1
|
Allahyari H, Shamsini L, Zamani H. Dual encapsulation of curcumin and ciprofloxacin in chitosan nanoparticles attenuates Pseudomonas aeruginosa virulence, elastinolytic potential and quorum sensing genes. Microb Pathog 2025; 202:107438. [PMID: 40032002 DOI: 10.1016/j.micpath.2025.107438] [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: 10/28/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Pseudomonas aeruginosa is an important human pathogen that is responsible for various human infections and able to develop resistance to a variety of antibiotics. Drug encapsulation may provide sustained and more efficient drug delivery, particularly in case of the drugs with low bioavailability. This study aims to characterize the antivirulence and anti-quorum sensing (QS) properties of curcumin and ciprofloxacin dually encapsulated in chitosan NPs (Cur-Cip-CsNPs). The nanoparticles were synthesized and characterized by SEM, FT-IR, Zeta Potential, and DLS analyses. The antibacterial and antivirulence effects of the Cip-CsNPs, Cur-CsNPs, and Cur-Cip-CsNPs against P. aeruginosa strains were investigated by well diffusion, biofilm and pyocyanin quantification, swarming, swimming, twitching, and proteolytic and elastinolytic activity assays. The mRNA transcript levels of the lasIR and lasAB genes were also determined by real-time PCR. Cur-Cip-CsNPs were more potent antibacterial agents against P. aeruginosa compared with other NPs and inhibited bacterial planktonic growth at 160 mg/mL, reduced biofilm formation by 72.5-86.5 % and pyocyanin levels by 80.2-80.6 %, and significantly inhibited flagellar and fimbrial motility of P. aeruginosa. Furthermore, bacterial proteolysis and elastinolytic activity were reduced more efficiently by Cur-Cip-CsNPs compared with other nanoformulations. The expression of the lasI, lasR, lasA, and lasB was attenuated more efficiently by Cur-Cip-CsNPs compared with Cip-CsNPs and Cur-CsNPs. This study presents an innovative approach to overcome the challenges due to antibiotic resistance and provides a new therapeutic option against P. aeruginosa infections.
Collapse
Affiliation(s)
| | - Leila Shamsini
- Department of Biology, University of Guilan, Rasht, Iran
| | | |
Collapse
|
2
|
Almatroudi A. Biofilm Resilience: Molecular Mechanisms Driving Antibiotic Resistance in Clinical Contexts. BIOLOGY 2025; 14:165. [PMID: 40001933 PMCID: PMC11852148 DOI: 10.3390/biology14020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025]
Abstract
Healthcare-associated infections pose a significant global health challenge, negatively impacting patient outcomes and burdening healthcare systems. A major contributing factor to healthcare-associated infections is the formation of biofilms, structured microbial communities encased in a self-produced extracellular polymeric substance matrix. Biofilms are critical in disease etiology and antibiotic resistance, complicating treatment and infection control efforts. Their inherent resistance mechanisms enable them to withstand antibiotic therapies, leading to recurrent infections and increased morbidity. This review explores the development of biofilms and their dual roles in health and disease. It highlights the structural and protective functions of the EPS matrix, which shields microbial populations from immune responses and antimicrobial agents. Key molecular mechanisms of biofilm resistance, including restricted antibiotic penetration, persister cell dormancy, and genetic adaptations, are identified as significant barriers to effective management. Biofilms are implicated in various clinical contexts, including chronic wounds, medical device-associated infections, oral health complications, and surgical site infections. Their prevalence in hospital environments exacerbates infection control challenges and underscores the urgent need for innovative preventive and therapeutic strategies. This review evaluates cutting-edge approaches such as DNase-mediated biofilm disruption, RNAIII-inhibiting peptides, DNABII proteins, bacteriophage therapies, antimicrobial peptides, nanoparticle-based solutions, antimicrobial coatings, and antimicrobial lock therapies. It also examines critical challenges associated with biofilm-related healthcare-associated infections, including diagnostic difficulties, disinfectant resistance, and economic implications. This review emphasizes the need for a multidisciplinary approach and underscores the importance of understanding biofilm dynamics, their role in disease pathogenesis, and the advancements in therapeutic strategies to combat biofilm-associated infections effectively in clinical settings. These insights aim to enhance treatment outcomes and reduce the burden of biofilm-related diseases.
