T.H.); (C.S.); (N.K.-P.); (T.W.)
| | - Steffen Stenger
- Institute for Medical Microbiology and Hygiene, University Hospital Ulm, 89081 Ulm, Germany;
| | - Armando Rodríguez
- Core Facility for Functional Peptidomics, Ulm Peptide Pharmaceuticals (U-PEP), Faculty of Medicine, Ulm University, 89081 Ulm, Germany; (A.R.); (L.S.)
- Core Unit of Mass Spectrometry and Proteomics, Faculty of Medicine, Ulm University, 89081 Ulm, Germany
| | - Ludger Ständker
- Core Facility for Functional Peptidomics, Ulm Peptide Pharmaceuticals (U-PEP), Faculty of Medicine, Ulm University, 89081 Ulm, Germany; (A.R.); (L.S.)
| | - Frank Rosenau
- Institute of Pharmaceutical Biotechnology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (M.H.); (V.A.)
- Max Planck Institute for Polymer Research Mainz, Ackermannweg 10, 55128 Mainz, Germany; (T.H.); (C.S.); (N.K.-P.); (T.W.)
- Correspondence: (A.-K.K.); (F.R.)
| |
Collapse
|
19
|
Yang Y, Li Y, Mei X, Yang M, Huang H, Du F, Wu J, He Y, Sun J, Wang H, He X, Zhu S, Li Y, Liu Y. Antimicrobial Terpenes Suppressed the Infection Process of Phytophthora in Fennel-Pepper Intercropping System. FRONTIERS IN PLANT SCIENCE 2022; 13:890534. [PMID: 35755704 PMCID: PMC9218821 DOI: 10.3389/fpls.2022.890534] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
The interactions between non-host roots and pathogens may be key to the inhibition of soilborne pathogens in intercropping systems. Fennel (Foeniculum vulgare) can be intercropped with a wide range of other plants to inhibit soilborne pathogens in biodiversity cultivation. However, the key compounds of fennel root exudates involved in the interactions between fennel roots and pathogens are still unknown. Here, a greenhouse experiment confirmed that intercropping with fennel suppressed pepper (Capsicum annuum) blight disease caused by Phytophthora capsici. Experimentally, the roots and root exudates of fennel can effectively interfere with the infection process of P. capsici at rhizosphere soil concentrations by attracting zoospores and inhibiting the motility of the zoospores and germination of the cystospores. Five terpene compounds (D-limonene, estragole, anethole, gamma-terpenes, and beta-myrcene) that were identified in the fennel rhizosphere soil and root exudates were found to interfere with P. capsica infection. D-limonene was associated with positive chemotaxis with zoospores, and a mixture of the five terpene compounds showed a strong synergistic effect on the infection process of P. capsici, especially for zoospore rupture. Furthermore, the five terpene compounds can induce the accumulation of reactive oxygen species (ROS), especially anethole, in hyphae. ROS accumulation may be one of the antimicrobial mechanisms of terpene compounds. Above all, we proposed that terpene compounds secreted from fennel root play a key role in Phytophthora disease suppression in this intercropping system.
Collapse
Affiliation(s)
- Yuxin Yang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Ying Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Xinyue Mei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Min Yang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Huichuan Huang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Fei Du
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Jiaqing Wu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Yiyi He
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Junwei Sun
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Haining Wang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Xiahong He
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Shusheng Zhu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Yingbin Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Yixiang Liu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
20
|
Combination effect of epsilon-poly-L-lysine and antibiotics against common bacterial pathogens. J Antibiot (Tokyo) 2022; 75:354-359. [PMID: 35459856 DOI: 10.1038/s41429-022-00523-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 02/08/2022] [Accepted: 03/17/2022] [Indexed: 12/29/2022]
Abstract
Epsilon-poly-L-lysine (EPL) is an antimicrobial peptide with low mammalian toxicity; thus, it is commonly used as food preservative. Here, the capacity of EPL to improve the efficacy of the antibiotics ampicillin (AMP), gentamycin (GEN), tetracycline (TCN), and methicillin (MET) against four bacterial pathogens, namely Pseudomonas aeruginosa PAO1, Klebsiella pneumoniae CG43, MET-sensitive Staphylococcus aureus ATCC 25923 (MSSA), and MET-resistant S. aureus ATCC 33591 (MRSA), was investigated. Some antibiotic-EPL combinations, i.e., AMP-EPL, GEN-EPL, and TCN-EPL, were particularly active against the pathogens through synergy, partial synergy, or additive effects. Additionally, MET-EPL displayed a partial synergistic effect against MRSA. GEN-EPL had the most powerful antimicrobial effect against MSSA: it eradicated the bacterium within an hour. Conversely, AMP-EPL and MET-EPL were the least potent combinations against MRSA, and TCN-EPL was least potent against K. pneumoniae; for these combinations, bactericidal activities occurred >10 h after initial treatments. All antibiotic-EPL treatments showed inhibitory activities against P. aeruginosa biofilm formation and enhanced preformed biofilm disruption in vitro. Similarly, the inhibition of biofilm formation on a porcine skin model was observed. Moreover, no significant cytotoxicity was detected for any antibiotic-EPL treatment in tests using Balb/3t3 fibroblasts. Given the rise in antibiotic-resistant bacteria, combining antibiotics with EPL may enhance antibiotic effectiveness, as shown in this study, while helping to avoid antimicrobial resistance.
Collapse
|
21
|
Kim J, Kim JC, Ahn J. Assessment of bacteriophage-encoded endolysin as a potent antimicrobial agent against antibiotic-resistant Salmonella Typhimurium. Microb Pathog 2022; 168:105576. [PMID: 35561980 DOI: 10.1016/j.micpath.2022.105576] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 11/25/2022]
Abstract
This study was designed to evaluate the potential of using newly purified Salmonella phage-encoded endolysin LysPB32 as novel antibiotic alternative. The endolysin LysPB32 was characterized by analyzing pH and thermal stability, lytic spectrum, antimicrobial activity, and mutant frequency against Salmonella Typhimurium KCCM 40253 (STKCCM), S. Typhimurium ATCC 19585 (STATCC), S. Typhimurium CCARM 8009 (STCCARM), Klebsiella pneumoniae ATCC 23357 (KPATCC), K. pneumoniae CCARM 10237 (KPCCARM), Pseudomonas aeruginosa ATCC 27853 (PAATCC), Listeria monocytogenes ATCC 1911 (LMATCC), Staphylococcus aureus ATCC 25923 (SAATCC), and S. aureus CCARM 3080 (SACCARM). The molecular weight of LysPB32 is 17 kDa that was classified as N-acetyl-β-d-muramidase. The optimum activity of LysPB32 against the outer membrane (OM) permeabilized STKCCM, STATCC, and STCCARM was observed at 37 °C and pH 6.5. LysPB32 had a broad spectrum of muralytic activity against antibiotic-sensitive STKCCM (41 mOD/min), STATCC (32 mOD/min), and SBKACC (25 mOD/min) and antibiotic-resistant STCCARM (35 mOD/min) and KPCCARM (31 mOD/min). The minimum inhibitory concentrations (MICs) of polymyxin B against STKCCM, STCCARM, and STATCC were decreased by 4-, 4-, and 8-folds, respectively, when treated with LysPB32. The combination of LysPB32 and polymyxin B effectively inhibited the growth of STKCCM, STCCARM, and STATCC after 24 h of incubation at 37 °C, showing 4.9-, 4.4-, and 3.3-log reductions, respectively. The mutant frequency was low in STKCCM, STCCARM, and STATCC treated with combination of LysPB32-polymyxin B system. The results suggest the LysPB32-polymyxin system can be a potential candidate for alternative therapeutic agent to control antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Junhwan Kim
- Department of Biomedical Science and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jin-Chul Kim
- Department of Biomedical Science and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Juhee Ahn
- Department of Biomedical Science and Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea.
