1
|
Gil-Gil T, Laborda P, Martínez JL, Hernando-Amado S. Use of adjuvants to improve antibiotic efficacy and reduce the burden of antimicrobial resistance. Expert Rev Anti Infect Ther 2025; 23:31-47. [PMID: 39670956 DOI: 10.1080/14787210.2024.2441891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/28/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION The increase in antibiotic resistance, together with the absence of novel antibiotics, makes mandatory the introduction of novel strategies to optimize the use of existing antibiotics. Among these strategies, the use of molecules that increase their activity looks promising. AREAS COVERED Different categories of adjuvants have been reviewed. Anti-resistance adjuvants increase the activity of antibiotics by inhibiting antibiotic resistance determinants. Anti-virulence approaches focus on the infection process itself; reducing virulence in combination with an antibiotic can improve therapeutic efficacy. Combination of phages with antibiotics can also be useful, since they present different mechanisms of action and targets. Finally, combining antibiotics with adjuvants in the same molecule may serve to improve antibiotics' efficacy and to overcome potential problems of differential pharmacokinetics/pharmacodynamics. EXPERT OPINION The successful combination of inhibitors of β-lactamases with β-lactams has shown that adjuvants can improve the efficacy of current antibiotics. In this sense, novel anti-resistance adjuvants able to inhibit efflux pumps are still needed, as well as anti-virulence compounds that improve the efficacy of antibiotics by interfering with the infection process. Although adjuvants may present different pharmacodynamics/pharmacokinetics than antibiotics, conjugates containing both compounds can solve this problem. Finally, already approved drugs can be a promising source of antibiotic adjuvants.
Collapse
Affiliation(s)
- Teresa Gil-Gil
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Pablo Laborda
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | | | | |
Collapse
|
2
|
Nguyen DD, Duong TH, Nguyen TP, Nguyen HT, Nguyen CH. Antibacterial Potential of Ethyl 3,5-Dibromoorsellinate, a Derivative of Diphenyl Ethers from Graphis handelii, against Methicillin-Resistant Staphylococcus aureus. ACS OMEGA 2024; 9:50012-50023. [PMID: 39713661 PMCID: PMC11656388 DOI: 10.1021/acsomega.4c09518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 12/24/2024]
Abstract
Staphylococcus aureus is a human pathogen responsible for a variety of diseases, from skin, soft tissue, and lung infections to severe cases such as meningitis, infective endocarditis, and bacteremia. The high level of antibiotic resistance in these pathogens, exemplified by methicillin-resistant Staphylococcus aureus (MRSA), necessitates the development of effective antibiotics. Thus, this work introduced the chemical synthesis of ethyl 3,5-dibromoorsellinate, a derivative of ethyl orsellinate from the lichen mycobiont of Graphis handelii, and its effectiveness against MRSA was assessed. Results showed that ethyl 3,5-dibromoorsellinate efficiently inhibited MRSA with a minimum inhibitory concentration (MIC) of 4 μg/mL, and the time-kill analysis showed the bactericidal effect of ethyl 3,5-dibromoorsellinate on MRSA at 8× MIC after 24 h. The compound also exhibited selective activity against MRSA compared with the human cell line, with a selectivity index of 12.5-fold. While ethyl 3,5-dibromoorsellinate exhibited an indifferent effect with ampicillin, this compound demonstrated antagonistic effects with kanamycin in the synergistic assessment. Additionally, ethyl 3,5-dibromoorsellinate demonstrated antibiofilm activity against MRSA starting from 0.25× MIC. The molecular docking investigation illustrated that ethyl 3,5-dibromoorsellinate binds with the penicillin-binding protein 2A of MRSA with a free energy of -42.5 to -45.7 kcal/mol. Given its promising antibacterial activities, ethyl 3,5-dibromoorsellinate warrants further investigation as a potential antibiotic option against MRSA.
Collapse
Affiliation(s)
- Dao Dinh Nguyen
- Faculty of
Biology-Biotechnology, University of Science, Vietnam National University Ho Chi Minh City, 227 Nguyen Van Cu, District 5, Ho Chi Minh City 749000, Vietnam
| | - Thuc-Huy Duong
- Department
of Chemistry, Ho Chi Minh City University
of Education, 280 An Duong Vuong, District 5, Ho Chi Minh
City 748342, Vietnam
| | - Thi-Phuong Nguyen
- NTT Hi-Tech
Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh, District
4, Ho Chi Minh City 700000, Vietnam
| | - Huy Truong Nguyen
- Faculty of
Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Chuong Hoang Nguyen
- Faculty of
Biology-Biotechnology, University of Science, Vietnam National University Ho Chi Minh City, 227 Nguyen Van Cu, District 5, Ho Chi Minh City 749000, Vietnam
| |
Collapse
|
3
|
Pani S, Mohapatra SS. Phenotypic heterogeneity in bacteria: the rise of antibiotic persistence, clinical implications, and therapeutic opportunities. Arch Microbiol 2024; 206:446. [PMID: 39460765 DOI: 10.1007/s00203-024-04173-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
The rising incidence of antimicrobial resistance (AMR) and the diminishing antibiotics discovery pipeline have created an unprecedented scenario where minor infections could become untreatable. AMR phenomenon is genetically encoded, and various genetic determinants have been implicated in their emergence and spread. Nevertheless, several non-genetic phenomena are also involved in antibiotic treatment failure which requires a systematic investigation. It has been observed that in an isogenic population of bacteria, not all cells behave or respond the same way to an antibiotic, because of the inherent heterogeneity among them. This heterogeneity is not always heritable but rather phenotypic. Three distinct types of phenotypic heterogeneity, namely tolerance, persistence, and heteroresistance have been observed in bacteria having significant clinical implications influencing the treatment outcome. While tolerance is when a population can survive high doses of antibiotics without changing the minimum inhibitory concentration (MIC) of the drug, persistence occurs in a subpopulation of bacteria that can survive exposure to high antibiotic doses. In contrast, when a subpopulation shows a very high MIC in comparison to the rest of the population, the phenomenon is called heteroresistance. In this article, we have highlighted bacterial persistence with a focus on their emergence and the underlying molecular mechanisms. Moreover, we have tried to associate the genome-wide methylation status with that of the heterogeneity at a single-cell level that may explain the role of epigenetic mechanisms in the development of persistence.
Collapse
Affiliation(s)
- Srimayee Pani
- Molecular Microbiology Lab, Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India
| | - Saswat S Mohapatra
- Molecular Microbiology Lab, Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur, Odisha, 760007, India.
| |
Collapse
|
4
|
Cassa MA, Gentile P, Girón-Hernández J, Ciardelli G, Carmagnola I. Smart self-defensive coatings with bacteria-triggered antimicrobial response for medical devices. Biomater Sci 2024; 12:5433-5449. [PMID: 39320148 DOI: 10.1039/d4bm00936c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Bacterial colonization and biofilm formation on medical devices represent one of the most urgent and critical challenges in modern healthcare. These issues not only pose serious threats to patient health by increasing the risk of infections but also exert a considerable economic burden on national healthcare systems due to prolonged hospital stays and additional treatments. To address this challenge, there is a need for smart, customized biomaterials for medical device fabrication, particularly through the development of surface modification strategies that prevent bacterial adhesion and the growth of mature biofilms. This review explores three bioinspired approaches through which antibacterial and antiadhesive coatings can be engineered to exhibit smart, stimuli-responsive features. This responsiveness is greatly valuable as it provides the coatings with a controlled, on-demand antibacterial response that is activated only in the presence of bacteria, functioning as self-defensive coatings. Such coatings can be designed to release antibacterial agents or change their surface properties/conformation in response to specific stimuli, like changes in pH, temperature, or the presence of bacterial enzymes. This targeted approach minimizes the risk of developing antibiotic resistance and reduces the need for continuous, high-dose antibacterial treatments, thereby preserving the natural microbiome and further reducing healthcare costs. The final part of the review reports a critical analysis highlighting the potential improvements and future evolutions regarding antimicrobial self-defensive coatings and their validation.
Collapse
Affiliation(s)
- Maria Antonia Cassa
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Torino 10129, Italy.
- Politecnico di Torino, Polito BIOmed Lab, Torino 10129, Italy
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Joel Girón-Hernández
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Gianluca Ciardelli
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Torino 10129, Italy.
- Politecnico di Torino, Polito BIOmed Lab, Torino 10129, Italy
- National Research Council, Institute for Chemical and Physical Processes (CNR-IPCF), Pisa 56124, Italy
| | - Irene Carmagnola
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Torino 10129, Italy.
- Politecnico di Torino, Polito BIOmed Lab, Torino 10129, Italy
| |
Collapse
|
5
|
D’Angelo C, Faggiano S, Imbimbo P, Viale E, Casillo A, Bettati S, Olimpo D, Tutino ML, Monti DM, Corsaro MM, Ronda L, Parrilli E. Pentadecanoic Acid-Releasing PDMS: Towards a New Material to Prevent S. epidermidis Biofilm Formation. Int J Mol Sci 2024; 25:10727. [PMID: 39409056 PMCID: PMC11476977 DOI: 10.3390/ijms251910727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Microbial biofilm formation on medical devices paves the way for device-associated infections. Staphylococcus epidermidis is one of the most common strains involved in such infections as it is able to colonize numerous devices, such as intravenous catheters, prosthetic joints, and heart valves. We previously reported the antibiofilm activity against S. epidermidis of pentadecanoic acid (PDA) deposited by drop-casting on the silicon-based polymer poly(dimethyl)siloxane (PDMS). This material exerted an antibiofilm activity by releasing PDA; however, a toxic effect on bacterial cells was observed, which could potentially favor the emergence of resistant strains. To develop a PDA-functionalized material for medical use and overcome the problem of toxicity, we produced PDA-doped PDMS by either spray-coating or PDA incorporation during PDMS polymerization. Furthermore, we created a strategy to assess the kinetics of PDA release using ADIFAB, a very sensitive free fatty acids fluorescent probe. Spray-coating resulted in the most promising strategy as the concentration of released PDA was in the range 0.8-1.5 μM over 21 days, ensuring long-term effectiveness of the antibiofilm molecule. Moreover, the new coated material resulted biocompatible when tested on immortalized human keratinocytes. Our results indicate that PDA spray-coated PDMS is a promising material for the production of medical devices endowed with antibiofilm activity.
Collapse
Affiliation(s)
- Caterina D’Angelo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Serena Faggiano
- Department of Food and Drug, University of Parma, Parco Area delle Scienze 23/A, 43124 Parma, Italy
- Institute of Biophysics, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (S.B.); (L.R.)
| | - Paola Imbimbo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Elisabetta Viale
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
| | - Angela Casillo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Stefano Bettati
- Institute of Biophysics, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (S.B.); (L.R.)
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
| | - Diana Olimpo
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Maria Luisa Tutino
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Daria Maria Monti
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Maria Michela Corsaro
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| | - Luca Ronda
- Institute of Biophysics, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (S.B.); (L.R.)
- Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
| | - Ermenegilda Parrilli
- Department of Chemical Sciences, University of Naples “Federico II”, Complesso Universitario Monte S. Angelo, Via Cintia 4, 80126 Naples, Italy; (C.D.); (P.I.); (A.C.); (D.O.); (M.L.T.); (D.M.M.); (M.M.C.)
| |
Collapse
|
6
|
Dhadwal S, Handa S, Chatterjee M, Banat IM. Sophorolipid: An Effective Biomolecule for Targeting Microbial Biofilms. Curr Microbiol 2024; 81:388. [PMID: 39367190 DOI: 10.1007/s00284-024-03892-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Biofilms are microbial aggregates encased in a matrix that is attached to biological or nonbiological surfaces and constitute serious problems in food, medical, and marine industries and can have major negative effects on both health and the economy. Biofilm's complex microbial community provides a resistant environment that is difficult to eradicate and is extremely resilient to antibiotics and sanitizers. There are various conventional techniques for combating biofilms, including, chemical removal, physical or mechanical removal, use of antibiotics and disinfectants to destroy biofilm producing organisms. In contrast to free living planktonic cells, biofilms are very resistant to these methods. Hence, new strategies that differ from traditional approaches are urgently required. Microbial world offers a wide range of effective "green" compounds such as biosurfactants. They outperform synthetic surfactants in terms of biodegradability, superior stabilization, and reduced toxicity concerns. They also have better antiadhesive and anti-biofilm capabilities which can be used to treat biofilm-related problems. Sophorolipids (SLs) are a major type of biosurfactants that have gained immense interest in the healthcare industries because of their antiadhesive and anti-biofilm properties. Sophorolipids may therefore prove to be attractive substances that can be used in biomedical applications as adjuvant to other antibiotics against some infections through growth inhibition and/or biofilm disruption.
Collapse
Affiliation(s)
- Sunidhi Dhadwal
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India
| | - Shristi Handa
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India
| | - Mary Chatterjee
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India.
| | - Ibrahim M Banat
- Faculty of Life & Health Sciences, University of Ulster, Coleraine, BT52 1SA, UK.
| |
Collapse
|
7
|
Gong F, Xin S, Liu X, He C, Yu X, Pan L, Zhang S, Gao H, Xu J. Multiple biological characteristics and functions of intestinal biofilm extracellular polymers: friend or foe? Front Microbiol 2024; 15:1445630. [PMID: 39224216 PMCID: PMC11367570 DOI: 10.3389/fmicb.2024.1445630] [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: 06/07/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The gut microbiota is vital to human health, and their biofilms significantly impact intestinal immunity and the maintenance of microbial balance. Certain pathogens, however, can employ biofilms to elude identification by the immune system and medical therapy, resulting in intestinal diseases. The biofilm is formed by extracellular polymorphic substances (EPS), which shield microbial pathogens from the host immune system and enhance its antimicrobial resistance. Therefore, investigating the impact of extracellular polysaccharides released by pathogens that form biofilms on virulence and defence mechanisms is crucial. In this review, we provide a comprehensive overview of current pathogenic biofilm research, deal with the role of extracellular polymers in the formation and maintenance of pathogenic biofilm, and elaborate different prevention and treatment strategies to provide an innovative approach to the treatment of intestinal pathogen-based diseases.
Collapse
Affiliation(s)
- Fengrong Gong
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaohui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinyi Yu
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Luming Pan
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sitian Zhang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Han Gao
- Department of Clinical Laboratory, Aerospace Center Hospital, Beijing, China
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Kaplan JB, Sukhishvili SA, Sailer M, Kridin K, Ramasubbu N. Aggregatibacter actinomycetemcomitans Dispersin B: The Quintessential Antibiofilm Enzyme. Pathogens 2024; 13:668. [PMID: 39204268 PMCID: PMC11357414 DOI: 10.3390/pathogens13080668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
The extracellular matrix of most bacterial biofilms contains polysaccharides, proteins, and nucleic acids. These biopolymers have been shown to mediate fundamental biofilm-related phenotypes including surface attachment, intercellular adhesion, and biocide resistance. Enzymes that degrade polymeric biofilm matrix components, including glycoside hydrolases, proteases, and nucleases, are useful tools for studying the structure and function of biofilm matrix components and are also being investigated as potential antibiofilm agents for clinical use. Dispersin B is a well-studied, broad-spectrum antibiofilm glycoside hydrolase produced by Aggregatibacter actinomycetemcomitans. Dispersin B degrades poly-N-acetylglucosamine, a biofilm matrix polysaccharide that mediates biofilm formation, stress tolerance, and biocide resistance in numerous Gram-negative and Gram-positive pathogens. Dispersin B has been shown to inhibit biofilm and pellicle formation; detach preformed biofilms; disaggregate bacterial flocs; sensitize preformed biofilms to detachment by enzymes, detergents, and metal chelators; and sensitize preformed biofilms to killing by antiseptics, antibiotics, bacteriophages, macrophages, and predatory bacteria. This review summarizes the results of nearly 100 in vitro and in vivo studies that have been carried out on dispersin B since its discovery 20 years ago. These include investigations into the biological function of the enzyme, its structure and mechanism of action, and its in vitro and in vivo antibiofilm activities against numerous bacterial species. Also discussed are potential clinical applications of dispersin B.
Collapse
Affiliation(s)
- Jeffrey B. Kaplan
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel;
| | - Svetlana A. Sukhishvili
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77843, USA;
| | | | - Khalaf Kridin
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel;
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Narayanan Ramasubbu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA;
| |
Collapse
|
9
|
Ralhan K, Iyer KA, Diaz LL, Bird R, Maind A, Zhou QA. Navigating Antibacterial Frontiers: A Panoramic Exploration of Antibacterial Landscapes, Resistance Mechanisms, and Emerging Therapeutic Strategies. ACS Infect Dis 2024; 10:1483-1519. [PMID: 38691668 PMCID: PMC11091902 DOI: 10.1021/acsinfecdis.4c00115] [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: 02/10/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024]
Abstract
The development of effective antibacterial solutions has become paramount in maintaining global health in this era of increasing bacterial threats and rampant antibiotic resistance. Traditional antibiotics have played a significant role in combating bacterial infections throughout history. However, the emergence of novel resistant strains necessitates constant innovation in antibacterial research. We have analyzed the data on antibacterials from the CAS Content Collection, the largest human-curated collection of published scientific knowledge, which has proven valuable for quantitative analysis of global scientific knowledge. Our analysis focuses on mining the CAS Content Collection data for recent publications (since 2012). This article aims to explore the intricate landscape of antibacterial research while reviewing the advancement from traditional antibiotics to novel and emerging antibacterial strategies. By delving into the resistance mechanisms, this paper highlights the need to find alternate strategies to address the growing concern.
Collapse
Affiliation(s)
| | | | - Leilani Lotti Diaz
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert Bird
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Ankush Maind
- ACS
International India Pvt. Ltd., Pune 411044, India
| | | |
Collapse
|
10
|
Li B, Mao J, Wu J, Mao K, Jia Y, Chen F, Liu J. Nano-Bio Interactions: Biofilm-Targeted Antibacterial Nanomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306135. [PMID: 37803439 DOI: 10.1002/smll.202306135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/10/2023] [Indexed: 10/08/2023]
Abstract
Biofilm is a spatially organized community formed by the accumulation of both microorganisms and their secretions, leading to persistent and chronic infections because of high resistance toward conventional antibiotics. In view of the tunable physicochemical properties and the related unique biological behavior (e.g., size-, shape-, and surface charge-dependent penetration, protein corona endowed targeting, catalytic- and electronic-related oxidative stress, optical- and magnetic-associated hyperthermia, etc.), nanomaterials-based therapeutics are widely used for the treatment of biofilm-associated infections. In this review, the biological characteristics of biofilm are introduced. And the nanomaterials-based antibacterial strategies are further discussed via biofilm targeting, including preventing biofilm formation, enhancing biofilm penetration, disrupting the mature biofilm, and acting as drug delivery systems. In which, the interactions between biofilm and nanomaterials include mechanical disruption, electron transfer, enzymatic degradation, oxidative stress, and hyperthermia. Additionally, the current advances of nanomaterials for antibacterial nanomaterials by biofilm targeting are summarized. This review aims to present a complete vision of antibacterial nanomaterials-biofilm (nano-bio) interactions, paving the way for the future development and clinical translation of effective antibacterial nanomedicines.
Collapse
Affiliation(s)
- Bo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jiahui Mao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Jiawei Wu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, P. R. China
| | - Kerou Mao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, P. R. China
| | - Yangrui Jia
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, P. R. China
| | - Fulin Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, P. R. China
| | - Jing Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing, 100190, P. R. China
| |
Collapse
|
11
|
Elkhalifa ME, Ashraf M, Ahmed A, Usman A, Hamdoon AA, Elawad MA, Almalki MG, Mosa OF, Niyazov LN, Ayaz M. Polyphenols and their nanoformulations as potential antibiofilm agents against multidrug-resistant pathogens. Future Microbiol 2024; 19:255-279. [PMID: 38305223 DOI: 10.2217/fmb-2023-0175] [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: 08/09/2023] [Accepted: 10/13/2023] [Indexed: 02/03/2024] Open
Abstract
The emergence of multidrug-resistant (MDR) pathogens is a major problem in the therapeutic management of infectious diseases. Among the bacterial resistance mechanisms is the development of an enveloped protein and polysaccharide-hydrated matrix called a biofilm. Polyphenolics have demonstrated beneficial antibacterial effects. Phenolic compounds mediate their antibiofilm effects via disruption of the bacterial membrane, deprivation of substrate, protein binding, binding to adhesion complex, viral fusion blockage and interactions with eukaryotic DNA. However, these compounds have limitations of chemical instability, low bioavailability, poor water solubility and short half-lives. Nanoformulations offer a promising solution to overcome these challenges by enhancing their antibacterial potential. This review summarizes the antibiofilm role of polyphenolics, their underlying mechanisms and their potential role as resistance-modifying agents.