Collapse
Affiliation(s)
- Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
3
|
Francolino R, Martino M, Nazzaro F, Sirignano C, Fratianni F, Coppola F, De Martino L, Formisano C, De Feo V. Chemical Profile and Bioactivities of Three Species of Mentha Growing in the Campania Region, Southern Italy. PLANTS (BASEL, SWITZERLAND) 2025; 14:360. [PMID: 39942924 PMCID: PMC11821235 DOI: 10.3390/plants14030360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025]
Abstract
The genus Mentha (Lamiaceae), comprising aromatic perennial plants widely distributed in temperate regions, holds significant medicinal and commercial value. This study aimed to investigate the chemical profile and bioactivities of hydroalcoholic extracts from Mentha longifolia (L.) L., Mentha pulegium L., and Mentha spicata L. harvested from the Campania region, Southern Italy. Chemical analysis using LC-HRESIMS/MS identified a total of 21 compounds. The extracts, particularly M. pulegium, exhibited notable antioxidant activity, evaluated through DPPH and FRAP assays, probably related to their chemical composition. Both M. pulegium and M. longifolia demonstrated a higher phenolic content, with M. pulegium also containing the highest levels of flavonoids. In addition, the extract's ability to inhibit biofilm formation was evaluated against several pathogenic strains, including Gram-positive bacteria (Listeria monocytogenes and Staphylococcus aureus) and Gram-negative bacteria (Acinetobacter baumannii, Pseudomonas aeruginosa, and Escherichia coli) using crystal violet and MTT assays. All extracts effectively inhibited biofilm formation in A. baumannii and P. aeruginosa, with M. pulegium also showing moderate activity against the metabolism of L. monocytogenes. The pronounced antibacterial and biofilm-inhibitory properties of M. pulegium highlight its potential for pharmaceutical applications.
Collapse
Affiliation(s)
- Rosaria Francolino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (R.F.); (M.M.); (V.D.F.)
| | - Mara Martino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (R.F.); (M.M.); (V.D.F.)
| | - Filomena Nazzaro
- Institute of Food Sciences, CNR, 83100 Avellino, Italy; (F.N.); (F.F.); (F.C.)
| | - Carmina Sirignano
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, 80131 Napoli, Italy;
| | - Florinda Fratianni
- Institute of Food Sciences, CNR, 83100 Avellino, Italy; (F.N.); (F.F.); (F.C.)
| | - Francesca Coppola
- Institute of Food Sciences, CNR, 83100 Avellino, Italy; (F.N.); (F.F.); (F.C.)
| | - Laura De Martino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (R.F.); (M.M.); (V.D.F.)
| | - Carmen Formisano
- Department of Pharmacy, School of Medicine and Surgery, University of Napoli Federico II, 80131 Napoli, Italy;
| | - Vincenzo De Feo
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy; (R.F.); (M.M.); (V.D.F.)
- Institute of Food Sciences, CNR, 83100 Avellino, Italy; (F.N.); (F.F.); (F.C.)
| |
Collapse
|
4
|
Grilc NK, Kristl J, Zupančič Š. Can polymeric nanofibers effectively preserve and deliver live therapeutic bacteria? Colloids Surf B Biointerfaces 2025; 245:114329. [PMID: 39486375 DOI: 10.1016/j.colsurfb.2024.114329] [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: 08/05/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Probiotics and live therapeutic bacteria (LTB), their strictly regulated therapeutic counterpart, are increasingly important in treating and preventing biofilm-related diseases. This necessitates new approaches to (i) preserve bacterial viability during manufacturing and storage and (ii) incorporate LTB into delivery systems for enhanced therapeutic efficacy. This review explores advances in probiotic and LTB product development, focusing on preservation, protection, and improved delivery. Preservation of bacteria can be achieved by drying methods that decelerate metabolism. These methods introduce stresses affecting viability which can be mitigated with suitable excipients like polymeric or low molecular weight stabilizers. The review emphasizes the incorporation of LTB into polymer-based nanofibers via electrospinning, enabling simultaneous drying, encapsulation, and delivery system production. Optimization of bacterial survival during electrospinning and storage is discussed, as well as controlled LTB release achievable through formulation design using gel-forming, gastroprotective, mucoadhesive, and pH-responsive polymers. Evaluation of the presence of the actual therapeutic strains, bacterial viability and activity by CFU enumeration or alternative analytical techniques is presented as a key aspect of developing effective and safe formulations with LTB. This review offers insights into designing delivery systems, especially polymeric nanofibers, for preservation and delivery of LTB, guiding readers in developing innovative biotherapeutic delivery systems.