| |
Collapse
|
22
|
Cesaro A, Torres MDT, Gaglione R, Dell'Olmo E, Di Girolamo R, Bosso A, Pizzo E, Haagsman HP, Veldhuizen EJA, de la Fuente-Nunez C, Arciello A. Synthetic Antibiotic Derived from Sequences Encrypted in a Protein from Human Plasma. ACS NANO 2022; 16:1880-1895. [PMID: 35112568 DOI: 10.1021/acsnano.1c04496] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Encrypted peptides have been recently found in the human proteome and represent a potential class of antibiotics. Here we report three peptides derived from the human apolipoprotein B (residues 887-922) that exhibited potent antimicrobial activity against drug-resistant Klebsiella pneumoniae, Acinetobacter baumannii, and Staphylococci both in vitro and in an animal model. The peptides had excellent cytotoxicity profiles, targeted bacteria by depolarizing and permeabilizing their cytoplasmic membrane, inhibited biofilms, and displayed anti-inflammatory properties. Importantly, the peptides, when used in combination, potentiated the activity of conventional antibiotics against bacteria and did not select for bacterial resistance. To ensure translatability of these molecules, a protease resistant retro-inverso variant of the lead encrypted peptide was synthesized and demonstrated anti-infective activity in a preclinical mouse model. Our results provide a link between human plasma and innate immunity and point to the blood as a source of much-needed antimicrobials.
Collapse
Affiliation(s)
- Angela Cesaro
- Department of Chemical Sciences, University of Naples Federico II, Naples I-80126, Italy
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Section Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, The Netherlands
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Rosa Gaglione
- Department of Chemical Sciences, University of Naples Federico II, Naples I-80126, Italy
- Istituto Nazionale di Biostrutture e Biosistemi (INBB), Rome 00136, Italy
| | - Eliana Dell'Olmo
- Department of Chemical Sciences, University of Naples Federico II, Naples I-80126, Italy
| | - Rocco Di Girolamo
- Department of Chemical Sciences, University of Naples Federico II, Naples I-80126, Italy
| | - Andrea Bosso
- Department of Biology, University of Naples Federico II, Naples I-80126, Italy
| | - Elio Pizzo
- Department of Biology, University of Naples Federico II, Naples I-80126, Italy
| | - Henk P Haagsman
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Section Molecular Host Defence, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, The Netherlands
| | - Edwin J A Veldhuizen
- Department of Biomolecular Health Sciences, Division of Infectious Diseases and Immunology, Section Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CL, The Netherlands
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Angela Arciello
- Department of Chemical Sciences, University of Naples Federico II, Naples I-80126, Italy
- Istituto Nazionale di Biostrutture e Biosistemi (INBB), Rome 00136, Italy
| |
Collapse
|
23
|
Singh A, Amod A, Pandey P, Bose P, Pingali MS, Shivalkar S, Varadwaj P, Sahoo A, Samanta S. Bacterial biofilm infections, their resistance to antibiotics therapy and current treatment strategies. Biomed Mater 2022; 17. [PMID: 35105823 DOI: 10.1088/1748-605x/ac50f6] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/01/2022] [Indexed: 11/11/2022]
Abstract
Nearly 80% of human chronic infections are caused due to bacterial biofilm formation. This is the most leading cause for failure of medical implants resulting in high morbidity and mortality. In addition, biofilms are also known to cause serious problems in food industry. Biofilm impart enhanced antibiotic resistance and become recalcitrant to host immune responses leading to persistent and recurrent infections. It makes the clinical treatment for biofilm infections very difficult. Reduced penetration of antibiotic molecules through EPS, mutation of the target site, accumulation of antibiotic degrading enzymes, enhanced expression of efflux pump genes are the probable causes for antibiotics resistance. Accordingly, strategies like administration of topical antibiotics and combined therapy of antibiotics with antimicrobial peptides are considered for alternate options to overcome the antibiotics resistance. A number of other remediation strategies for both biofilm inhibition and dispersion of established biofilm have been developed. The metallic nanoparticles and their oxides have recently gained a tremendous thrust as antibiofilm therapy for their unique features. This present comprehensive review gives the understanding of antibiotic resistance mechanisms of biofilm and provides an overview of various currently available biofilm remediation strategies, focusing primarily on the applications of metallic nanoparticles and their oxides.
Collapse
Affiliation(s)
- Anirudh Singh
- Indian Institute of Information Technology Allahabad, Allahabad, UP, India, Allahabad, Uttar Pradesh, 211012, INDIA
| | - Ayush Amod
- Indian Institute of Information Technology Allahabad, UP, India, Allahabad, Uttar Pradesh, 211012, INDIA
| | | | - Pranay Bose
- KIIT University, Bhubaneswar, Odisha, India, Bhubaneswar, Orissa, 751024, INDIA
| | - M Shivapriya Pingali
- Indian Institute of Information Technology Allahabad, UP, India, Allahabad, Uttar Pradesh, 211012, INDIA
| | - Saurabh Shivalkar
- Applied Sciences, IIIT Allahabad, UP, India, Allahabad, 211012, INDIA
| | - Pritish Varadwaj
- Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, India, Allahabad, Uttar Pradesh, 211012, INDIA
| | - Amaresh Sahoo
- Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, UP, India, Allahabad, Uttar Pradesh, 211012, INDIA
| | - Sintu Samanta
- Applied Sciences, Indian Institute of Information Technology Allahabad, Allahabad, India, Allahabad, Uttar Pradesh, 211012, INDIA
| |
Collapse
|
24
|
Chen YC, Yang Y, Zhang C, Chen HY, Chen F, Wang KJ. A Novel Antimicrobial Peptide Sparamosin 26-54 From the Mud Crab Scylla paramamosain Showing Potent Antifungal Activity Against Cryptococcus neoformans. Front Microbiol 2021; 12:746006. [PMID: 34690992 PMCID: PMC8531530 DOI: 10.3389/fmicb.2021.746006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Due to the increasing prevalence of drug-resistant fungi and the limitations of current treatment strategies to fungal infections, exploration and development of new antifungal drugs or substituents are necessary. In the study, a novel antimicrobial peptide, named Sparamosin, was identified in the mud crab Scylla paramamosain, which contains a signal peptide of 22 amino acids and a mature peptide of 54 amino acids. The antimicrobial activity of its synthetic mature peptide and two truncated peptides (Sparamosin1-25 and Sparamosin26-54) were determined. The results showed that Sparamosin26-54 had the strongest activity against a variety of Gram-negative bacteria, Gram-positive bacteria and fungi, in particular had rapid fungicidal kinetics (killed 99% Cryptococcus neoformans within 10 min) and had potent anti-biofilm activity against C. neoformans, but had no cytotoxic effect on mammalian cells. The RNA-seq results showed that after Sparamosin26-54 treatment, the expression of genes involved in cell wall component biosynthesis, cell wall integrity signaling pathway, anti-oxidative stress, apoptosis and DNA repair were significantly up-regulated, indicating that Sparamosin26-54 might disrupt the cell wall of C. neoformans, causing oxidative stress, DNA damage and cell apoptosis. The underlying mechanism was further confirmed. Sparamosin26-54 could bind to several phospholipids in the cell membrane and effectively killed C. neoformans through disrupting the integrity of the cell wall and cell membrane observed by electron microscope and staining assay. In addition, it was found that the accumulation of reactive oxygen species (ROS) increased, the mitochondrial membrane potential (MMP) was disrupted, and DNA fragmentation was induced after Sparamosin26-54 treatment, which are all hallmarks of apoptosis. Taken together, Sparamosin26-54 has a good application prospect as an effective antimicrobial agent, especially for C. neoformans infections.