Collapse
Affiliation(s)
- Modawy Em Elkhalifa
- Public Health Department, Health Sciences College at Al-Leith, Umm Al-Qura University, Al leith, KSA
- Faculty of Public and Environmental Health, University of Khartoum, Khartoum, Sudan
| | - Muhammad Ashraf
- Department of Pharmacy, University of Malakand, Chakdara, Dir (L), KP, 18800, Pakistan
| | - Alshebli Ahmed
- Public Health Department, Health Sciences College at Al-Leith, Umm Al-Qura University, Al leith, KSA
- Faculty of Public and Environmental Health, University of Khartoum, Khartoum, Sudan
| | - Assad Usman
- Department of Pharmacy, University of Malakand, Chakdara, Dir (L), KP, 18800, Pakistan
| | - Alashary Ae Hamdoon
- Public Health Department, Health Sciences College at Al-Leith, Umm Al-Qura University, Al leith, KSA
- Faculty of Public and Environmental Health, University of Khartoum, Khartoum, Sudan
| | - Mohammed A Elawad
- Public Health Department, Health Sciences College at Al-Leith, Umm Al-Qura University, Al leith, KSA
- Faculty of Public and Environmental Health, University of Khartoum, Khartoum, Sudan
| | - Meshari G Almalki
- Public Health Department, Health Sciences College at Al-Leith, Umm Al-Qura University, Al leith, KSA
| | - Osama F Mosa
- Public Health Department, Health Sciences College at Al-Leith, Umm Al-Qura University, Al leith, KSA
| | - Laziz N Niyazov
- Medical Chemistry Department, Bukhara State Medical Institute Named After Abu Ali Ibn Sino, Bukhara, Uzbekistan
| | - Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Chakdara, Dir (L), KP, 18800, Pakistan
| |
Collapse
|
12
|
Wang X, Liu M, Yu C, Li J, Zhou X. Biofilm formation: mechanistic insights and therapeutic targets. MOLECULAR BIOMEDICINE 2023; 4:49. [PMID: 38097907 PMCID: PMC10721784 DOI: 10.1186/s43556-023-00164-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Biofilms are complex multicellular communities formed by bacteria, and their extracellular polymeric substances are observed as surface-attached or non-surface-attached aggregates. Many types of bacterial species found in living hosts or environments can form biofilms. These include pathogenic bacteria such as Pseudomonas, which can act as persistent infectious hosts and are responsible for a wide range of chronic diseases as well as the emergence of antibiotic resistance, thereby making them difficult to eliminate. Pseudomonas aeruginosa has emerged as a model organism for studying biofilm formation. In addition, other Pseudomonas utilize biofilm formation in plant colonization and environmental persistence. Biofilms are effective in aiding bacterial colonization, enhancing bacterial resistance to antimicrobial substances and host immune responses, and facilitating cell‒cell signalling exchanges between community bacteria. The lack of antibiotics targeting biofilms in the drug discovery process indicates the need to design new biofilm inhibitors as antimicrobial drugs using various strategies and targeting different stages of biofilm formation. Growing strategies that have been developed to combat biofilm formation include targeting bacterial enzymes, as well as those involved in the quorum sensing and adhesion pathways. In this review, with Pseudomonas as the primary subject of study, we review and discuss the mechanisms of bacterial biofilm formation and current therapeutic approaches, emphasizing the clinical issues associated with biofilm infections and focusing on current and emerging antibiotic biofilm strategies.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ming Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chuanjiang Yu
- Institute for Cancer Genetics, Columbia University, New York, NY, 10032, USA
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Xikun Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Lyons B, Balkaran JPR, Dunn-Lawless D, Lucian V, Keller SB, O’Reilly CS, Hu L, Rubasingham J, Nair M, Carlisle R, Stride E, Gray M, Coussios C. Sonosensitive Cavitation Nuclei-A Customisable Platform Technology for Enhanced Therapeutic Delivery. Molecules 2023; 28:7733. [PMID: 38067464 PMCID: PMC10708135 DOI: 10.3390/molecules28237733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Ultrasound-mediated cavitation shows great promise for improving targeted drug delivery across a range of clinical applications. Cavitation nuclei-sound-sensitive constructs that enhance cavitation activity at lower pressures-have become a powerful adjuvant to ultrasound-based treatments, and more recently emerged as a drug delivery vehicle in their own right. The unique combination of physical, biological, and chemical effects that occur around these structures, as well as their varied compositions and morphologies, make cavitation nuclei an attractive platform for creating delivery systems tuned to particular therapeutics. In this review, we describe the structure and function of cavitation nuclei, approaches to their functionalization and customization, various clinical applications, progress toward real-world translation, and future directions for the field.
Collapse
Affiliation(s)
- Brian Lyons
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Joel P. R. Balkaran
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Darcy Dunn-Lawless
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Veronica Lucian
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Sara B. Keller
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Colm S. O’Reilly
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX1 3PJ, UK;
| | - Luna Hu
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Jeffrey Rubasingham
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Malavika Nair
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Robert Carlisle
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Michael Gray
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| | - Constantin Coussios
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, UK; (J.P.R.B.); (D.D.-L.); (V.L.); (S.B.K.); (L.H.); (J.R.); (M.N.); (R.C.); (E.S.); (M.G.)
| |
Collapse
|
14
|
Baidamshina DR, Trizna EY, Goncharova SS, Sorokin AV, Lavlinskaya MS, Melnik AP, Gafarova LF, Kharitonova MA, Ostolopovskaya OV, Artyukhov VG, Sokolova EA, Holyavka MG, Bogachev MI, Kayumov AR, Zelenikhin PV. The Effect of Ficin Immobilized on Carboxymethyl Chitosan on Biofilms of Oral Pathogens. Int J Mol Sci 2023; 24:16090. [PMID: 38003281 PMCID: PMC10671066 DOI: 10.3390/ijms242216090] [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: 10/13/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
In the last decade, Ficin, a proteolytic enzyme extracted from the latex sap of the wild fig tree, has been widely investigated as a promising tool for the treatment of microbial biofilms, wound healing, and oral care. Here we report the antibiofilm properties of the enzyme immobilized on soluble carboxymethyl chitosan (CMCh) and CMCh itself. Ficin was immobilized on CMCh with molecular weights of either 200, 350 or 600 kDa. Among them, the carrier with a molecular weight of 200 kDa bound the maximum amount of enzyme, binding up to 49% of the total protein compared to 19-32% of the total protein bound to other CMChs. Treatment with pure CMCh led to the destruction of biofilms formed by Streptococcus salivarius, Streptococcus gordonii, Streptococcus mutans, and Candida albicans, while no apparent effect on Staphylococcus aureus was observed. A soluble Ficin was less efficient in the destruction of the biofilms formed by Streptococcus sobrinus and S. gordonii. By contrast, treatment with CMCh200-immobilized Ficin led to a significant reduction of the biofilms of the primary colonizers S. gordonii and S. mutans. In model biofilms obtained by the inoculation of swabs from teeth of healthy volunteers, the destruction of the biofilm by both soluble and immobilized Ficin was observed, although the degree of the destruction varied between artificial plaque samples. Nevertheless, combined treatment of oral Streptococci biofilm by enzyme and chlorhexidine for 3 h led to a significant decrease in the viability of biofilm-embedded cells, compared to solely chlorhexidine application. This suggests that the use of either soluble or immobilized Ficin would allow decreasing the amount and/or concentration of the antiseptics required for oral care or improving the efficiency of oral cavity sanitization.
Collapse
Affiliation(s)
- Diana R. Baidamshina
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| | - Elena Yu. Trizna
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| | - Svetlana S. Goncharova
- Department of Biophysics and Biotechnology, Voronezh State University, 394018 Voronezh, Russia; (S.S.G.); (A.V.S.); (M.S.L.); (V.G.A.); (M.G.H.)
| | - Andrey V. Sorokin
- Department of Biophysics and Biotechnology, Voronezh State University, 394018 Voronezh, Russia; (S.S.G.); (A.V.S.); (M.S.L.); (V.G.A.); (M.G.H.)
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 299053 Sevastopol, Russia
| | - Maria S. Lavlinskaya
- Department of Biophysics and Biotechnology, Voronezh State University, 394018 Voronezh, Russia; (S.S.G.); (A.V.S.); (M.S.L.); (V.G.A.); (M.G.H.)
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 299053 Sevastopol, Russia
| | - Anastasia P. Melnik
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| | - Leysan F. Gafarova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| | - Maya A. Kharitonova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| | - Olga V. Ostolopovskaya
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| | - Valeriy G. Artyukhov
- Department of Biophysics and Biotechnology, Voronezh State University, 394018 Voronezh, Russia; (S.S.G.); (A.V.S.); (M.S.L.); (V.G.A.); (M.G.H.)
| | - Evgenia A. Sokolova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| | - Marina G. Holyavka
- Department of Biophysics and Biotechnology, Voronezh State University, 394018 Voronezh, Russia; (S.S.G.); (A.V.S.); (M.S.L.); (V.G.A.); (M.G.H.)
- Laboratory of Bioresource Potential of Coastal Area, Institute for Advanced Studies, Sevastopol State University, 299053 Sevastopol, Russia
| | - Mikhail I. Bogachev
- Biomedical Engineering Research Centre, St. Petersburg Electrotechnical University, 197022 St. Petersburg, Russia;
| | - Airat R. Kayumov
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
- Interdepartment Research Laboratory, Kazan State Academy of Veterinary Medicine Named after N. E. Bauman, 420029 Kazan, Russia
| | - Pavel V. Zelenikhin
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia; (D.R.B.); (E.Y.T.); (A.P.M.); (L.F.G.); (M.A.K.); (O.V.O.); (E.A.S.); (A.R.K.)
| |
Collapse
|
15
|
Yang S, Li X, Cang W, Mu D, Ji S, An Y, Wu R, Wu J. Biofilm tolerance, resistance and infections increasing threat of public health. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:233-247. [PMID: 37933277 PMCID: PMC10625689 DOI: 10.15698/mic2023.11.807] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 11/08/2023]
Abstract
Microbial biofilms can cause chronic infection. In the clinical setting, the biofilm-related infections usually persist and reoccur; the main reason is the increased antibiotic resistance of biofilms. Traditional antibiotic therapy is not effective and might increase the threat of antibiotic resistance to public health. Therefore, it is urgent to study the tolerance and resistance mechanism of biofilms to antibiotics and find effective therapies for biofilm-related infections. The tolerance mechanism and host reaction of biofilm to antibiotics are reviewed, and bacterial biofilm related diseases formed by human pathogens are discussed thoroughly. The review also explored the role of biofilms in the development of bacterial resistance mechanisms and proposed therapeutic intervention strategies for biofilm related diseases.
Collapse
Affiliation(s)
- Shanshan Yang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Xinfei Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
| | - Weihe Cang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
| | - Delun Mu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Shuaiqi Ji
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Yuejia An
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| |
Collapse
|
16
|
Kalia VC, Patel SKS, Lee JK. Bacterial biofilm inhibitors: An overview. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115389. [PMID: 37634478 DOI: 10.1016/j.ecoenv.2023.115389] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
Bacteria that cause infectious diseases adopt biofilms as one of their most prevalent lifestyles. Biofilms enable bacteria to tolerate environmental stress and evade antibacterial agents. This bacterial defense mechanism has rendered the use of antibiotics ineffective for the treatment of infectious diseases. However, many highly drug-resistant microbes have rapidly emerged owing to such treatments. Different signaling mechanisms regulate bacterial biofilm formation, including cyclic dinucleotide (c-di-GMP), small non-coding RNAs, and quorum sensing (QS). A cell density-dependent phenomenon, QS is associated with c-di-GMP (a global messenger), which regulates gene expression related to adhesion, extracellular matrix production, the transition from the planktonic to biofilm stage, stability, pathogenicity, virulence, and acquisition of nutrients. The article aims to provide information on inhibiting biofilm formation and disintegrating mature/preformed biofilms. This treatment enables antimicrobials to target the free-living/exposed bacterial cells at lower concentrations than those needed to treat bacteria within the biofilm. Therefore, a complementary action of antibiofilm and antimicrobial agents can be a robust strategic approach to dealing with infectious diseases. Taken together, these molecules have broad implications for human health.
Collapse
Affiliation(s)
- Vipin Chandra Kalia
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sanjay K S Patel
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jung-Kul Lee
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
17
|
Pan X, Liu W, Du Q, Zhang H, Han D. Recent Advances in Bacterial Persistence Mechanisms. Int J Mol Sci 2023; 24:14311. [PMID: 37762613 PMCID: PMC10531727 DOI: 10.3390/ijms241814311] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The recurrence of bacterial infectious diseases is closely associated with bacterial persisters. This subpopulation of bacteria can escape antibiotic treatment by entering a metabolic status of low activity through various mechanisms, for example, biofilm, toxin-antitoxin modules, the stringent response, and the SOS response. Correspondingly, multiple new treatments are being developed. However, due to their spontaneous low abundance in populations and the lack of research on in vivo interactions between persisters and the host's immune system, microfluidics, high-throughput sequencing, and microscopy techniques are combined innovatively to explore the mechanisms of persister formation and maintenance at the single-cell level. Here, we outline the main mechanisms of persister formation, and describe the cutting-edge technology for further research. Despite the significant progress regarding study techniques, some challenges remain to be tackled.