Collapse
Affiliation(s)
- Nina Katarina Grilc
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Julijana Kristl
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Špela Zupančič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia.
| |
Collapse
|
5
|
Monteiro R, Silva E, Pereira MO, Sousa AM. Mechanistic Insights into Succinic Acid as an Adjuvant for Ciprofloxacin in Treating Pseudomonas aeruginosa Growing Within Cystic Fibrosis Airway Mucus. Microorganisms 2024; 12:2538. [PMID: 39770741 PMCID: PMC11678660 DOI: 10.3390/microorganisms12122538] [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: 10/31/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Pseudomonas aeruginosa is a major cause of chronic respiratory infections in patients with cystic fibrosis (CF), with biofilm formation contributing to its persistence and antibiotic resistance. This study aimed to gain insights into the mechanistic action of succinic acid as a ciprofloxacin adjuvant against clinically relevant CF isolates, including small colony variants and mucoid strains, and a ciprofloxacin-resistant strain grown within CF dense mucus. Time-kill assays in artificial CF mucus, along with planktonic and surface-attached biofilm experiments, were used to assess the activity of succinic acid alone and in combination with sublethal ciprofloxacin concentrations. Succinic acid demonstrated an adjuvant effect of ciprofloxacin against P. aeruginosa grown within CF mucus at pH levels below pKa1 during the early bacterial growth stages. In examining planktonic growth and biofilms under these conditions, we found that succinic acid demonstrated strong antibacterial and antibiofilm properties. Conversely, succinic acid activity decreased at later growth stages, though it enhanced the ciprofloxacin effect, especially against mucoid biofilms. Moreover, we noted that, in dense CF mucus, succinic acid activity was attenuated compared to a non-CF environment, indicating diffusion challenges. These findings underscore the potential of succinic acid as a therapeutic adjuvant for improving antibiotic treatment outcomes and overcoming biofilm-associated resistance in CF.
Collapse
Affiliation(s)
- Rosana Monteiro
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
| | - Eduarda Silva
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
| | - Maria Olivia Pereira
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Margarida Sousa
- Centre of Biological Engineering, LIBRO—Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (R.M.); (E.S.); (M.O.P.)
- LABBELS—Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
6
|
Meirelles LA, Vayena E, Debache A, Schmidt E, Rossy T, Distler T, Hatzimanikatis V, Persat A. Pseudomonas aeruginosa faces a fitness trade-off between mucosal colonization and antibiotic tolerance during airway infection. Nat Microbiol 2024; 9:3284-3303. [PMID: 39455898 DOI: 10.1038/s41564-024-01842-3] [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: 01/16/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024]
Abstract
Pseudomonas aeruginosa frequently causes antibiotic-recalcitrant pneumonia, but the mechanisms driving its adaptation during human infections remain unclear. To reveal the selective pressures and adaptation strategies at the mucosal surface, here we investigated P. aeruginosa growth and antibiotic tolerance in tissue-engineered airways by transposon insertion sequencing (Tn-seq). Metabolic modelling based on Tn-seq data revealed the nutritional requirements for P. aeruginosa growth, highlighting reliance on glucose and lactate and varying requirements for amino acid biosynthesis. Tn-seq also revealed selection against biofilm formation during mucosal growth in the absence of antibiotics. Live imaging in engineered organoids showed that biofilm-dwelling cells remained sessile while colonizing the mucosal surface, limiting nutrient foraging and reduced growth. Conversely, biofilm formation increased antibiotic tolerance at the mucosal surface. Moreover, mutants with exacerbated biofilm phenotypes protected less tolerant but more cytotoxic strains, contributing to phenotypic heterogeneity. P. aeruginosa must therefore navigate conflicting physical and biological selective pressures to establish chronic infections.