Collapse
Affiliation(s)
- Yan-Chao Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ying Yang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Chang Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Hui-Yun Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
- Fujian Innovation Research Institute for Marine Biological Antimicrobial Peptide Industrial Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
25
|
Bolosov IA, Panteleev PV, Sychev SV, Sukhanov SV, Mironov PA, Myshkin MY, Shenkarev ZO, Ovchinnikova TV. Dodecapeptide Cathelicidins of Cetartiodactyla: Structure, Mechanism of Antimicrobial Action, and Synergistic Interaction With Other Cathelicidins. Front Microbiol 2021; 12:725526. [PMID: 34484167 PMCID: PMC8415029 DOI: 10.3389/fmicb.2021.725526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/19/2021] [Indexed: 12/02/2022] Open
Abstract
In this study, dodecapeptide cathelicidins were shown to be widespread antimicrobial peptides among the Cetruminantia clade. In particular, we investigated the dodecapeptide from the domestic goat Capra hircus, designated as ChDode and its unique ortholog from the sperm whale Physeter catodon (PcDode). ChDode contains two cysteine residues, while PcDode consists of two dodecapeptide building blocks and contains four cysteine residues. The recombinant analogs of the peptides were obtained by heterologous expression in Escherichia coli cells. The structures of the peptides were studied by circular dichroism (CD), FTIR, and NMR spectroscopy. It was demonstrated that PcDode adopts a β-hairpin structure in water and resembles β-hairpin antimicrobial peptides, while ChDode forms a β-structural antiparallel covalent dimer, stabilized by two intermonomer disulfide bonds. Both peptides reveal a significant right-handed twist about 200 degrees per 8 residues. In DPC micelles ChDode forms flat β-structural tetramers by antiparallel non-covalent association of the dimers. The tetramers incorporate into the micelles in transmembrane orientation. Incorporation into the micelles and dimerization significantly diminished the amplitude of backbone motions of ChDode at the picosecond-nanosecond timescale. When interacting with negatively charged membranes containing phosphatidylethanolamine (PE) and phosphatidylglycerol (PG), the ChDode peptide adopted similar oligomeric structure and was capable to form ion-conducting pores without membrane lysis. Despite modest antibacterial activity of ChDode, a considerable synergistic effect of this peptide in combination with another goat cathelicidin – the α-helical peptide ChMAP-28 was observed. This effect is based on an increase in permeability of bacterial membranes. In turn, this mechanism can lead to an increase in the efficiency of the combined action of the synergistic pair ChMAP-28 with the Pro-rich peptide mini-ChBac7.5Nα targeting the bacterial ribosome.
Collapse
Affiliation(s)
- Ilia A Bolosov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Pavel V Panteleev
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | - Sergei V Sychev
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Stanislav V Sukhanov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Pavel A Mironov
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail Yu Myshkin
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Zakhar O Shenkarev
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | - Tatiana V Ovchinnikova
- M. M. Shemyakin and Yu. A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
26
|
Gan BH, Gaynord J, Rowe SM, Deingruber T, Spring DR. The multifaceted nature of antimicrobial peptides: current synthetic chemistry approaches and future directions. Chem Soc Rev 2021; 50:7820-7880. [PMID: 34042120 PMCID: PMC8689412 DOI: 10.1039/d0cs00729c] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 12/13/2022]
Abstract
Bacterial infections caused by 'superbugs' are increasing globally, and conventional antibiotics are becoming less effective against these bacteria, such that we risk entering a post-antibiotic era. In recent years, antimicrobial peptides (AMPs) have gained significant attention for their clinical potential as a new class of antibiotics to combat antimicrobial resistance. In this review, we discuss several facets of AMPs including their diversity, physicochemical properties, mechanisms of action, and effects of environmental factors on these features. This review outlines various chemical synthetic strategies that have been applied to develop novel AMPs, including chemical modifications of existing peptides, semi-synthesis, and computer-aided design. We will also highlight novel AMP structures, including hybrids, antimicrobial dendrimers and polypeptides, peptidomimetics, and AMP-drug conjugates and consider recent developments in their chemical synthesis.
Collapse
Affiliation(s)
- Bee Ha Gan
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Josephine Gaynord
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Sam M Rowe
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - Tomas Deingruber
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| | - David R Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
| |
Collapse
|
27
|
Shang D, Han X, Du W, Kou Z, Jiang F. Trp-Containing Antibacterial Peptides Impair Quorum Sensing and Biofilm Development in Multidrug-Resistant Pseudomonas aeruginosa and Exhibit Synergistic Effects With Antibiotics. Front Microbiol 2021; 12:611009. [PMID: 33643239 PMCID: PMC7906020 DOI: 10.3389/fmicb.2021.611009] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pseudomonas aeruginosa uses quorum sensing (QS) to control virulence, biofilm formation and antibiotic efflux pump expression. The development of effective small molecules targeting the QS system and biofilm formation represents a novel attractive strategy. In this present study, the effects of a series of Trp-containing peptides on the QS-regulated virulence and biofilm development of multidrug-resistant P. aeruginosa, as well as their synergistic antibacterial activity with three classes of traditional chemical antibiotics were investigated. The results showed that Trp-containing peptides at low concentrations reduced the production of QS-regulated virulence factors by downregulating the gene expression of both the las and rhl systems in the strain MRPA0108. Biofilm formation was inhibited in a concentration-dependent manner, which was associated with extracellular polysaccharide production inhibition by downregulating pelA, algD, and pslA transcription. These changes correlated with alterations in the extracellular production of pseudomonal virulence factors and swarming motility. In addition, the combination of Trp-containing peptides at low concentration with the antibiotics ceftazidime and piperacillin provided synergistic effects. Notably, L11W and L12W showed the highest synergy with ceftazidime and piperacillin. A mechanistic study demonstrated that the Trp-containing peptides, especially L12W, significantly decreased β-lactamase activity and expression of efflux pump genes OprM, MexX, and MexA, resulting in a reduction in antibiotic efflux from MRPA0108 cells and thus increasing the antibacterial activity of these antibiotics against MRPA0108.
Collapse
Affiliation(s)
- Dejing Shang
- School of Life Sciences, Liaoning Normal University, Dalian, China
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Xue Han
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Wanying Du
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Zhiru Kou
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Fengquan Jiang
- Clinical Laboratory Department of the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
28
|
Patel N, Swavey S, Robinson J. A Cationic Porphyrin, ZnPor, Disassembles Pseudomonas aeruginosa Biofilm Matrix, Kills Cells Directly, and Enhances Antibiotic Activity of Tobramycin. Antibiotics (Basel) 2020; 9:E875. [PMID: 33291344 PMCID: PMC7762324 DOI: 10.3390/antibiotics9120875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/28/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
One of the greatest threats to human health is the rise in antibiotic-resistant bacterial infections. Pseudomonas aeruginosa (PsA) is an "opportunistic" pathogen known to cause life-threatening infections in immunocompromised individuals and is the most common pathogen in adults with cystic fibrosis (CF). We report here a cationic zinc (II) porphyrin, ZnPor, that effectively kills planktonic and biofilm-associated cells of PsA. In standard tests against 16-18 h-old biofilms, concentrations as low as 16 µg/mL resulted in the extensive disruption and detachment of the matrix. The pre-treatment of biofilms for 30 min with ZnPor at minimum inhibitory concentration (MIC) levels (4 µg/mL) substantially enhanced the ability of tobramycin (Tobra) to kill biofilm-associated cells. We demonstrate the rapid uptake and accumulation of ZnPor in planktonic cells even in dedicated heme-uptake system mutants (ΔPhu, ΔHas, and the double mutant). Furthermore, uptake was unaffected by the ionophore carbonyl cyanide m-chlorophenyl hydrazine (CCCP). Cells pre-exposed to ZnPor took up the cell-impermeant dye SYTOXTM Green in a concentration-dependent manner. The accumulation of ZnPor did not result in cell lysis, nor did the cells develop resistance. Taken together, these properties make ZnPor a promising candidate for treating multi-drug-resistant infections, including persistent, antibiotic-resistant biofilms.