Collapse
Affiliation(s)
- Xiaozhou Pan
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Wenxin Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Qingqing Du
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| | - Dingding Han
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai 200062, China
| |
Collapse
|
18
|
Ramya RK, Theraka K, Ramprasadh SV, Bharathi SV, Srinivasan S, Jacob S, Kuila A. Pragmatic Treatment Strategies for Polyaromatic Hydrocarbon Remediation and Anti-biofouling from Surfaces Using Nano-enzymes: a Review. Appl Biochem Biotechnol 2023; 195:5479-5496. [PMID: 35138553 DOI: 10.1007/s12010-022-03848-1] [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] [Accepted: 01/31/2022] [Indexed: 11/02/2022]
Abstract
In this review, two important environmental pollutants have been considered for its potential remediation using microbial-derived nano-enzymes. Firstly, polyaromatic hydrocarbons (PAHs) are one of the major industrial contaminants in the environment due to their ubiquitous occurrence, toxicity, and proclivity for bioaccumulation. Secondly, biofouling due to biofilm-forming organisms that impact tremendous economic and environmental consequences in many industries, especially marine vessels where it causes an increase in hydrodynamic drag, which results in a loss of ship speed at constant power or a power increase to maintain the same speed with higher fuel consumption and emissions into the atmosphere, particularly Green House Gases (GHGs). Among the remediation strategies, biological routes are found to be promising, efficient, and sustainable. Natural ligninolytic enzymes such as MnP, LiP, laccase, peroxidases, and polysaccharide and protein degradative enzymes are found to be highly efficient for PAH degradation and antifouling respectively. However, large-scale usage of these enzymes is difficult due to various reasons like their poor stability, adaptation, and high-cost production of these enzymes. In recent years, the use of nanoparticles, particularly nano-enzymes, is found to be an innovative and synergistic approach to detoxify contaminated areas with concomitant maintenance of enzyme stability.
Collapse
Affiliation(s)
- Rajesh Khanna Ramya
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu Dist, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - Karthikeyan Theraka
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu Dist, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - Swaminathan Viji Ramprasadh
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu Dist, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - Sundaramoorthy Vijaya Bharathi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu Dist, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - S Srinivasan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu Dist, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India
| | - Samuel Jacob
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, Chengalpattu Dist, SRM Nagar, Kattankulathur, 603203, Tamil Nadu, India.
| | - Arindam Kuila
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Banasthali, Rajasthan, 304022, India.
| |
Collapse
|
19
|
Zhao N, Curry D, Evans RE, Isguven S, Freeman T, Eisenbrey JR, Forsberg F, Gilbertie JM, Boorman S, Hilliard R, Dastgheyb SS, Machado P, Stanczak M, Harwood M, Chen AF, Parvizi J, Shapiro IM, Hickok NJ, Schaer TP. Microbubble cavitation restores Staphylococcus aureus antibiotic susceptibility in vitro and in a septic arthritis model. Commun Biol 2023; 6:425. [PMID: 37069337 PMCID: PMC10110534 DOI: 10.1038/s42003-023-04752-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/24/2023] [Indexed: 04/19/2023] Open
Abstract
Treatment failure in joint infections is associated with fibrinous, antibiotic-resistant, floating and tissue-associated Staphylococcus aureus aggregates formed in synovial fluid (SynF). We explore whether antibiotic activity could be increased against Staphylococcus aureus aggregates using ultrasound-triggered microbubble destruction (UTMD), in vitro and in a porcine model of septic arthritis. In vitro, when bacterially laden SynF is diluted, akin to the dilution achieved clinically with lavage and local injection of antibiotics, amikacin and ultrasound application result in increased bacterial metabolism, aggregate permeabilization, and a 4-5 log decrease in colony forming units, independent of microbubble destruction. Without SynF dilution, amikacin + UTMD does not increase antibiotic activity. Importantly, in the porcine model of septic arthritis, no bacteria are recovered from the SynF after treatment with amikacin and UTMD-ultrasound without UTMD is insufficient. Our data suggest that UTMD + antibiotics may serve as an important adjunct for the treatment of septic arthritis.
Collapse
Affiliation(s)
- Neil Zhao
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dylan Curry
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rachel E Evans
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Selin Isguven
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Theresa Freeman
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - John R Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jessica M Gilbertie
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Sophie Boorman
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Rachel Hilliard
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Sana S Dastgheyb
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Priscilla Machado
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Stanczak
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Marc Harwood
- Rothman Orthopaedic Institute, Philadelphia, PA, USA
| | - Antonia F Chen
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Javad Parvizi
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
- Rothman Orthopaedic Institute, Philadelphia, PA, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Noreen J Hickok
- Department of Orthopaedic Surgery, Sidney Kimmel College, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Thomas P Schaer
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA.
| |
Collapse
|
20
|
Upadhyay A, Pal D, Kumar A. Combinatorial enzyme therapy: A promising neoteric approach for bacterial biofilm disruption. Process Biochem 2023. [DOI: 10.1016/j.procbio.2023.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
21
|
Mohamad F, Alzahrani RR, Alsaadi A, Alrfaei BM, Yassin AEB, Alkhulaifi MM, Halwani M. An Explorative Review on Advanced Approaches to Overcome Bacterial Resistance by Curbing Bacterial Biofilm Formation. Infect Drug Resist 2023; 16:19-49. [PMID: 36636380 PMCID: PMC9830422 DOI: 10.2147/idr.s380883] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/06/2022] [Indexed: 01/05/2023] Open
Abstract
The continuous emergence of multidrug-resistant pathogens evoked the development of innovative approaches targeting virulence factors unique to their pathogenic cascade. These approaches aimed to explore anti-virulence or anti-infective therapies. There are evident concerns regarding the bacterial ability to create a superstructure, the biofilm. Biofilm formation is a crucial virulence factor causing difficult-to-treat, localized, and systemic infections. The microenvironments of bacterial biofilm reduce the efficacy of antibiotics and evade the host's immunity. Producing a biofilm is not limited to a specific group of bacteria; however, Pseudomonas aeruginosa, Acinetobacter baumannii, and Staphylococcus aureus biofilms are exemplary models. This review discusses biofilm formation as a virulence factor and the link to antimicrobial resistance. In addition, it explores insights into innovative multi-targeted approaches and their physiological mechanisms to combat biofilms, including natural compounds, phages, antimicrobial photodynamic therapy (aPDT), CRISPR-Cas gene editing, and nano-mediated techniques.
Collapse
Affiliation(s)
- F Mohamad
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Raghad R Alzahrani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia,Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ahlam Alsaadi
- Infectious Diseases Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Bahauddeen M Alrfaei
- Stem Cells and Regenerative Medicine, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Alaa Eldeen B Yassin
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Manal M Alkhulaifi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia,Manal M Alkhulaifi, P.O. Box 55670, Riyadh, 11544, Tel +966 (11) 805-1685, Email
| | - Majed Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia,Correspondence: Majed Halwani, P.O. Box 3660, Mail Code 1515 (KAIMRC), Riyadh, 11481, Tel +966 (11) 429-4433, Fax +966 (11) 429-4440, Email ;
| |
Collapse
|
22
|
The Bovhyaluronidase Azoximer (Longidaza ®) Disrupts Candida albicans and Candida albicans-Bacterial Mixed Biofilms and Increases the Efficacy of Antifungals. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58121710. [PMID: 36556912 PMCID: PMC9782602 DOI: 10.3390/medicina58121710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022]
Abstract
Background and Objectives: Candida albicans causes various diseases ranging from superficial mycoses to life-threatening systemic infections often associated with biofilm formation, including mixed fungal−bacterial consortia. The biofilm matrix protects cells, making Candida extremely resistant to treatment. Here, we show that the bovhyaluronidase azoximer (Longidaza®) in vitro destroys the biofilm formed by either C. albicans alone or mixed with bacteria, this way decreasing the concentrations of antimicrobials required for the pathogen’s eradication. Materials and Methods: Bovhyaluronidase azoximer, Longidaza® was obtained from NPO Petrovax Pharm Ltd., Moscow, Russia as lyophilized powder. The antifungal activity was assessed by microdilution assay and CFUs counting. Antibiofilm activity was evaluated via biofilms staining and scanning electron microscopy. Results: Thus, treatment with Longidaza® reduced the biofilm biomass of nine C. albicans clinical isolates by 30−60%, while mixed biofilms of C. albicans with various bacteria were destroyed by 30−40%. Furthermore, the concentration of fluconazole required to achieve a similar reduction of the residual respiratory activity of detached cell clumps of four C. albicans isolates has been reduced four-fold when combined with Longidaza®. While in the biofilm, two of four isolates became significantly more susceptible to fluconazole in combination with Longidaza®. Conclusion: Taken together, our data indicate that Longidaza® is capable of suppression of tissues and artificial surfaces biofouling by C. albicans biofilms, as well as facilitating drug penetration into the cell clumps, this way decreasing the effective MIC of antifungals.
Collapse
|
23
|
Poilvache H, Van Bambeke F, Cornu O. Development of an innovative in vivo model of PJI treated with DAIR. Front Med (Lausanne) 2022; 9:984814. [PMID: 36314026 PMCID: PMC9606572 DOI: 10.3389/fmed.2022.984814] [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: 07/02/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Prosthetic Joint Infection (PJI) are catastrophic complications of joint replacement. Debridement, implant retention, and antibiotic therapy (DAIR) is the usual strategy in acute infections but fails in 45% of MRSA infections. We describe the development of a model of infected arthroplasty in rabbits, treated with debridement and a course of vancomycin with clinically relevant dosage. Materials and methods A total of 15 rabbits were assigned to three groups: vancomycin pharmacokinetics (A), infection (B), and DAIR (C). All groups received a tibial arthroplasty using a Ti-6Al-4V implant. Groups B and C were infected per-operatively with a 5.5 log10 MRSA inoculum. After 1 week, groups C infected knees were surgically debrided. Groups A and C received 1 week of vancomycin. Pharmacokinetic profiles were obtained in group A following 1st and 5th injections. Animals were euthanized 2 weeks after the arthroplasty. Implants and tissue samples were processed for bacterial counts and histology. Results Average vancomycin AUC0–12 h were 213.0 mg*h/L (1st injection) and 207.8 mg*h/L (5th injection), reaching clinical targets. All inoculated animals were infected. CFUs were reproducible in groups B. A sharp decrease in CFU was observed in groups C. Serum markers and leukocytes counts increased significantly in infected groups. Conclusion We developed a reproducible rabbit model of PJI treated with DAIR, using vancomycin at clinically relevant concentrations.