Collapse
Affiliation(s)
- Lucas A Meirelles
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Evangelia Vayena
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Auriane Debache
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eric Schmidt
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tamara Rossy
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tania Distler
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Vassily Hatzimanikatis
- Laboratory of Computational Systems Biotechnology, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Alexandre Persat
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
7
|
Stewart PS, Kim J, James G, Yi F, Stechmiller J, Weaver M, Kelly DL, Fisher S, Schultz G, Lyon D. Association of biofilm and microbial metrics with healing rate in older adults with chronic venous leg ulcers. Wound Repair Regen 2024; 32:858-871. [PMID: 39425525 PMCID: PMC11585430 DOI: 10.1111/wrr.13230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
The presence of microbial biofilms in many human chronic wounds led to the hypothesis that biofilms delay healing of these wounds. We tested this hypothesis in a population of 117 older individuals with venous leg ulcers who were receiving standardised therapy, including frequent debridement. Debridement specimens were analysed for the amount of bacterial biomass by two independent methods: a microscopic approach that scored the relative size and number of bacterial aggregates, interpreted as a biofilm metric, and conventional enumeration by agar plating for viable bacteria. The plating protocol yielded three distinct values: the total viable bacterial count, bleach-tolerant bacteria, and the log reduction in viable bacteria upon bleach treatment. Wound healing rates over an 8-week observation period were calculated as the rate of decrease of the equivalent diameter of the wound. There was no statistically significant association between wound healing and the biofilm metric in any of the three analyses performed (p ≥0.15). In all three statistical tests, wound healing was associated with the log reduction caused by bleach treatment (p ≤0.004); wounds that harboured bacteria that were more bleach-susceptible healed more slowly. A refinement of the model of chronic wound infection pathogenesis is proposed in which dormant bacteria constitute a persistent nidus and outgrowth of metabolically active cells impairs healing. This model constitutes a new hypothesis as metabolic activity was not directly measured in this investigation.
Collapse
Affiliation(s)
- Philip S. Stewart
- Center for Biofilm Engineering, Montana State University
- Department of Chemical and Biological Engineering, Montana State University
| | - Junglyun Kim
- Chungnam National University College of Nursing
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Garth James
- Center for Biofilm Engineering, Montana State University
- Department of Chemical and Biological Engineering, Montana State University
| | - Fan Yi
- Department of Mathematics and Statistical Science, University of Idaho
| | - Joyce Stechmiller
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Michael Weaver
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Debra L. Kelly
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Steve Fisher
- Center for Biofilm Engineering, Montana State University
| | | | - Debra Lyon
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| |
Collapse
|
8
|
Nickerson CA, McLean RJC, Barrila J, Yang J, Thornhill SG, Banken LL, Porterfield DM, Poste G, Pellis NR, Ott CM. Microbiology of human spaceflight: microbial responses to mechanical forces that impact health and habitat sustainability. Microbiol Mol Biol Rev 2024; 88:e0014423. [PMID: 39158275 PMCID: PMC11426028 DOI: 10.1128/mmbr.00144-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
SUMMARYUnderstanding the dynamic adaptive plasticity of microorganisms has been advanced by studying their responses to extreme environments. Spaceflight research platforms provide a unique opportunity to study microbial characteristics in new extreme adaptational modes, including sustained exposure to reduced forces of gravity and associated low fluid shear force conditions. Under these conditions, unexpected microbial responses occur, including alterations in virulence, antibiotic and stress resistance, biofilm formation, metabolism, motility, and gene expression, which are not observed using conventional experimental approaches. Here, we review biological and physical mechanisms that regulate microbial responses to spaceflight and spaceflight analog environments from both the microbe and host-microbe perspective that are relevant to human health and habitat sustainability. We highlight instrumentation and technology used in spaceflight microbiology experiments, their limitations, and advances necessary to enable next-generation research. As spaceflight experiments are relatively rare, we discuss ground-based analogs that mimic aspects of microbial responses to reduced gravity in spaceflight, including those that reduce mechanical forces of fluid flow over cell surfaces which also simulate conditions encountered by microorganisms during their terrestrial lifecycles. As spaceflight mission durations increase with traditional astronauts and commercial space programs send civilian crews with underlying health conditions, microorganisms will continue to play increasingly critical roles in health and habitat sustainability, thus defining a new dimension of occupational health. The ability of microorganisms to adapt, survive, and evolve in the spaceflight environment is important for future human space endeavors and provides opportunities for innovative biological and technological advances to benefit life on Earth.