Collapse
Affiliation(s)
- Neha Patel
- Department of Biology, University of Dayton, Dayton, OH 45469, USA;
| | - Shawn Swavey
- Department of Chemistry, University of Dayton, Dayton, OH 45469, USA;
| | - Jayne Robinson
- Department of Biology, University of Dayton, Dayton, OH 45469, USA;
- Integrated Science and Engineering Center, University of Dayton, Dayton, OH 45469, USA
| |
Collapse
|
29
|
Lei W, Krolla P, Schwartz T, Levkin PA. Controlling Geometry and Flow Through Bacterial Bridges on Patterned Lubricant-Infused Surfaces (pLIS). SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004575. [PMID: 33216442 DOI: 10.1002/smll.202004575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/11/2020] [Indexed: 06/11/2023]
Abstract
Spatial control of bacteria and biofilms on surfaces is necessary to understand the biofilm formation and the social interactions between bacterial communities, which could provide useful hints to study the biofilm-involved diseases. Here patterned lubricant-infused surfaces (pLIS) are utilized to fabricate connective structures named "bacterial bridges" between bacterial colonies of Pseudomonas aeruginosa by a simple dewetting method. It is demonstrated that the bacteria attached to hydrophilic areas and bacteria precipitated on lubricant infused borders both contribute to the formation of bacterial bridges. The geometry and distribution of bridges can be controlled using predesigned superhydrophobic-hydrophilic patterns. It is demonstrated that bacterial bridges connecting bacteria colonies act as bio-microfluidic channels and can transport liquids, nutrients, and antibacterial substances between neighboring bacteria clusters. Thus, bacterial bridges can be used to study formation, spreading, and development of bacterial colonies, and communication within and between isolated biofilms.
Collapse
Affiliation(s)
- Wenxi Lei
- Institute of Biological and Chemical Systems - Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Peter Krolla
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Thomas Schwartz
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Pavel A Levkin
- Institute of Biological and Chemical Systems - Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
- Institute of Organic Chemistry, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| |
Collapse
|
30
|
Lei W, Demir K, Overhage J, Grunze M, Schwartz T, Levkin PA. Droplet-Microarray: Miniaturized Platform for High-Throughput Screening of Antimicrobial Compounds. ACTA ACUST UNITED AC 2020; 4:e2000073. [PMID: 32875737 DOI: 10.1002/adbi.202000073] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/20/2020] [Indexed: 01/12/2023]
Abstract
Currently, there are no time-saving and cost-effective high-throughput screening methods for the evaluation of bacterial drug-resistance. In this study, a droplet microarray (DMA) system is established as a miniaturized platform for high-throughput screening of antibacterial compounds using the emerging, opportunistic human pathogen Pseudomonas aeruginosa (P. aeruginosa) as a target. Based on the differences in wettability of DMA slides, a rapid method for generating microarrays of nanoliter-sized droplets containing bacteria is developed. The bacterial growth in droplets is evaluated using fluorescence. The new method enables immediate screening with libraries of antibiotics. A novel simple colorimetric readout method compatible with the nanoliter size of the droplets is established. Furthermore, the drug-resistance of P. aeruginosa 49, a multi-resistant strain from an environmental isolate, is investigated. This study demonstrates the potential of the DMA platform for the rapid formation of microarrays of bacteria for high-throughput drug screening.
Collapse
Affiliation(s)
- Wenxi Lei
- Institute of Biological and Chemical Systems-Functional Molecular Systems, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Konstantin Demir
- Institute of Biological and Chemical Systems-Functional Molecular Systems, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Joerg Overhage
- Department of Health Sciences, Carleton University, Ottawa, ON, K1S 5B6, Canada
| | - Michael Grunze
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Thomas Schwartz
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Pavel A Levkin
- Institute of Biological and Chemical Systems-Functional Molecular Systems, Karlsruhe Institute of Technology, Hermann-von-Helmholtz Platz 1, Eggenstein-Leopoldshafen, 76344, Germany.,Institute of Organic Chemistry, Karlsruhe Institute of Technology, Karlsruhe, 76131, Germany
| |
Collapse
|
31
|
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: 28] [Impact Index Per Article: 7.0] [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
|
32
|
Zhang H, Yang Y, Mei X, Li Y, Wu J, Li Y, Wang H, Huang H, Yang M, He X, Zhu S, Liu Y. Phenolic Acids Released in Maize Rhizosphere During Maize-Soybean Intercropping Inhibit Phytophthora Blight of Soybean. FRONTIERS IN PLANT SCIENCE 2020; 11:886. [PMID: 32849668 PMCID: PMC7399372 DOI: 10.3389/fpls.2020.00886] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/29/2020] [Indexed: 05/20/2023]
Abstract
Interspecies interactions play a key role in soil-borne disease suppression in intercropping systems. However, there are limited data on the underlying mechanisms of soil-borne Phytophthora disease suppression. Here, a field experiment confirmed the effects of maize and soybean intercropping on Phytophthora blight of soybean caused by Phytophthora sojae. Experimentally, the roots and root exudates of maize were found to attract P. sojae zoospores and inhibit their motility and the germination of cystospores. Furthermore, five phenolic acids (p-coumaric acid, cinnamic acid, p-hydroxybenzoic acid, vanillic acid, and ferulic acid) that were consistently identified in the root exudates and rhizosphere soil of maize were found to interfere with the infection behavior of P. sojae. Among them, cinnamic acid was associated with significant chemotaxis in zoospores, and p-coumaric acid and cinnamic acid showed strong antimicrobial activity against P. sojae. However, in the rhizosphere soil of soybean, only p-hydroxybenzoic acid, low concentrations of vanillic acid, and ferulic acid were identified. Importantly, the coexistence of five phenolic acids in the maize rhizosphere compared with three phenolic acids in the soybean rhizosphere showed strong synergistic antimicrobial activity against the infection behavior of P. sojae. In summary, the types and concentrations of phenolic acids in maize and soybean rhizosphere soils were found to be crucial factors for Phytophthora disease suppression in this intercropping system.
Collapse
Affiliation(s)
- He Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Yuxin Yang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Xinyue Mei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Ying Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Jiaqing Wu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Yiwen Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Huiling Wang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
| | - Huichuan Huang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Min Yang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Xiahong He
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Shusheng Zhu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| | - Yixiang Liu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, College of Plant Protection, Yunnan Agricultural University, Kunming, China
- China France Plantomix Joint Laboratory, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
33
|
Yang Y, Chen F, Chen HY, Peng H, Hao H, Wang KJ. A Novel Antimicrobial Peptide Scyreprocin From Mud Crab Scylla paramamosain Showing Potent Antifungal and Anti-biofilm Activity. Front Microbiol 2020; 11:1589. [PMID: 32849331 PMCID: PMC7396596 DOI: 10.3389/fmicb.2020.01589] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022] Open
Abstract
Natural antimicrobial peptides (AMPs) are potential antibiotic alternatives. Marine crustaceans are thought to generate more powerful and various AMPs to protect themselves from infections caused by pathogenic microorganisms in their complex aquatic habitat, thus becoming one of the most promising sources of AMPs or other bioactive substances. In the study, a novel protein was identified as an interacting partner of male-specific AMP SCY2 in Scylla paramamosain and named scyreprocin. The recombinant product of scyreprocin (rScyreprocin) was successfully expressed in Escherichia coli. rScyreprocin exerted potent, broad-spectrum antifungal, antibacterial, and anti-biofilm activity (minimum inhibitory concentrations from 0.5 to 32 μM) through differential modes of action, including disruption of cell membrane integrity and induction of cell apoptosis, and has rapid bactericidal (in 0.5–2 h) and fungicidal (in 8–10 h) kinetics. In addition to its fungicidal activity against planktonic fungi, rScyreprocin also prevented the adhesion of fungal cells, inhibited biofilm formation, and eradicated the mature biofilms. Moreover, rScyreprocin showed a profound inhibitory effect on spore germination of Aspergillus spp. (minimum inhibitory concentrations from 4 to 8 μM). This peptide was not cytotoxic to murine and mammalian cells and could increase the survival rate of Oryzias melastigma under the challenge of Vibrio harveyi. Taken together, the novel AMP scyreprocin would be a promising alternative to antibiotics used in aquaculture and medicine.