Collapse
Affiliation(s)
- Hervé Poilvache
- Neuro Musculo-Skeletal Laboratory, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium,Cellular and Molecular Pharmacology Laboratory, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Orthopedic Surgery and Traumatology Department, Cliniques universitaires Saint-Luc, Brussels, Belgium,*Correspondence: Hervé Poilvache,
| | - Françoise Van Bambeke
- Cellular and Molecular Pharmacology Laboratory, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Olivier Cornu
- Neuro Musculo-Skeletal Laboratory, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Brussels, Belgium,Orthopedic Surgery and Traumatology Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
24
|
Ji H, Hu H, Tang Q, Kang X, Liu X, Zhao L, Jing R, Wu M, Li G, Zhou X, Liu J, Wang Q, Cong H, Wu L, Qin Y. Precisely controlled and deeply penetrated micro-nano hybrid multifunctional motors with enhanced antibacterial activity against refractory biofilm infections. JOURNAL OF HAZARDOUS MATERIALS 2022; 436:129210. [PMID: 35739732 DOI: 10.1016/j.jhazmat.2022.129210] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
The biofilm resistance of microorganisms has severe economic and environmental implications, especially the contamination of facilities associated with human life, including medical implants, air-conditioning systems, water supply systems, and food-processing equipment, resulting in the prevalence of infectious diseases. Once bacteria form biofilms, their antibiotic resistance can increase by 10-1,000-fold, posing a great challenge to the treatment of related diseases. In order to overcome the contamination of bacterial biofilm, destroying the biofilm's matrix so as to solve the penetration depth dilemma of antibacterial agents is the most effective way. Here, a magnetically controlled multifunctional micromotor was developed by using H2O2 as the fuel and MnO2 as the catalyst to treat bacterial biofilm infection. In the presence of H2O2, the as-prepared motors could be self-propelled by the generated oxygen microbubbles. Thereby, the remotely controlled motors could drill into the EPS of biofilm and disrupt them completely with the help of bubbles. Finally, the generated highly toxic •OH could efficiently kill the unprotected bacteria. This strategy combined the mechanical damage, highly toxic •OH, and precise magnetic guidance in one system, which could effectively eliminate biologically infectious fouling in microchannels within 10 min, possessing a wide range of practical application prospects especially in large scale and complex infection sites.
Collapse
Affiliation(s)
- Haiwei Ji
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Haolu Hu
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Qu Tang
- Department of Laboratory Medicine, Affiliated hospital of Nantong University, No. 20, Xisi Road, Nantong 226001, Jiangsu, China
| | - Xiaoxia Kang
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Xiaodi Liu
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Lingfeng Zhao
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Rongrong Jing
- Department of Laboratory Medicine, Affiliated hospital of Nantong University, No. 20, Xisi Road, Nantong 226001, Jiangsu, China
| | - Mingmin Wu
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Guo Li
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Xiaobo Zhou
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Jinxia Liu
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Qi Wang
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China
| | - Hui Cong
- Department of Laboratory Medicine, Affiliated hospital of Nantong University, No. 20, Xisi Road, Nantong 226001, Jiangsu, China.
| | - Li Wu
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China.
| | - Yuling Qin
- School of Public Health, Nantong University, No. 9, Seyuan Road, Nantong 226019, Jiangsu, China.
| |
Collapse
|
25
|
An Overview of Biofilm Formation-Combating Strategies and Mechanisms of Action of Antibiofilm Agents. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081110. [PMID: 35892912 PMCID: PMC9394423 DOI: 10.3390/life12081110] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022]
Abstract
Biofilm formation on surfaces via microbial colonization causes infections and has become a major health issue globally. The biofilm lifestyle provides resistance to environmental stresses and antimicrobial therapies. Biofilms can cause several chronic conditions, and effective treatment has become a challenge due to increased antimicrobial resistance. Antibiotics available for treating biofilm-associated infections are generally not very effective and require high doses that may cause toxicity in the host. Therefore, it is essential to study and develop efficient anti-biofilm strategies that can significantly reduce the rate of biofilm-associated healthcare problems. In this context, some effective combating strategies with potential anti-biofilm agents, including plant extracts, peptides, enzymes, lantibiotics, chelating agents, biosurfactants, polysaccharides, organic, inorganic, and metal nanoparticles, etc., have been reviewed to overcome biofilm-associated healthcare problems. From their extensive literature survey, it can be concluded that these molecules with considerable structural alterations might be applied to the treatment of biofilm-associated infections, by evaluating their significant delivery to the target site of the host. To design effective anti-biofilm molecules, it must be assured that the minimum inhibitory concentrations of these anti-biofilm compounds can eradicate biofilm-associated infections without causing toxic effects at a significant rate.
Collapse
|
26
|
A previously uncharacterized gene, PA2146, contributes to biofilm formation and drug tolerance across the ɣ-Proteobacteria. NPJ Biofilms Microbiomes 2022; 8:54. [PMID: 35798749 PMCID: PMC9262955 DOI: 10.1038/s41522-022-00314-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 06/14/2022] [Indexed: 01/11/2023] Open
Abstract
Transcriptomic studies have revealed a large number of uncharacterized genes that are differentially expressed in biofilms, which may be important in regulating biofilm phenotypes such as resistance to antimicrobial agents. To identify biofilm genes of unknown function in P. aeruginosa, we made use of RNA-seq and selected 27 uncharacterized genes that were induced upon biofilm growth. Biofilms by respective mutants were subsequently analyzed for two biofilm characteristics, the biofilm architecture and drug susceptibility. The screen revealed 12 out of 27 genes to contribute to biofilm formation and 13 drug susceptibility, with 8 genes affecting both biofilm phenotypes. Amongst the genes affecting both biofilm phenotypes was PA2146, encoding a small hypothetical protein that exhibited some of the most substantial increases in transcript abundance during biofilm growth by P. aeruginosa PAO1 and clinical isolates. PA2146 is highly conserved in ɣ-proteobacteria. Inactivation of PA2146 affected both biofilm phenotypes in P. aeruginosa PAO1, with inactivation of homologs in Klebsiella pneumoniae and Escherichia coli having similar effects. Heterologous expression of PA2146 homologs complemented the P. aeruginosa ∆PA2146, suggesting that PA2146 homologs substitute for and play a similar role as PA2146 in P. aeruginosa.
Collapse
|
27
|
Anti-infective DNase I coatings on polydopamine functionalized titanium surfaces by alternating current electrophoretic deposition. Anal Chim Acta 2022; 1218:340022. [DOI: 10.1016/j.aca.2022.340022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/28/2022] [Accepted: 05/28/2022] [Indexed: 11/23/2022]
|
28
|
Kaur H, Kaur A, Soni SK, Rishi P. Microbially-derived cocktail of carbohydrases as an anti-biofouling agents: a 'green approach'. BIOFOULING 2022; 38:455-481. [PMID: 35673761 DOI: 10.1080/08927014.2022.2085566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Enzymes, also known as biocatalysts, display vital properties like high substrate specificity, an eco-friendly nature, low energy inputs, and cost-effectiveness. Among their numerous known applications, enzymes that can target biofilms or their components are increasingly being investigated for their anti-biofouling action, particularly in healthcare, food manufacturing units and environmental applications. Enzymes can target biofilms at different levels like during the attachment of microorganisms, formation of exopolymeric substances (EPS), and their disruption thereafter. In this regard, a consortium of carbohydrases that can target heterogeneous polysaccharides present in the EPS matrix may provide an effective alternative to conventional chemical anti-biofouling methods. Further, for complete annihilation of biofilms, enzymes can be used alone or in conjunction with other antimicrobial agents. Enzymes hold the promise to replace the conventional methods with greener, more economical, and more efficient alternatives. The present article explores the potential and future perspectives of using carbohydrases as effective anti-biofilm agents.
Collapse
Affiliation(s)
- Harmanpreet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Arashdeep Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
29
|
Bacterial biofilms and their resistance mechanisms: a brief look at treatment with natural agents. Folia Microbiol (Praha) 2022; 67:535-554. [DOI: 10.1007/s12223-022-00955-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/29/2022] [Indexed: 12/14/2022]
|
30
|
Abstract
Biofilms are recalcitrant to antimicrobials, partly due to the barrier effect of their matrix. The use of hydrolytic enzymes capable to degrade matrix constituents has been proposed as an alternative strategy against biofilm-related infections. This study aimed to determine whether hydrolytic enzymes could potentiate the activity of antimicrobials against hard-to-treat interkingdom biofilms comprising two bacteria and one fungus. We studied the activity of a series of enzymes alone or in combination, followed or not by antimicrobial treatment, against single-, dual- or three-species biofilms of Staphylococcus aureus, Escherichia coli, and Candida albicans, by measuring their residual biomass or culturable cells. Two hydrolytic enzymes, subtilisin A and lyticase, were identified as the most effective to reduce the biomass of C. albicans biofilm. When targeting interkingdom biofilms, subtilisin A alone was the most effective enzyme to reduce biomass of all biofilms, followed by lyticase combined with an enzymatic cocktail composed of cellulase, denarase, and dispersin B that proved previously active against bacterial biofilms. The subsequent incubation with antimicrobials further reduced the biomass. Enzymes alone did not reduce culturable cells in most cases and did not interfere with the cidal effects of antimicrobials. Therefore, this work highlights the potential interest of pre-exposing interkingdom biofilms to hydrolytic enzymes to reduce their biomass besides the number of culturable cells, which was not achieved when using antimicrobials alone. IMPORTANCE Biofilms are recalcitrant to antimicrobial treatments. This problem is even more critical when dealing with polymicrobial, interkingdom biofilms, including both bacteria and fungi, as these microorganisms cooperate to strengthen the biofilm and produce a complex matrix. Here, we demonstrate that the protease subtilisin A used alone, or a cocktail containing lyticase, cellulase, denarase, and dispersin B markedly reduce the biomass of interkingdom biofilms and cooperate with antimicrobials to act upon these recalcitrant forms of infection. This work may open perspectives for the development of novel adjuvant therapies against biofilm-related infections.
Collapse
|
31
|
Lahiri D, Nag M, Dey A, Sarkar T, Ray RR, Rebezov M, Shariati MA, Thiruvengadam M, Simal‐Gandara J. Immobilized enzymes as potent antibiofilm agent. Biotechnol Prog 2022; 38:e3281. [PMID: 35690881 PMCID: PMC9786792 DOI: 10.1002/btpr.3281] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/03/2022] [Accepted: 06/09/2022] [Indexed: 12/30/2022]
Abstract
Biofilm has been a point of concern in hospitals and various industries. They not only cause various chronic infections but are also responsible for the degradation of various medical appliances. Since the last decade, various alternate strategies are being adopted to combat the biofilm formed on various biotic and abiotic surfaces. The use of enzymes as a potent anti-fouling agent is proved to be of utmost importance as the enzymes can inhibit biofilm formation in an eco-friendly and cost-effective way. The physical and chemical immobilization of the enzyme not only leads to the improvement of thermostability and reusability of the enzyme, but also gains better efficiency of biofilm removal. Immobilization of amylase, cellobiohydrolase, pectinase, subtilisin A and β-N-acetyl-glucosaminidase (DspB) are proved to be most effective in inhibition of biofilm formation and removal of matured biofilm than their free forms. Hence, these immobilized enzymes provide greater eradication of biofilm formed on various surfaces and are coming up to be the potent antibiofilm agent.