Collapse
Affiliation(s)
- Cheryl A. Nickerson
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | | | - Jennifer Barrila
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | - Jiseon Yang
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | | | - Laura L. Banken
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, Arizona, USA
| | - D. Marshall Porterfield
- Department of Agricultural & Biological Engineering, Purdue University, West Lafayette, Indiana, USA
| | - George Poste
- Complex Adaptive Systems Initiative, Arizona State University, Tempe, Arizona, USA
| | | | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, Texas, USA
| |
Collapse
|
9
|
Iungin O, Shydlovska O, Moshynets O, Vasylenko V, Sidorenko M, Mickevičius S, Potters G. Metal-based nanoparticles: an alternative treatment for biofilm infection in hard-to-heal wounds. J Wound Care 2024; 33:xcix-cx. [PMID: 38588056 DOI: 10.12968/jowc.2024.33.sup4a.xcix] [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] [Indexed: 04/10/2024]
Abstract
Metal-based nanoparticles (MNPs) are promoted as effective compounds in the treatment of bacterial infections and as possible alternatives to antibiotics. These MNPs are known to affect a broad spectrum of microorganisms using a multitude of strategies, including the induction of reactive oxygen species and interaction with the inner structures of the bacterial cells. The aim of this review was to summarise the latest studies about the effect of metal-based nanoparticles on pathogenic bacterial biofilm formed in wounds, using the examples of Gram-positive bacterium Staphylococcus aureus and Gram-negative bacterium Pseudomonas aeruginosa, as well as provide an overview of possible clinical applications.
Collapse
Affiliation(s)
- Olga Iungin
- 1 Kyiv National University of Technologies and Design (KNUTD), Kyiv, Ukraine
- 2 Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Olga Shydlovska
- 1 Kyiv National University of Technologies and Design (KNUTD), Kyiv, Ukraine
| | - Olena Moshynets
- 2 Institute of Molecular Biology and Genetics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Volodymyr Vasylenko
- 3 Vytautas Magnus University, Faculty of Natural Science, Akademija, Lithuania
| | - Marina Sidorenko
- 3 Vytautas Magnus University, Faculty of Natural Science, Akademija, Lithuania
| | - Saulius Mickevičius
- 3 Vytautas Magnus University, Faculty of Natural Science, Akademija, Lithuania
| | - Geert Potters
- 4 Antwerp Maritime Academy, Antwerp, Belgium
- 5 University of Antwerp, Antwerp, Belgium
| |
Collapse
|
10
|
Li J, Yu Y, Zhou Y, Song J, Yang A, Wang M, Li Y, Wan M, Zhang C, Yang H, Bai Y, Wong WL, Pu H, Feng X. Multi-targeting oligopyridiniums: Rational design for biofilm dispersion and bacterial persister eradication. Bioorg Chem 2024; 144:107163. [PMID: 38306825 DOI: 10.1016/j.bioorg.2024.107163] [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: 10/19/2023] [Revised: 01/09/2024] [Accepted: 01/27/2024] [Indexed: 02/04/2024]
Abstract
The development of effective antibacterial drugs to combat bacterial infections, particularly the biofilm-related infections, remains a challenge. There are two important features of bacterial biofilms, which are well-known critical factors causing biofilms hard-to-treat in clinical, including the dense and impermeable extracellular polymeric substances (EPS) and the metabolically repressed dormant and persistent bacterial population embedded. These characteristics largely increase the difficulty for regular antibiotic treatment due to insufficient penetration into EPS. In addition, the dormant bacteria are insensitive to the growth-inhibiting mechanism of traditional antibiotics. Herein, we explore the potential of a series of new oligopyridinium-based oligomers bearing a multi-biomacromolecule targeting function as the potent bacterial biofilm eradication agent. These oligomers were rationally designed to be "charge-on-backbone" that can offer a special alternating amphiphilicity. This novel and unique feature endows high affinity to bacterial membrane lipids, DNAs as well as proteins. Such a broad multi-targeting nature of molecules not only enables its penetration into EPS, but also plays vital roles in the bactericidal mechanism of action that is highly effective against dormant and persistent bacteria. Our in vitro, ex vivo, and in vivo studies demonstrated that OPc3, one of the most effective derivatives, was able to offer excellent antibacterial potency against a variety of bacteria and effectively eliminate biofilms in zebrafish models and mouse wound biofilm infection models.