Collapse
Affiliation(s)
- Ying Yang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Hui-Yun Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Hui Peng
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Hua Hao
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean and Earth Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
34
|
Lin S, Li H, Tao Y, Liu J, Yuan W, Chen Y, Liu Y, Liu S. In Vitro and in Vivo Evaluation of Membrane-Active Flavone Amphiphiles: Semisynthetic Kaempferol-Derived Antimicrobials against Drug-Resistant Gram-Positive Bacteria. J Med Chem 2020; 63:5797-5815. [DOI: 10.1021/acs.jmedchem.0c00053] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Shuimu Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Hongxia Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yiwen Tao
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Jiayong Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Wenchang Yuan
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yongzhi Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Ying Liu
- Guangdong Key Laboratory of Optical Fiber Sensing and Communications, Institute of Photonics Technology, Jinan University, Guangzhou, Guangdong 510632, P. R. China
| | - Shouping Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
35
|
Isayenko OY, Knysh OV, Kotsar OV, Ryzhkova TN, Dyukareva GI. Simultaneous and sequential influence of metabolite complexes of Lactobacillus rhamnosus and Saccharomyces boulardii and antibiotics against poly-resistant Gram-negative bacteria. REGULATORY MECHANISMS IN BIOSYSTEMS 2020. [DOI: 10.15421/022021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
For the first time the poly-resistant strains of Gram-negative microorganisms were studied for the sensitivity to combined simultaneous and sequential influence of metabolic complexes of Lactobacillus rhamnosus GG and Saccharomyces boulardii, obtained by the author’s method without using the growth media, with antibiotics. The synergic activity of antibacterial preparations and metabolic complexes of L. rhamnosus GG and S. boulardii were studied using modified disk-diffusive method of Kirby-Bauer. During the sequential method of testing (at first the microorganisms were incubated with structural components and metabolites, then their sensitivity to the antibacterial preparations was determined), we observed increase in the diameters of the zones of growth inhibition of Pseudomonas aeruginosa PR to the typical antibiotics (gentamicin, amіcyl, ciprofloxacin, сefotaxime) and non-typical (lincomycin, levomycetin) depending on the tested combinations. Acinetobacter baumannii PR exhibited lower susceptibility: growth inhibition was seen for the combination with ciprofloxacin, сefotaxime, levomycetin. Susceptibility of Lelliottia amnigena (Enterobacter amnigenus) PR increased to levofloxacin, lincomycin. The zones of growth inhibition of Klebsiella pneumoniae PR increased to gentamicin, amіcyl, tetracycline, сeftriaxone. Maximum efficiency was determined during sequential combination of antibiotics with separate metabolic complexes of L. rhamnosus and S. boulardii, and also their combination (to 15.2, 20.2 and 15.4 mm respectively) compared with their simultaneous use (to 12.2, 15.2 and 13.0 mm respectively) for all the tested poly-resistant pathogens, regardless of the mechanism of action of antibacterial preparation. Metabolic complexes of L. rhamnosus GG and S. boulardii, due to increase in the susceptibility of microorganisms, can decrease the therapeutic concentration of antibiotic, slow the probability of the development of resistance of microorganisms, and are therefore promising candidates for developing “accompanying medications” to antibiotics and antimicrobial preparations of new generation.
Collapse
|
36
|
Duperthuy M. Antimicrobial Peptides: Virulence and Resistance Modulation in Gram-Negative Bacteria. Microorganisms 2020; 8:microorganisms8020280. [PMID: 32092866 PMCID: PMC7074834 DOI: 10.3390/microorganisms8020280] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 02/03/2023] Open
Abstract
Growing resistance to antibiotics is one of the biggest threats to human health. One of the possibilities to overcome this resistance is to use and develop alternative molecules such as antimicrobial peptides (AMPs). However, an increasing number of studies have shown that bacterial resistance to AMPs does exist. Since AMPs are immunity molecules, it is important to ensure that their potential therapeutic use is not harmful in the long term. Recently, several studies have focused on the adaptation of Gram-negative bacteria to subinhibitory concentrations of AMPs. Such concentrations are commonly found in vivo and in the environment. It is therefore necessary to understand how bacteria detect and respond to low concentrations of AMPs. This review focuses on recent findings regarding the impact of subinhibitory concentrations of AMPs on the modulation of virulence and resistance in Gram-negative bacteria.
Collapse
Affiliation(s)
- Marylise Duperthuy
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Succ. Centre-ville, Montréal, QC H3C 3J7, Canada
| |
Collapse
|
37
|
Majid A, Naz F, Khaskheli MH. Structural Plasticity of EAK-16 Peptide Inducing Vesicle Membrane Leakage. Protein Pept Lett 2020; 27:801-807. [PMID: 32003653 DOI: 10.2174/0929866527666200129141116] [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: 08/23/2019] [Revised: 11/11/2019] [Accepted: 11/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ionic complementary peptide EAK-16 has been studies for anticancer drug delivery application. This is a 16 residues, short sequence peptide has ability to trosnform into micro/nanoparticle via self-assembly. However, it is still not clear that how this can bind with cell membrane to induce membrane leakage or delivering their cargo inside cell membrane. OBJECTIVE The main objective of this work was to understand behaviour of secondary structure conformation of peptide in solution and at lipid membrane interfaces and membrane permeability of synthetic ionic complementary peptide EAK-16. The corresponding secondary structure conformation was evaluated. METHODS We performed biophysical investigation to probe the interaction of synthesised ionic complementary peptide (EAK-16) with dimyristoylphospholcholine (DMPC) and dimyristoylphosphoserine (DMPS) membrane interfaces. The folding behaviours of EAK-16 were studied with Circular Dichroism (CD) spectroscopy. Membrane leakage with peptide was confirmed with calcein leakage assay. RESULTS Our finding of this study showed that in aqueous phase EAK-16 was predominantly folded into β-sheets. The temperature could alter the β-sheets. However, in DMPC and DMPS membrane interfaces, EAK-16 adopted helical conformation. EAK-16 has preference in perturbing anionic compared Zwitterionic lipid vesicles. This study proposed that hydrophobic grooves of EAK-16 might be a key in the association with lipid bilayers. Secondly, a charge distribution of ionic residues would also support the orientation at lipid bilayers. This peptide membrane association would facilitate the membrane destabilisation. CONCLUSION This study demonstrated the supporting evidence that EAK-16 could interact with lipid membranes and conforming to helical structure, while the helical conformation induced the lipid membrane leakage. Overall, this study provides a physical rationale that ionic complementary peptide can be a useful tool for designing and development of novel antibiotics and anticancer agents along its previous drug delivery applications.