Collapse
Affiliation(s)
- Dibyajit Lahiri
- Department of BiotechnologyUniversity of Engineering & ManagementKolkataWest BengalIndia
| | - Moupriya Nag
- Department of BiotechnologyUniversity of Engineering & ManagementKolkataWest BengalIndia
| | - Ankita Dey
- Department of MicrobiologyBelle Vue ClinicsKolkataWest BengalIndia
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda PolytechnicWest Bengal State Council of Technical Education, Govtment of West BengalMalda732102India
| | - Rina Rani Ray
- Department of BiotechnologyMaulana Abul Kalam Azad University of TechnologyHaringhataWest BengalIndia
| | - Maksim Rebezov
- V. M. Gorbatov Federal Research Center for Food SystemsMoscowRussian Federation,Prokhorov General Physics Institute of the Russian Academy of SciencesMoscowRussian Federation,Liaocheng UniversityLiaochengShandongChina
| | - Mohammad Ali Shariati
- Liaocheng UniversityLiaochengShandongChina,Department of Scientific ResearchK. G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University)MoscowRussian Federation
| | - Muthu Thiruvengadam
- Department of Crop ScienceCollege of Sanghuh Life Science, Konkuk UniversitySeoulSouth Korea
| | - Jesus Simal‐Gandara
- Universidade de Vigo, Nutrition and Bromatology GroupAnalytical Chemistry and Food Science Department, Faculty of ScienceOurenseSpain
| |
Collapse
|
32
|
Thorn CR, Howell PL, Wozniak DJ, Prestidge CA, Thomas N. Enhancing the therapeutic use of biofilm-dispersing enzymes with smart drug delivery systems. Adv Drug Deliv Rev 2021; 179:113916. [PMID: 34371086 DOI: 10.1016/j.addr.2021.113916] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 12/18/2022]
Abstract
Biofilm-dispersing enzymes degrade the extracellular polymeric matrix surrounding bacterial biofilms, disperse the microbial community and increase their susceptibility to antibiotics and immune cells. Challenges for the clinical translation of biofilm-dispersing enzymes involve their susceptibility to denaturation, degradation, and clearance upon administration in vivo. Drug delivery systems aim to overcome these limitations through encapsulation, stabilization and protection from the exterior environment, thereby maintaining the enzymatic activity. Smart drug delivery systems offer target specificity, releasing payloads at the site of infection while minimizing unnecessary systemic exposure. This review highlights critical advances of biofilm-dispersing enzymes as a novel therapeutic approach for biofilm-associated infections. We explore how smart, bio-responsive delivery systems overcome the limiting factors of biofilm-dispersing enzymes and summarize the key systems designed. This review will guide future developments, focusing on utilizing selective and specific therapies in a targeted fashion to meet the unmet therapeutic needs of biofilm infections.
Collapse
Affiliation(s)
- Chelsea R Thorn
- University of South Australia, Clinical and Health Sciences, North Tce, Adelaide, SA 5000, Australia; The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, SA 5000, Australia; Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, SA 5000, Australia
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Daniel J Wozniak
- Departments of Microbial Infection and Immunity, Ohio State University, Columbus, OH 43210, USA
| | - Clive A Prestidge
- University of South Australia, Clinical and Health Sciences, North Tce, Adelaide, SA 5000, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, SA 5000, Australia
| | - Nicky Thomas
- University of South Australia, Clinical and Health Sciences, North Tce, Adelaide, SA 5000, Australia; The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia; ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, SA 5000, Australia; Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, SA 5000, Australia.
| |
Collapse
|
33
|
Abstract
Microbes are hardly seen as planktonic species and are most commonly found as biofilm communities in cases of chronic infections. Biofilms are regarded as a biological condition, where a large group of microorganisms gets adhered to a biotic or abiotic surface. In this context, Pseudomonas aeruginosa, a Gram-negative nosocomial pathogen is the main causative organism responsible for life-threatening and persistent infections in individuals affected with cystic fibrosis and other lung ailments. The bacteria can form a strong biofilm structure when it adheres to a surface suitable for the development of a biofilm matrix. These bacterial biofilms pose higher natural resistance to conventional antibiotic therapy due to their multiple tolerance mechanisms. This prevailing condition has led to an increasing rate of treatment failures associated with P. aeruginosa biofilm infections. A better understanding of the effect of a diverse group of antibiotics on established biofilms would be necessary to avoid inappropriate treatment strategies. Hence, the search for other alternative strategies as effective biofilm treatment options has become a growing area of research. The current review aims to give an overview of the mechanisms governing biofilm formation and the different strategies employed so far in the control of biofilm infections caused by P. aeruginosa. Moreover, this review can also help researchers to search for new antibiofilm agents to tackle the effect of biofilm infections that are currently imprudent to conventional antibiotics.
Collapse
|
34
|
Opdensteinen P, Dietz SJ, Gengenbach BB, Buyel JF. Expression of Biofilm-Degrading Enzymes in Plants and Automated High-Throughput Activity Screening Using Experimental Bacillus subtilis Biofilms. Front Bioeng Biotechnol 2021; 9:708150. [PMID: 34621728 PMCID: PMC8490750 DOI: 10.3389/fbioe.2021.708150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022] Open
Abstract
Biofilm-forming bacteria are sources of infections because they are often resistant to antibiotics and chemical removal. Recombinant biofilm-degrading enzymes have the potential to remove biofilms gently, but they can be toxic toward microbial hosts and are therefore difficult to produce in bacteria. Here, we investigated Nicotiana species for the production of such enzymes using the dispersin B-like enzyme Lysobacter gummosus glyco 2 (Lg2) as a model. We first optimized transient Lg2 expression in plant cell packs using different subcellular targeting methods. We found that expression levels were transferable to differentiated plants, facilitating the scale-up of production. Our process yielded 20 mg kg−1 Lg2 in extracts but 0.3 mg kg−1 after purification, limited by losses during depth filtration. Next, we established an experimental biofilm assay to screen enzymes for degrading activity using different Bacillus subtilis strains. We then tested complex and chemically defined growth media for reproducible biofilm formation before converting the assay to an automated high-throughput screening format. Finally, we quantified the biofilm-degrading activity of Lg2 in comparison with commercial enzymes against our experimental biofilms, indicating that crude extracts can be screened directly. This ability will allow us to combine high-throughput expression in plant cell packs with automated activity screening.
Collapse
Affiliation(s)
- P Opdensteinen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.,Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| | - S J Dietz
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| | - B B Gengenbach
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.,Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| | - J F Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.,Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
35
|
Garrido V, Piñero‐Lambea C, Rodriguez‐Arce I, Paetzold B, Ferrar T, Weber M, Garcia‐Ramallo E, Gallo C, Collantes M, Peñuelas I, Serrano L, Grilló M, Lluch‐Senar M. Engineering a genome-reduced bacterium to eliminate Staphylococcus aureus biofilms in vivo. Mol Syst Biol 2021; 17:e10145. [PMID: 34612607 PMCID: PMC8493563 DOI: 10.15252/msb.202010145] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
Bacteria present a promising delivery system for treating human diseases. Here, we engineered the genome-reduced human lung pathogen Mycoplasma pneumoniae as a live biotherapeutic to treat biofilm-associated bacterial infections. This strain has a unique genetic code, which hinders gene transfer to most other bacterial genera, and it lacks a cell wall, which allows it to express proteins that target peptidoglycans of pathogenic bacteria. We first determined that removal of the pathogenic factors fully attenuated the chassis strain in vivo. We then designed synthetic promoters and identified an endogenous peptide signal sequence that, when fused to heterologous proteins, promotes efficient secretion. Based on this, we equipped the chassis strain with a genetic platform designed to secrete antibiofilm and bactericidal enzymes, resulting in a strain capable of dissolving Staphylococcus aureus biofilms preformed on catheters in vitro, ex vivo, and in vivo. To our knowledge, this is the first engineered genome-reduced bacterium that can fight against clinically relevant biofilm-associated bacterial infections.
Collapse
Affiliation(s)
- Victoria Garrido
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Institute of Agrobiotechnology (IdAB; CSIC‐Navarra Government)MutilvaSpain
| | - Carlos Piñero‐Lambea
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Pulmobiotics LtdBarcelonaSpain
| | - Irene Rodriguez‐Arce
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Institute of Agrobiotechnology (IdAB; CSIC‐Navarra Government)MutilvaSpain
| | - Bernhard Paetzold
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- S‐Biomedic N.V.BeerseBelgium
| | - Tony Ferrar
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Marc Weber
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Eva Garcia‐Ramallo
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Pulmobiotics LtdBarcelonaSpain
| | - Carolina Gallo
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - María Collantes
- RADIOMIN Research GroupClínica Universidad de NavarraPamplonaSpain
- IdiSNA, Navarra Institute for Health ResearchPamplonaSpain
| | - Iván Peñuelas
- RADIOMIN Research GroupClínica Universidad de NavarraPamplonaSpain
- IdiSNA, Navarra Institute for Health ResearchPamplonaSpain
| | - Luis Serrano
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- ICREABarcelonaSpain
| | - María‐Jesús Grilló
- Institute of Agrobiotechnology (IdAB; CSIC‐Navarra Government)MutilvaSpain
| | - María Lluch‐Senar
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Pulmobiotics LtdBarcelonaSpain
- Basic Sciences DepartmentFaculty of Medicine and Health SciencesUniversitat Internacional de CatalunyaSant Cugat del VallèsSpain
| |
Collapse
|
36
|
An Enzybiotic Regimen for the Treatment of Methicillin-Resistant Staphylococcus aureus Orthopaedic Device-Related Infection. Antibiotics (Basel) 2021; 10:antibiotics10101186. [PMID: 34680767 PMCID: PMC8533017 DOI: 10.3390/antibiotics10101186] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022] Open
Abstract
Orthopaedic device-related infection (ODRI) presents a significant challenge to the field of orthopaedic and trauma surgery. Despite extensive treatment involving surgical debridement and prolonged antibiotic therapy, outcomes remain poor. This is largely due to the unique abilities of Staphylococcus aureus, the most common causative agent of ODRI, to establish and protect itself within the host by forming biofilms on implanted devices and staphylococcal abscess communities (SACs). There is a need for novel antimicrobials that can readily target such features. Enzybiotics are a class of antimicrobial enzymes derived from bacteria and bacteriophages, which function by enzymatically degrading bacterial polymers essential to bacterial survival or biofilm formation. Here, we apply an enzybiotic-based combination regimen to a set of in vitro models as well as in a murine ODRI model to evaluate their usefulness in eradicating established S. aureus infection, compared to classical antibiotics. We show that two chimeric endolysins previously selected for their functional efficacy in human serum in combination with a polysaccharide depolymerase reduce bacterial CFU numbers 10,000-fold in a peg model and in an implant model of biofilm. The enzyme combination also completely eradicates S. aureus in a SAC in vitro model where classical antibiotics are ineffective. In an in vivo ODRI model in mice, the antibiofilm effects of this enzyme regimen are further enhanced when combined with a classical gentamicin/vancomycin treatment. In a mouse model of methicillin-resistant S. aureus (MRSA) ODRI following a fracture repair, a combined local enzybiotic/antibiotic treatment regimen showed a significant CFU reduction in the device and the surrounding soft tissue, as well as significant prevention of weight loss. These outcomes were superior to treatment with antibiotics alone. Overall, this study demonstrates that the addition of enzybiotics, which are distinguished by their extremely rapid killing efficacy and antibiofilm activities, can enhance the treatment of severe MRSA ODRI.