Collapse
Affiliation(s)
- Jiaqi Li
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yue Yu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Yu Zhou
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Junfeng Song
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Anming Yang
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Min Wang
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Youzhi Li
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China
| | - Muyang Wan
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Chunhui Zhang
- College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Huan Yang
- Xuzhou Key Laboratory of Laboratory Diagnostics, School of Medical Technology, Xuzhou Medical University, Xuzhou 221004, China.
| | - Yugang Bai
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| | - Wing-Leung Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon 999077, Hong Kong, China Hong Kong Special Administrative Region.
| | - Huangsheng Pu
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China; College of Biology, Hunan University, Changsha, Hunan 410082, China; College of Advanced Interdisciplinary Studies & Hunan Provincial Key Laboratory of Novel Nano Optoelectronic Information Materials and Devices, National University of Defense Technology, Changsha, Hunan 410073, China; Nanhu Laser Laboratory, National University of Defense Technology, Changsha 410073, China.
| | - Xinxin Feng
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, and School of Chemistry and Chemical Engineering, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
11
|
Sousa AM, Ferreira D, Rodrigues LR, Pereira MO. Aptamer-based therapy for fighting biofilm-associated infections. J Control Release 2024; 367:522-539. [PMID: 38295992 DOI: 10.1016/j.jconrel.2024.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/06/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
Biofilms are key players in the pathogenesis of most of chronic infections associated with host tissue or fluids and indwelling medical devices. These chronic infections are hard to be treated due to the increased biofilms tolerance towards antibiotics in comparison to planktonic (or free living) cells. Despite the advanced understanding of their formation and physiology, biofilms continue to be a challenge and there is no standardized therapeutic approach in clinical practice to eradicate them. Aptamers offer distinctive properties, including excellent affinity, selectivity, stability, making them valuable tools for therapeutic purposes. This review explores the flexibility and designability of aptamers as antibiofilm drugs but, importantly, as targeting tools for diverse drug and delivery systems. It highlights specific examples of application of aptamers in biofilms of diverse species according to different modes of action including inhibition of motility and adhesion, blocking of quorum sensing molecules, and dispersal of biofilm-cells to planktonic state. Moreover, it discusses the limitations and challenges that impaired an increased success of the use of aptamers on biofilm management, as well as the opportunities related to aptamers modifications that can significantly expand their applicability on the biofilm field.
Collapse
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| | - Débora Ferreira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Lígia Raquel Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
12
|
Neveu M, Quinn R, Barge LM, Craft KL, German CR, Getty S, Glein C, Parra M, Burton AS, Cary F, Corpolongo A, Fifer L, Gangidine A, Gentry D, Georgiou CD, Haddadin Z, Herbold C, Inaba A, Jordan SF, Kalucha H, Klier P, Knicely K, Li AY, McNally P, Millan M, Naz N, Raj CG, Schroedl P, Timm J, Yang Z. Future of the Search for Life: Workshop Report. ASTROBIOLOGY 2024; 24:114-129. [PMID: 38227837 DOI: 10.1089/ast.2022.0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
The 2-week, virtual Future of the Search for Life science and engineering workshop brought together more than 100 scientists, engineers, and technologists in March and April 2022 to provide their expert opinion on the interconnections between life-detection science and technology. Participants identified the advances in measurement and sampling technologies they believed to be necessary to perform in situ searches for life elsewhere in our Solar System, 20 years or more in the future. Among suggested measurements for these searches, those pertaining to three potential indicators of life termed "dynamic disequilibrium," "catalysis," and "informational polymers" were identified as particularly promising avenues for further exploration. For these three indicators, small breakout groups of participants identified measurement needs and knowledge gaps, along with corresponding constraints on sample handling (acquisition and processing) approaches for a variety of environments on Enceladus, Europa, Mars, and Titan. Despite the diversity of these environments, sample processing approaches all tend to be more complex than those that have been implemented on missions or envisioned for mission concepts to date. The approaches considered by workshop breakout groups progress from nondestructive to destructive measurement techniques, and most involve the need for fluid (especially liquid) sample processing. Sample processing needs were identified as technology gaps. These gaps include technology and associated sampling strategies that allow the preservation of the thermal, mechanical, and chemical integrity of the samples upon acquisition; and to optimize the sample information obtained by operating suites of instruments on common samples. Crucially, the interplay between science-driven life-detection strategies and their technological implementation highlights the need for an unprecedented level of payload integration and extensive collaboration between scientists and engineers, starting from concept formulation through mission deployment of life-detection instruments and sample processing systems.