Collapse
Affiliation(s)
- Abdul Majid
- Department of Biochemistry, Shah Abdul Latif University, Khairpur, Pakistan
| | - Farah Naz
- Department of Biochemistry, Shah Abdul Latif University, Khairpur, Pakistan
| | | |
Collapse
|
38
|
Tyagi N, Kumar A. Understanding effect of interaction of nanoparticles and antibiotics on bacteria survival under aquatic conditions: Knowns and unknowns. ENVIRONMENTAL RESEARCH 2020; 181:108945. [PMID: 31806288 DOI: 10.1016/j.envres.2019.108945] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/22/2019] [Accepted: 11/17/2019] [Indexed: 06/10/2023]
Abstract
The review provides a comprehensive overview of the available state-of-the-art of nanoparticles (NPs) and antibiotics (ABs) occurrence and their fate in the natural aquatic settings by addressing different research questions and the challenges faced while addressing those questions. Firstly, understand the interaction of NPs and ABs with themselves in addition to other matrix components (presence of natural organic matter, bacteria, biofilms, other anthropogenic pollutants and metals from natural sources). Secondly, summarize the bactericidal activity of NP and AB due to reactive oxygen species (ROS) production. The complete information was gathered from database and analysed as per the conjectured questions under laboratory versus environmental-relevant conditions (1. Fate of NPs and ABs, and 2. Will the presence of NPs and ABs alone and their mixtures influence the ROS concentration and antibacterial activity), and proposed six reactions to describe the fate of NP and AB in natural aquatic settings. However, laboratory-based studies revealed that NP and AB fate largely depend on the ionic strength, organic matter content and pH of the matrix whereas field based information is missing about this. The former was performed at sterile conditions using sophisticated instruments and standard protocol as compared to latter and can't be replicated under natural aquatic settings due to lack of: (i) accurate environmental concentration of NPs and ABs, (ii) knowledge of bacterial type and their concentration, (iii) optimized protocol and tracking systems. The author's recommendation is to verify the proposed reactions experimentally by using the frequently found pairs of NPs and ABs in the natural aquatic settings. Further, ranked them on their decreasing order of toxicity and informed regulatory bodies for further action. Overall research is needed in the suggested directions to reduce uncertainty behind the impacts of NPs and ABs on the aquatic settings and their role in bactericidal activity.
Collapse
Affiliation(s)
- Neha Tyagi
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| | - Arun Kumar
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| |
Collapse
|
39
|
Machineni L. Effects of biotic and abiotic factors on biofilm growth dynamics and their heterogeneous response to antibiotic challenge. J Biosci 2020. [DOI: 10.1007/s12038-020-9990-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
40
|
Machineni L. Effects of biotic and abiotic factors on biofilm growth dynamics and their heterogeneous response to antibiotic challenge. J Biosci 2020; 45:25. [PMID: 32020907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Over the last couple of decades, with the crisis of new antimicrobial arsenal, multidrug-resistant clinical pathogens have been observed extensively. In clinical and medical settings, these persistent pathogens predominantly grow as complex heterogeneous structures enmeshed in a self-produced exopolysaccharide matrix, termed as biofilms. Since biofilms can rapidly form by adapting new environmental surroundings and have potential effect on human health, it is critical to study them promptly and consistently. Biofilm infections are challenging in the contamination of medical devices and implantations, food processing and pharmaceutical industrial settings, and in dental area caries, periodontitis and so on. The persistence of infections associated with biofilms has been mainly attributed to the increased antibiotic resistance offered by the cells growing in biofilms. In fact, it is well known that this recalcitrance of bacterial biofilms is multifactorial, and there are several resistance mechanisms that may act in parallel in order to provide an enhanced level of resistance to the biofilm. In combination, distinct resistance mechanisms significantly decrease our ability to control and eradicate biofilm-associated infections with current antimicrobial arsenal. In addition, various factors are known to influence the process of biofilm formation, growth dynamics, and their heterogeneous response towards antibiotic therapy. The current review discusses the contribution of cellular and physiochemical factors on the growth dynamics of biofilm, especially their role in antibiotic resistance mechanisms of bacterial population living in surface attached growth mode. A systematic investigation on the effects and treatment of biofilms may pave the way for novel therapeutic strategies to prevent and treat biofilms in healthcare and industrial settings.
Collapse
|
41
|
Berditsch M, Afonin S, Reuster J, Lux H, Schkolin K, Babii O, Radchenko DS, Abdullah I, William N, Middel V, Strähle U, Nelson A, Valko K, Ulrich AS. Supreme activity of gramicidin S against resistant, persistent and biofilm cells of staphylococci and enterococci. Sci Rep 2019; 9:17938. [PMID: 31784584 PMCID: PMC6884456 DOI: 10.1038/s41598-019-54212-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/06/2019] [Indexed: 01/03/2023] Open
Abstract
Three promising antibacterial peptides were studied with regard to their ability to inhibit the growth and kill the cells of clinical strains of Staphylococcus aureus, Enterococcus faecalis and Enterococcus faecium. The multifunctional gramicidin S (GS) was the most potent, compared to the membranotropic temporin L (TL), being more effective than the innate-defence regulator IDR-1018 (IDR). These activities, compared across 16 strains as minimal bactericidal and minimal inhibitory concentrations (MIC), are independent of bacterial resistance pattern, phenotype variations and/or biofilm-forming potency. For S. aureus strains, complete killing is accomplished by all peptides at 5 × MIC. For E. faecalis strains, only GS exhibits a rapid bactericidal effect at 5 × MIC, while TL and IDR require higher concentrations. The biofilm-preventing activities of all peptides against the six strains with the largest biofilm biomass were compared. GS demonstrates the lowest minimal biofilm inhibiting concentrations, whereas TL and IDR are consistently less effective. In mature biofilms, only GS completely kills the cells of all studied strains. We compare the physicochemical properties, membranolytic activities, model pharmacokinetics and eukaryotic toxicities of the peptides and explain the bactericidal, antipersister and antibiofilm activities of GS by its elevated stability, pronounced cell-penetration ability and effective utilization of multiple modes of antibacterial action.
Collapse
Affiliation(s)
- Marina Berditsch
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Sergii Afonin
- KIT, Institute of Biological Interfaces (IBG-2), Karlsruhe, 76021, Germany
| | - Jennifer Reuster
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Hannah Lux
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Kristina Schkolin
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany
| | - Oleg Babii
- KIT, Institute of Biological Interfaces (IBG-2), Karlsruhe, 76021, Germany
| | - Dmytro S Radchenko
- Enamine Ltd., Kyiv, 02094, Ukraine.,Taras Shevchenko National University of Kyiv, Kyiv, 01601, Ukraine
| | - Issah Abdullah
- University College London (UCL), UCL School of Pharmacy, London, WC1N 1AX, United Kingdom
| | - Nicola William
- University of Leeds, School of Chemistry, Leeds, LS9 2JT, United Kingdom
| | - Volker Middel
- KIT, Institute of Toxicology and Genetics (ITG), Eggenstein-Leopoldshafen, 76344, Germany
| | - Uwe Strähle
- KIT, Institute of Toxicology and Genetics (ITG), Eggenstein-Leopoldshafen, 76344, Germany
| | - Andrew Nelson
- University of Leeds, School of Chemistry, Leeds, LS9 2JT, United Kingdom
| | - Klara Valko
- University College London (UCL), UCL School of Pharmacy, London, WC1N 1AX, United Kingdom
| | - Anne S Ulrich
- Karlsruhe Institute of Technology (KIT), Institute of Organic Chemistry (IOC), Karlsruhe, 76131, Germany. .,KIT, Institute of Biological Interfaces (IBG-2), Karlsruhe, 76021, Germany.