Collapse
|
37
|
Thorn CR, Raju D, Lacdao I, Gilbert S, Sivarajah P, Howell PL, Prestidge CA, Thomas N. Protective Liquid Crystal Nanoparticles for Targeted Delivery of PslG: A Biofilm Dispersing Enzyme. ACS Infect Dis 2021; 7:2102-2115. [PMID: 33908759 DOI: 10.1021/acsinfecdis.1c00014] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The glycoside hydrolase, PslG, attacks and degrades the dominant Psl polysaccharide in the exopolymeric substance (EPS) matrix of Pseudomonas aeruginosa biofilms and is a promising therapy to potentiate the effect of antibiotics. However, the need for coadministration with an antibiotic and the potential susceptibility of PslG to proteolysis highlights the need for an effective delivery system. Here, we compared liposomes versus lipid liquid crystal nanoparticles (LCNPs) loaded with PslG and tobramycin as potential formulation approaches to (1) protect PslG from proteolysis, (2) trigger the enzyme's release in the presence of bacteria, and (3) improve the total antimicrobial effect in vitro and in vivo in a Caenorhabditis elegans infection model. LCNPs were an effective formulation strategy for PslG and tobramycin that better protected the enzyme against proteolysis, triggered and sustained the release of PslG, improved the antimicrobial effect by 10-100-fold, and increased the survival of C. elegans infected with P. aeruginosa. Digestible LCNPs had the advantage of triggering the enzyme's release in the presence of bacteria. However, compared to nondigestible LCNPs, negligible differences arose between the LCNPs' ability to protect PslG from proteolysis and potentiate the antimicrobial activity in combination with tobramycin. In C. elegans, the improved antimicrobial efficacy was comparable to tobramycin-LCNPs, although the PslG + tobramycin-LCNPs achieved a greater than 10-fold reduction in bacteria compared to the unformulated combination. Herewith, LCNPs are showcased as a promising protective delivery system for novel biofilm dispersing enzymes combined with antibiotics, enabling infection-directed therapy and improved performance.
Collapse
Affiliation(s)
- Chelsea R. Thorn
- Clinical and Health Science, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
- The Basil Hetzel Institute for Translational Health Research, Woodville, South Australia 5011, Australia
- ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia
- Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
| | - Deepa Raju
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Ira Lacdao
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Stephanie Gilbert
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Piyanka Sivarajah
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - P. Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Clive A. Prestidge
- Clinical and Health Science, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
- ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia
| | - Nicky Thomas
- Clinical and Health Science, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
- The Basil Hetzel Institute for Translational Health Research, Woodville, South Australia 5011, Australia
- ARC Centre for Excellence in Bio-Nano Science and Technology, Adelaide, South Australia 5000, Australia
- Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, South Australia 5000, Australia
| |
Collapse
|
38
|
Khalifa AA, Bakr HM, Farouk OA. Biomaterials and technologies in the management of periprosthetic infection after total hip arthroplasty: An updated review. JOURNAL OF MUSCULOSKELETAL SURGERY AND RESEARCH 2021; 5:142-151. [DOI: 10.25259/jmsr_51_2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Although total hip arthroplasty (THA) is considered one of the most efficacious procedures for managing various hip conditions, failures due to different mechanisms are still being reported. Periprosthetic joint infection (PJI) is one of the devastating causes of failure and revision of THA. PJI carries a burden on the patient, the surgeon, and the health-care system. The diagnosis and management of PJIs carry many morbidities and increased treatment costs. The development of PJI is multifactorial, including issues related to the patient’s general condition, the surgeon’s efficiency, surgical technique, and the implants used. Recent advances in the area of diagnosis and predicting PJI as well as introducing new technologies and biomaterials update for the prevention and treatment of PJI. Local implant coatings, advancement in the bearing surfaces technologies, and new technologies such as immunotherapy and bacteriophage therapy were introduced and suggested as contemporary PJI eradication solutions. In this review, we aimed at discussing some of the newly introduced materials and technologies for the sake of PJI control.
Collapse
Affiliation(s)
- Ahmed A. Khalifa
- Department of Orthopedics, Qena Faculty of Medicine and University Hospital, South Valley University, Qena, Egypt
| | - Hatem M. Bakr
- Department of Orthopedics and Traumatology, Assiut University Hospital, Assiut, Egypt,
| | - Osama A. Farouk
- Department of Orthopedics and Traumatology, Assiut University Hospital, Assiut, Egypt,
| |
Collapse
|
39
|
Blackman LD, Qu Y, Cass P, Locock KES. Approaches for the inhibition and elimination of microbial biofilms using macromolecular agents. Chem Soc Rev 2021; 50:1587-1616. [PMID: 33403373 DOI: 10.1039/d0cs00986e] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biofilms are complex three-dimensional structures formed at interfaces by the vast majority of bacteria and fungi. These robust communities have an important detrimental impact on a wide range of industries and other facets of our daily lives, yet their removal is challenging owing to the high tolerance of biofilms towards conventional antimicrobial agents. This key issue has driven an urgent search for new innovative antibiofilm materials. Amongst these emerging approaches are highly promising materials that employ aqueous-soluble macromolecules, including peptides, proteins, synthetic polymers, and nanomaterials thereof, which exhibit a range of functionalities that can inhibit biofilm formation or detach and destroy organisms residing within established biofilms. In this Review, we outline the progress made in inhibiting and removing biofilms using macromolecular approaches, including a spotlight on cutting-edge materials that respond to environmental stimuli for "on-demand" antibiofilm activity, as well as synergistic multi-action antibiofilm materials. We also highlight materials that imitate and harness naturally derived species to achieve new and improved biomimetic and biohybrid antibiofilm materials. Finally, we share some speculative insights into possible future directions for this exciting and highly significant field of research.
Collapse
Affiliation(s)
- Lewis D Blackman
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia.
| | - Yue Qu
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia and Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Peter Cass
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia.
| | | |
Collapse
|
40
|
Lin Y, Chen J, Zhou X, Li Y. Inhibition of Streptococcus mutans biofilm formation by strategies targeting the metabolism of exopolysaccharides. Crit Rev Microbiol 2021; 47:667-677. [PMID: 33938347 DOI: 10.1080/1040841x.2021.1915959] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Dental caries is one of the most prevalent and costly biofilm-associated infectious diseases affecting most of the world's population. In particular, dental caries is driven by dysbiosis of the dental biofilm adherent to the enamel surface. Specific types of acid-producing bacteria, especially Streptococcus mutans, colonize the dental surface and cause damage to the hard tooth structure in the presence of fermentable carbohydrates. Streptococcus mutans has been established as the major cariogenic pathogen responsible for human dental caries, with a high ability to form biofilms. The exopolysaccharide (EPS) matrix, mainly contributed by S. mutans, has been considered as a virulence determinant of cariogenic biofilm. As EPS is an important virulence factor, targeting EPS metabolism could be useful in preventing cariogenic biofilm formation. This review summarizes plausible strategies targeting S. mutans biofilms by degrading EPS structure, inhibiting EPS production, and disturbing the EPS metabolism-related gene expression and regulatory systems.
Collapse
Affiliation(s)
- Yongwang Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiamin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Khan J, Tarar SM, Gul I, Nawaz U, Arshad M. Challenges of antibiotic resistance biofilms and potential combating strategies: a review. 3 Biotech 2021; 11:169. [PMID: 33816046 DOI: 10.1007/s13205-021-02707-w] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
In this modern era, medicine is facing many alarming challenges. Among different challenges, antibiotics are gaining importance. Recent years have seen unprecedented increase in knowledge and understanding of various factors that are root cause of the spread and development of resistance in microbes against antibiotics. The infection results in the formation of microbial colonies which are termed as biofilms. However, it has been found that a multiple factors contribute in the formation of antimicrobial resistance. Due to higher dose of Minimum Bactericidal Concentration (MBC) as well as of Minimum Inhibitory Concentration (MIC), a large batch of antibiotics available today are of no use as they are ineffective against infections. Therefore, to control infections, there is dire need to adopt alternative treatment for biofilm infection other than antibiotics. This review highlights the latest techniques that are being used to cure the menace of biofilm infections. A wide range of mechanisms has been examined with particular attention towards avenues which can be proved fruitful in the treatment of biofilms. Besides, newer strategies, i.e., matrix centered are also discussed as alternative therapeutic techniques including modulating microbial metabolism, matrix degrading enzyme, photodynamic therapy, natural compounds quorum sensing and nanotechnology which are being used to disrupt extra polymeric substances (EPS) matrix of desired bacterial biofilms.
Collapse
Affiliation(s)
- Javairia Khan
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Sumbal Mudassar Tarar
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Iram Gul
- Department of Earth and Environmental Sciences, Hazara University, Mansehra, Pakistan
| | - Uzam Nawaz
- Department of Statistics, The Women University Multan, Multan, Pakistan
| | - Muhammad Arshad
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology (NUST), Islamabad, Pakistan
| |
Collapse
|
42
|
Jakubovics NS, Goodman SD, Mashburn-Warren L, Stafford GP, Cieplik F. The dental plaque biofilm matrix. Periodontol 2000 2021; 86:32-56. [PMID: 33690911 PMCID: PMC9413593 DOI: 10.1111/prd.12361] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
| | - Steven D Goodman
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Lauren Mashburn-Warren
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Graham P Stafford
- Integrated Biosciences, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Fabian Cieplik
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
43
|
Jara J, Alarcón F, Monnappa AK, Santos JI, Bianco V, Nie P, Ciamarra MP, Canales Á, Dinis L, López-Montero I, Valeriani C, Orgaz B. Self-Adaptation of Pseudomonas fluorescens Biofilms to Hydrodynamic Stress. Front Microbiol 2021; 11:588884. [PMID: 33510716 PMCID: PMC7835673 DOI: 10.3389/fmicb.2020.588884] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/14/2020] [Indexed: 11/26/2022] Open
Abstract
In some conditions, bacteria self-organize into biofilms, supracellular structures made of a self-produced embedding matrix, mainly composed of polysaccharides, DNA, proteins, and lipids. It is known that bacteria change their colony/matrix ratio in the presence of external stimuli such as hydrodynamic stress. However, little is still known about the molecular mechanisms driving this self-adaptation. In this work, we monitor structural features of Pseudomonas fluorescens biofilms grown with and without hydrodynamic stress. Our measurements show that the hydrodynamic stress concomitantly increases the cell density population and the matrix production. At short growth timescales, the matrix mediates a weak cell-cell attractive interaction due to the depletion forces originated by the polymer constituents. Using a population dynamics model, we conclude that hydrodynamic stress causes a faster diffusion of nutrients and a higher incorporation of planktonic bacteria to the already formed microcolonies. This results in the formation of more mechanically stable biofilms due to an increase of the number of crosslinks, as shown by computer simulations. The mechanical stability also relies on a change in the chemical compositions of the matrix, which becomes enriched in carbohydrates, known to display adhering properties. Overall, we demonstrate that bacteria are capable of self-adapting to hostile hydrodynamic stress by tailoring the biofilm chemical composition, thus affecting both the mesoscale structure of the matrix and its viscoelastic properties that ultimately regulate the bacteria-polymer interactions.