| |
Collapse
|
42
|
Shang D, Liu Y, Jiang F, Ji F, Wang H, Han X. Synergistic Antibacterial Activity of Designed Trp-Containing Antibacterial Peptides in Combination With Antibiotics Against Multidrug-Resistant Staphylococcus epidermidis. Front Microbiol 2019; 10:2719. [PMID: 31824473 PMCID: PMC6886405 DOI: 10.3389/fmicb.2019.02719] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/08/2019] [Indexed: 01/10/2023] Open
Abstract
Multidrug resistance among various bacterial strains is leading to worldwide resistance to a wide range of antibiotics. Combination therapy involving current antibiotics and other biological or chemical molecules represents an attractive novel strategy. In this study, we investigated the synergistic antibacterial activity of a series of Trp-containing antimicrobial peptides (AMPs) with four classes of traditional chemical antibiotics that are inactive against multidrug-resistant Staphylococcus epidermidis (MRSE) in vitro and in vivo. Among the antibiotics that we studied, penicillin, ampicillin and erythromycin showed a distinct synergistic effect in combination with all of the Trp-containing AMPs, represented by a fractional inhibitory concentration index (FICI) of <0.5. The antibacterial activities were noticeably improved, with 32-to 64-fold reductions in the MIC values for ampicillin and 16- to 32-fold reductions in the MIC values for erythromycin and penicillin. Tetracycline showed synergistic activity with only I1WL5W but additive activity with L11W, L12W, and I4WL5W. Ceftazidime exhibited additive activity with the Trp-containing peptides. In addition, the antibiotics in combination with the peptide significantly inhibited biofilm formation by MRSE 1208. A mechanistic study demonstrated that the Trp-containing peptides, especially I1WL5W and I4WL5W, which contain two tryptophan residues, disrupted bacterial inner and outer membranes, which promoted antibiotic delivery into the cytoplasm and access to cytoplasmic targets; however, L11W and L12W may have increased intracellular antibiotic concentrations by decreasing blaZ, tet(m) and msrA expression. Importantly, strong synergistic activity against the MRSE 1208 strain was observed for the combination of I1WL5W and penicillin in a mouse infection model. Thus, the combination of AMPs and traditional antibiotics could be a promising option for the prevention of acute and chronic infections caused by MRSE.
Collapse
Affiliation(s)
- Dejing Shang
- School of Life Sciences, Liaoning Normal University, Dalian, China.,Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Yue Liu
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Fengquan Jiang
- School of Life Sciences, Liaoning Normal University, Dalian, China.,Clinical Laboratory Department of the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Fangyu Ji
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - He Wang
- School of Life Sciences, Liaoning Normal University, Dalian, China
| | - Xue Han
- School of Life Sciences, Liaoning Normal University, Dalian, China
| |
Collapse
|
43
|
Isayenko OY. Synergistic activity of filtrates of Lactobacillus rhamnosus and Saccharomyces boulardii and antibacterial preparations against Corynebacterium spp. REGULATORY MECHANISMS IN BIOSYSTEMS 2019. [DOI: 10.15421/021966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We present the results of the first study of the combined influence of the biologically active substances Lactobacillus rhamnosus GG ATCC 53103 and Saccharomyces boulardii, obtained by the author’s method, and antibacterial agents on Corynebacterium spp. The first area of research was the study of increasing the sensitivity of toxigenic microorganisms to antimicrobial drugs due to the consecutive effects of the structural components and metabolites of L. rhamnosus GG and S. boulardii and antibacterial drugs on Corynebacterium spp. tox+. The greatest increase in the sensitivity of test-cultures of corynebacteria to penicillin (by 19.4 mm), imipenem (by 15.0 mm), vancomycin (by 12.0 mm), gentamicin (by 11.0 mm), ciprofloxacin (by 9.8 mm), erythromycin (by 9.6 mm), cefotaxime (by 9.5 mm) occurred due to the products of lactobacteria and a combination of metabolites of lactobacteria and saccharomycetes. The second area of research was the study of the synergic activity of substances L. rhamnosus GG and S. boulardii and traditional antibacterial drugs manifested by their simultaneous effect on Corynebacterium spp. Maximum potentiation of azithromycin (by 4.6 mm), erythromycin (by 4.5 mm), cefotaxime (by 2.2 mm), ceftriaxone (by 1.6 mm) and ampicillin (by 1.0 mm) relative to corynebacteria was also observed under the influence of lactobacteria metabolites and a combination of lactobacteria and saccharomycetes metabolites. Different degrees of manifestation of the combined action of biologically active substances L. rhamnosus GG and S. boulardii with antibiotics were determined, which depended on the selected combinations, the method of influence on the microorganism, the individual sensitivity of the test-cultures, the activity of the test filtrates and the initial concentration of the producers used to obtain the products of vital activity of lactobacteria and saccharomyces. The presented complexes of structural components and metabolites of L. rhamnosus GG and S. boulardii, obtained without the use of traditional nutrient media, by increasing the bioavailability of pathogenic pathogens can reduce the required concentration of the antibiotic, continuing their use, and suspend the likelihood of pathogens developing resistance to microorganisms. This makes them promising candidates both for the development of "accompaniment-preparations" for antibiotics for the additional therapy of infectious diseases of different etiology, and for the creation of a new direction of antimicrobial agents with multifunctional capabilities. Synergistic activity of filtrates L. rhamnosus GG and S. boulardii and antibacterial preparations against Corynebacterium spp.
Collapse
|
44
|
Rončević T, Puizina J, Tossi A. Antimicrobial Peptides as Anti-Infective Agents in Pre-Post-Antibiotic Era? Int J Mol Sci 2019; 20:E5713. [PMID: 31739573 PMCID: PMC6887943 DOI: 10.3390/ijms20225713] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Resistance to antibiotics is one of the main current threats to human health and every year multi-drug resistant bacteria are infecting millions of people worldwide, with many dying as a result. Ever since their discovery, some 40 years ago, the antimicrobial peptides (AMPs) of innate defense have been hailed as a potential alternative to conventional antibiotics due to their relatively low potential to elicit resistance. Despite continued effort by both academia and start-ups, currently there are still no antibiotics based on AMPs in use. In this study, we discuss what we know and what we do not know about these agents, and what we need to know to successfully translate discovery to application. Understanding the complex mechanics of action of these peptides is the main prerequisite for identifying and/or designing or redesigning novel molecules with potent biological activity. However, other aspects also need to be well elucidated, i.e., the (bio)synthetic processes, physiological and pathological contexts of their activity, and a quantitative understanding of how physico-chemical properties affect activity. Research groups worldwide are using biological, biophysical, and algorithmic techniques to develop models aimed at designing molecules with the necessary blend of antimicrobial potency and low toxicity. Shedding light on some open questions may contribute toward improving this process.
Collapse
Affiliation(s)
- Tomislav Rončević
- Department of Biology, Faculty of Science, University of Split, 21000 Split, Croatia;
- Laboratory for Aquaculture, Institute of Oceanography and Fisheries, 21000 Split, Croatia
| | - Jasna Puizina
- Department of Biology, Faculty of Science, University of Split, 21000 Split, Croatia;
| | - Alessandro Tossi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| |
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
|
Giacomucci S, Cros CDN, Perron X, Mathieu-Denoncourt A, Duperthuy M. Flagella-dependent inhibition of biofilm formation by sub-inhibitory concentration of polymyxin B in Vibrio cholerae. PLoS One 2019; 14:e0221431. [PMID: 31430343 PMCID: PMC6701800 DOI: 10.1371/journal.pone.0221431] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/06/2019] [Indexed: 01/02/2023] Open
Abstract
Biofilm formation is a common strategy used by bacteria in order to survive and persist in the environment. In Vibrio cholerae (V. cholerae), a Gram-negative pathogen responsible for the cholera disease, biofilm-like aggregates are important for the pathogenesis and disease transmission. Biofilm formation is initiated by the attachment of the bacteria to a surface, followed by maturation stages involving the formation of a biofilm matrix. In V. cholerae, flagella are essential for the initial step of biofilm formation, allowing the bacteria to swim and to detect a surface. In this study, we explored the effect of polymyxin B (PmB), a cationic bacterial antimicrobial peptide, on biofilm formation in pathogenic V. cholerae strains belonging to the O1 and O139 serotypes. We found that sub-inhibitory concentration of PmB induces a reduction of the biofilm formation by V. cholerae O1 and O139. Experiment on preformed biofilm demonstrated that the biofilm formation inhibition occurs at the initial step of biofilm formation, where the flagella are essential. We further characterize the effect of PmB on V. cholerae flagellation. Our results demonstrate that the flagellin expression is not reduced in presence of sub-inhibitory concentration of PmB. However, a decrease of the abundance of flagellin associated with the bacterial cells together with an increase in the secretome was observed. Electron microscopy observations also suggest that the abundance of aflagellated bacteria increases upon PmB supplementation. Finally, in agreement with the effect on the flagellation, a reduction of the bacterial motility is observed. Altogether, our results suggest that the PmB affect V. cholerae flagella resulting in a decrease of the motility and a compromised ability to form biofilm.