Collapse
Affiliation(s)
- Josué Jara
- Departamento de Farmacia Galénica y Tecnología Alimentaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisco Alarcón
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Madrid, Spain.,Departamento de Ingeniería Física, Universidad de Guanajuato, León, Mexico
| | - Ajay K Monnappa
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Valentino Bianco
- Departamento de Química Física, Universidad Complutense de Madrid, Madrid, Spain
| | - Pin Nie
- Nanyang Technological University, Singapore, Singapore
| | | | - Ángeles Canales
- Departamento de Química Orgánica, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Dinis
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Madrid, Spain
| | - Iván López-Montero
- Instituto de Investigación Biomédica Hospital 12 de Octubre (imas12), Madrid, Spain.,Departamento de Química Física, Universidad Complutense de Madrid, Madrid, Spain
| | - Chantal Valeriani
- Departamento de Estructura de la Materia, Física Térmica y Electrónica, Universidad Complutense de Madrid, Madrid, Spain
| | - Belén Orgaz
- Departamento de Farmacia Galénica y Tecnología Alimentaria, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
44
|
Li P, Gao Z, Tan Z, Xiao J, Wei L, Chen Y. New developments in anti-biofilm intervention towards effective management of orthopedic device related infections (ODRI's). BIOFOULING 2021; 37:1-35. [PMID: 33618584 DOI: 10.1080/08927014.2020.1869725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 06/12/2023]
Abstract
Orthopedic device related infections (ODRI's) represent a difficult to treat situation owing to their biofilm based nature. Biofilm infections once established are difficult to eradicate even with an aggressive treatment regimen due to their recalcitrance towards antibiotics and immune attack. The involvement of antibiotic resistant pathogens as the etiological agent further worsens the overall clinical picture, pressing on the need to look into alternative treatment strategies. The present review highlightes the microbiological challenges associated with treatment of ODRI's due to biofilm formation on the implant surface. Further, it details the newer anti-infective modalities that work either by preventing biofilm formation and/or through effective disruption of the mature biofilms formed on the medical implant. The study, therefore aims to provide a comprehensive insight into the newer anti-biofilm interventions (non-antibiotic approaches) and a better understanding of their mechanism of action essential for improved management of orthopedic implant infections.
Collapse
Affiliation(s)
- Ping Li
- Department of Orthopedics, Ya'an People's Hospital, Yaan City, China
| | - Zhenwu Gao
- Department of Orthopedics, Shanxi Bethune Hospital, Taiyuan City, China
| | - Zhenwei Tan
- Department of Orthopedics, Western Theater Air Force Hospital of PLA, Chengdu, China
| | - Jun Xiao
- Department of Orthopedics, Ya'an People's Hospital, Yaan City, China
| | - Li Wei
- Nursing Department, Three Gorges Hospital Affiliated to Chongqing University, Chongqing, China
| | - Yirui Chen
- Department of Orthopedics, Three Gorges Hospital Affiliated to Chongqing University, Chongqing, China
| |
Collapse
|
45
|
|
46
|
Biofilms as Promoters of Bacterial Antibiotic Resistance and Tolerance. Antibiotics (Basel) 2020; 10:antibiotics10010003. [PMID: 33374551 PMCID: PMC7822488 DOI: 10.3390/antibiotics10010003] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/15/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistant bacteria are a global threat for human and animal health. However, they are only part of the problem of antibiotic failure. Another bacterial strategy that contributes to their capacity to withstand antimicrobials is the formation of biofilms. Biofilms are associations of microorganisms embedded a self-produced extracellular matrix. They create particular environments that confer bacterial tolerance and resistance to antibiotics by different mechanisms that depend upon factors such as biofilm composition, architecture, the stage of biofilm development, and growth conditions. The biofilm structure hinders the penetration of antibiotics and may prevent the accumulation of bactericidal concentrations throughout the entire biofilm. In addition, gradients of dispersion of nutrients and oxygen within the biofilm generate different metabolic states of individual cells and favor the development of antibiotic tolerance and bacterial persistence. Furthermore, antimicrobial resistance may develop within biofilms through a variety of mechanisms. The expression of efflux pumps may be induced in various parts of the biofilm and the mutation frequency is induced, while the presence of extracellular DNA and the close contact between cells favor horizontal gene transfer. A deep understanding of the mechanisms by which biofilms cause tolerance/resistance to antibiotics helps to develop novel strategies to fight these infections.
Collapse
|
47
|
Ricardo SIC, Anjos IIL, Monge N, Faustino CMC, Ribeiro IAC. A Glance at Antimicrobial Strategies to Prevent Catheter-Associated Medical Infections. ACS Infect Dis 2020; 6:3109-3130. [PMID: 33245664 DOI: 10.1021/acsinfecdis.0c00526] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Urinary and intravascular catheters are two of the most used invasive medical devices; however, microbial colonization of catheter surfaces is responsible for most healthcare-associated infections (HAIs). Several antimicrobial-coated catheters are available, but recurrent antibiotic therapy can decrease their potential activity against resistant bacterial strains. The aim of this Review is to question the actual effectiveness of currently used (coated) catheters and describe the progress and promise of alternative antimicrobial coatings. Different strategies have been reviewed with the common goal of preventing biofilm formation on catheters, including release-based approaches using antibiotics, antiseptics, nitric oxide, 5-fluorouracil, and silver as well as contact-killing approaches employing quaternary ammonium compounds, chitosan, antimicrobial peptides, and enzymes. All of these strategies have given proof of antimicrobial efficacy by modifying the physiology of pathogens or disrupting their structural integrity. The aim for synergistic approaches using multitarget processes and the combination of both antifouling and bactericidal properties holds potential for the near future. Despite intensive research in biofilm preventive strategies, laboratorial studies still present some limitations since experimental conditions usually are not the same and also differ from biological conditions encountered when the catheter is inserted in the human body. Consequently, in most cases, the efficacy data obtained from in vitro studies is not properly reflected in the clinical setting. Thus, further well-designed clinical trials and additional cytotoxicity studies are needed to prove the efficacy and safety of the developed antimicrobial strategies in the prevention of biofilm formation at catheter surfaces.
Collapse
Affiliation(s)
- Susana I. C. Ricardo
- Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Inês I. L. Anjos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Nuno Monge
- Centro Interdisciplinar de Estudos Educacionais (CIED), Escola Superior de Educação de Lisboa, Instituto Politécnico de Lisboa, Campus de Benfica do IPL, 1549-003 Lisboa, Portugal
| | - Célia M. C. Faustino
- Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Isabel A. C. Ribeiro
- Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Avenida Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| |
Collapse
|
48
|
Baidamshina DR, Koroleva VA, Trizna EY, Pankova SM, Agafonova MN, Chirkova MN, Vasileva OS, Akhmetov N, Shubina VV, Porfiryev AG, Semenova EV, Sachenkov OA, Bogachev MI, Artyukhov VG, Baltina TV, Holyavka MG, Kayumov AR. Anti-biofilm and wound-healing activity of chitosan-immobilized Ficin. Int J Biol Macromol 2020; 164:4205-4217. [DOI: 10.1016/j.ijbiomac.2020.09.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 01/08/2023]
|
49
|
Deciphering Streptococcal Biofilms. Microorganisms 2020; 8:microorganisms8111835. [PMID: 33233415 PMCID: PMC7700319 DOI: 10.3390/microorganisms8111835] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Streptococci are a diverse group of bacteria, which are mostly commensals but also cause a considerable proportion of life-threatening infections. They colonize many different host niches such as the oral cavity, the respiratory, gastrointestinal, and urogenital tract. While these host compartments impose different environmental conditions, many streptococci form biofilms on mucosal membranes facilitating their prolonged survival. In response to environmental conditions or stimuli, bacteria experience profound physiologic and metabolic changes during biofilm formation. While investigating bacterial cells under planktonic and biofilm conditions, various genes have been identified that are important for the initial step of biofilm formation. Expression patterns of these genes during the transition from planktonic to biofilm growth suggest a highly regulated and complex process. Biofilms as a bacterial survival strategy allow evasion of host immunity and protection against antibiotic therapy. However, the exact mechanisms by which biofilm-associated bacteria cause disease are poorly understood. Therefore, advanced molecular techniques are employed to identify gene(s) or protein(s) as targets for the development of antibiofilm therapeutic approaches. We review our current understanding of biofilm formation in different streptococci and how biofilm production may alter virulence-associated characteristics of these species. In addition, we have summarized the role of surface proteins especially pili proteins in biofilm formation. This review will provide an overview of strategies which may be exploited for developing novel approaches against biofilm-related streptococcal infections.
Collapse
|
50
|
Van der Gucht M, Aktan MK, Hendrix H, Velde GV, Paeshuyse J, Braem A, Lavigne R. qDNase assay: A quantitative method for real-time assessment of DNase activity on coated surfaces. Biochem Biophys Res Commun 2020; 534:1003-1006. [PMID: 33121680 DOI: 10.1016/j.bbrc.2020.10.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 10/17/2020] [Indexed: 11/17/2022]
Abstract
DNase coatings show great potential to prevent biofilm formation in various applications of the medical implant, food and marine industry. However, straightforward and quantitative methods to characterize the enzymatic activity of these coatings are currently not available. We here introduce the qDNase assay, a quantitative, real-time method to characterize the activity of DNase coatings. The assay combines (1) the use of an oligonucleotide probe, which fluoresces upon cleavage by coated DNases, and (2) the continuous read-out of the fluorescent signal within a microplate fluorometer format. The combination of these two properties results in a real-time fluorescent signal that is used to directly quantify the activity of DNase coatings. As a proof of concept, bovine DNase I coatings were immobilized on titanium by means of chemical grafting and their activity was estimated at 3.87 × 10-4 U. To our knowledge, the qDNase assay provides the first approach to report the activity of a DNase coating in absolute DNase activity units. This assay will not only serve to compare existing DNase coating methods more accurately, but will also enable the rational design of new DNase coating methods in the future.
Collapse
Affiliation(s)
- Marie Van der Gucht
- KU Leuven, Department of Biosystems, Laboratory of Gene Technology, Kasteelpark Arenberg 21 - box 2462, B-3001, Leuven, Belgium
| | - Merve Kübra Aktan
- KU Leuven, Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering Research Group, Kasteelpark Arenberg 44 - box 2450, B-3001, Leuven, Belgium
| | - Hanne Hendrix
- KU Leuven, Department of Biosystems, Laboratory of Gene Technology, Kasteelpark Arenberg 21 - box 2462, B-3001, Leuven, Belgium
| | - Greetje Vande Velde
- KU Leuven, Department of Imaging & Pathology, Biomedical MRI / MoSAIC, Herestraat 49, building O&N1 - box 505, B-3000, Leuven, Belgium
| | - Jan Paeshuyse
- KU Leuven, Department of Biosystems, Laboratory of Host Pathogen Interaction in Livestock, Kasteelpark Arenberg 30 - box 2472, B-3001, Leuven, Belgium
| | - Annabel Braem
- KU Leuven, Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering Research Group, Kasteelpark Arenberg 44 - box 2450, B-3001, Leuven, Belgium
| | - Rob Lavigne
- KU Leuven, Department of Biosystems, Laboratory of Gene Technology, Kasteelpark Arenberg 21 - box 2462, B-3001, Leuven, Belgium.
| |
Collapse
|