Collapse
Affiliation(s)
- Sean Giacomucci
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Succ. Centre-ville, Montréal, Québec, Canada
| | - Candice Danabé-Nieto Cros
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Succ. Centre-ville, Montréal, Québec, Canada
| | - Xavier Perron
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Succ. Centre-ville, Montréal, Québec, Canada
| | - Annabelle Mathieu-Denoncourt
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Succ. Centre-ville, Montréal, Québec, Canada
| | - Marylise Duperthuy
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Succ. Centre-ville, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
47
|
Development of a nebramine-cyclam conjugate as an antibacterial adjuvant to potentiate β-lactam antibiotics against multidrug-resistant P. aeruginosa. J Antibiot (Tokyo) 2019; 72:816-826. [PMID: 31420586 DOI: 10.1038/s41429-019-0221-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 12/21/2022]
Abstract
The β-lactams are the most widely used class of antibiotics due to their safety, effectiveness, and spectrum of activity. As a result of their ubiquitous usage, there has been a steady rise in β-lactam resistant Gram-negative bacteria, especially Pseudomonas aeruginosa, resulting in limited treatment options. P. aeruginosa can develop multidrug-resistant phenotypes using a multifaceted approach of β-lactamase expression, decreased porin production and increased efflux. Current β-lactamase inhibitors address drug hydrolyzing enzymes but may not be as effective in phenotypes with reduced permeability and/or overexpressed efflux pumps. Herein, we present the synthesis and biological evaluation of a nebramine-cyclam conjugate molecule that is able to potentiate β-lactam antibiotics, as well as other legacy antibiotics, against P. aeruginosa in vitro. Combination studies show that this adjuvant is able to synergize with β-lactams such as aztreonam and ceftazidime against multidrug-resistant and extremely drug-resistant clinical isolates through a hypothesized mechanism of outer membrane permeabilization. Importantly, the addition of low concentrations (8 µM) of the nontoxic nebramine-cyclam conjugate is able to further potentiate existing β-lactam/β-lactamase inhibitor combinations in β-lactamase-harboring P. aeruginosa strains. These data support a potential application of the nebramine-cyclam conjugate as an adjuvant for treating infections caused by P. aeruginosa strains that utilize multiple mechanisms of resistance.
Collapse
|
48
|
The Microbial Lipopeptide Paenibacterin Disrupts Desiccation Resistance in Salmonella enterica Serovars Tennessee and Eimsbuettel. Appl Environ Microbiol 2019; 85:AEM.00739-19. [PMID: 31101609 DOI: 10.1128/aem.00739-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/07/2019] [Indexed: 01/20/2023] Open
Abstract
Salmonella enterica is increasingly linked to disease outbreaks associated with consumption of low-water-activity (low-aw) foods. Persistence of the pathogen in these foods was attributed to its ability to implement desiccation resistance mechanisms. Published knowledge about methods that disrupt desiccation resistance in S. enterica is lacking. We hypothesize that strong membrane-active compounds disrupt the desiccation resistance that S. enterica may acquire in low-aw foods or environments. The newly discovered antimicrobial lipopeptide paenibacterin was the membrane-active agent investigated in this study. Strains of S. enterica serovars Tennessee and Eimsbuettel, with a history of association with low-moisture foods, were investigated. The viability of these strains did not decrease significantly during dehydration and subsequent storage in the dehydrated state. Considering that the paenibacterin MIC against S. enterica strains was 8 μg/ml, concentrations of 4 to 16 μg/ml paenibacterin were tested. Within this range, desiccation-adapted S. Eimsbuettel was much more tolerant to the antimicrobial agent than the desiccation-adapted S. Tennessee. Pretreatment with 8 μg/ml paenibacterin increased inactivation of S. enterica during desiccation. The use of paenibacterin at 16 μg/ml or higher concentrations resulted in leakage of intracellular potassium ions from desiccation-adapted cells. Paenibacterin significantly decreased the biosynthesis of the intracellular osmoprotectant solute, trehalose, in a concentration-dependent manner. Treatment with 64 μg/ml paenibacterin increased the permeability of the cytoplasmic membranes of desiccation-adapted cells. Transcription of the desiccation-related genes proV, STM1494, kdpA, and otsB in response to paenibacterin treatment was investigated using reverse transcription-quantitative PCR. Transcription of some of these genes was downregulated in a concentration- and strain-dependent manner.IMPORTANCE Salmonella enterica adapts effectively and persists for a long time in low-aw foods or environments through resistance mechanisms to desiccation stress. Desiccation-resistant cells compromise food safety and constitute a serious health hazard. Strategies to combat desiccation resistance in S. enterica are needed to sensitize the pathogen to lethal processes used in food preservation. The study proved that the membrane-active lipopeptide paenibacterin disrupts the resistance in desiccation-adapted S. enterica, as measured by phenotypic, biochemical, and genetic analyses. This study highlighted the role of the lipopeptide paenibacterin in disrupting mechanisms employed by S. enterica to resist desiccation. This knowledge may lead to the design of novel control measures to improve the safety of low-aw foods.
Collapse
|
49
|
Mullick P, Mukherjee S, Das G, Ramesh A. Generation of a Hydroxyapatite Nanocarrier through Biomineralization Using Cell-Free Extract of Lactic Acid Bacteria for Antibiofilm Application. ACS APPLIED BIO MATERIALS 2019; 2:2927-2936. [DOI: 10.1021/acsabm.9b00293] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Priya Mullick
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Sandipan Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Gopal Das
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Aiyagari Ramesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| |
Collapse
|
50
|
Characterization and Synergistic Antimicrobial Evaluation of Lipopeptides from Bacillus amyloliquefaciens Isolated from Oil-Contaminated Soil. Int J Microbiol 2019; 2019:3704198. [PMID: 30956662 PMCID: PMC6431436 DOI: 10.1155/2019/3704198] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 02/17/2019] [Indexed: 12/02/2022] Open
Abstract
Lipopeptides show great potential for biomedical application. Several lipopeptides exhibit narrow and broad-spectrum inhibition activities. The aim of the study is to characterize the lipopeptides produced by B. amyloliquefaciens strain MD4-12 and evaluate the synergistic antimicrobial activity in combination with a conventional antibiotic against Gram-negative bacteria. B. amyloliquefaciens strain MD4-12 was isolated from oil-contaminated soil. The isolate was cultivated in McKeen medium, and the lipopeptides were isolated by precipitation and extraction with methanol. Characterization of the lipopeptides by ESI-MS gave nine mass ion peaks with m/z 994–1072, resulted from protonating of the main ions in [M + H]+ and [M + Na]+ ion form. These mass ion peaks attributed to surfactin homologs. By tandem mass spectrometry, five variants of surfactin with the same amino acid sequence in peptide moiety could be revealed. The peptide moiety contains seven amino acids identified as Glu-Leu/Ile-Leu-Val-Asp-Leu-Leu/Ile while the fatty acid moiety comprises a different length of chain from C12 to C16. Surfactin showed antibacterial activity against various Gram-positive and Gram-negative bacteria. Combination surfactin with ampicillin showed a synergistic effect against P. aeruginosa ATCC 27853.
Collapse
|