Collapse
Affiliation(s)
- Marc Neveu
- Department of Astronomy, University of Maryland, College Park, Maryland, USA
- NASA Goddard Space Flight Center, Greenbelt, Maryland, USA
| | - Richard Quinn
- NASA Ames Research Center, Moffett Field, California, USA
| | - Laura M Barge
- NASA Jet Propulsion Laboratory, California Institute of Technology, Pasadena, California, USA
| | - Kathleen L Craft
- Applied Physics Laboratory, Johns Hopkins University, Laurel, Maryland, USA
| | | | | | | | - Macarena Parra
- NASA Ames Research Center, Moffett Field, California, USA
| | | | - Francesca Cary
- Hawai'i Institute of Geophysics and Planetology, University of Hawai'i, Mānoa, Hawaii, USA
| | - Andrea Corpolongo
- Department of Geosciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lucas Fifer
- Department of Earth and Space Sciences, University of Washington, Seattle, Washington, USA
| | - Andrew Gangidine
- Office of Development, Yale University, New Haven, Connecticut, USA
| | - Diana Gentry
- NASA Ames Research Center, Moffett Field, California, USA
| | | | - Zaid Haddadin
- Department of Electrical and Computer Engineering, University of California, San Diego, California, USA
| | - Craig Herbold
- School of Biological Sciences, University of Canterbury, Christchurch, New Zealand
| | - Aila Inaba
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
| | - Seán F Jordan
- School of Chemical Sciences, Dublin City University, Dublin, Ireland
| | - Hemani Kalucha
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, California, USA
| | - Pavel Klier
- NASA Ames Research Center, Moffett Field, California, USA
- NASA Postdoctoral Program, Oak Ridge Associated Universities, Oak Ridge, Tennessee, USA
| | - Kas Knicely
- Geophysical Institute, University of Alaska, Fairbanks, Alaska, USA
| | - An Y Li
- Department of Earth and Space Sciences, University of Washington, Seattle, Washington, USA
| | - Patrick McNally
- Space Physics Research Laboratory, University of Michigan, Ann Arbor, Michigan, USA
| | - Maëva Millan
- Laboratory Atmosphere and Space Observations, Guyancourt, France
| | - Neveda Naz
- Department of Chemistry, Tufts University, Medford, Massachusetts, USA
| | - Chinmayee Govinda Raj
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Peter Schroedl
- Department of Biology, Boston University, Boston, Massachusetts, USA
| | - Jennifer Timm
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, New Jersey, USA
| | - Ziming Yang
- Department of Chemistry, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
13
|
Lazarus E, Meyer AS, Ikuma K, Rivero IV. Three dimensional printed biofilms: Fabrication, design and future biomedical and environmental applications. Microb Biotechnol 2024; 17:e14360. [PMID: 38041693 PMCID: PMC10832517 DOI: 10.1111/1751-7915.14360] [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: 07/01/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 12/03/2023] Open
Abstract
Three dimensional printing has emerged as a widely acceptable strategy for the fabrication of mammalian cell laden constructs with complex microenvironments for tissue engineering and regenerative medicine. More recently 3D printed living materials containing microorganisms have been developed and matured into living biofilms. The potential for engineered 3D biofilms as in vitro models for biomedical applications, such as antimicrobial susceptibility testing, and environmental applications, such as bioleaching, bioremediation, and wastewater purification, is extensive but the need for an in-depth understanding of the structure-function relationship between the complex construct and the microorganism response still exists. This review discusses 3D printing fabrication methods for engineered biofilms with specific structural features. Next, it highlights the importance of bioink compositions and 3D bioarchitecture design. Finally, a brief overview of current and potential applications of 3D printed biofilms in environmental and biomedical fields is discussed.
Collapse
Affiliation(s)
- Emily Lazarus
- Department Industrial and Systems EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
| | - Anne S. Meyer
- Department of BiologyUniversity of RochesterRochesterNew YorkUSA
| | - Kaoru Ikuma
- Department of Civil, Construction, and Environmental EngineeringIowa State UniversityAmesIowaUSA
| | - Iris V. Rivero
- Department Industrial and Systems EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
- Department of Biomedical EngineeringRochester Institute of TechnologyRochesterNew YorkUSA
- Department of Industrial and Systems EngineeringUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
14
|
Coenye T. Biofilm antimicrobial susceptibility testing: where are we and where could we be going? Clin Microbiol Rev 2023; 36:e0002423. [PMID: 37812003 PMCID: PMC10732061 DOI: 10.1128/cmr.00024-23] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/27/2023] [Indexed: 10/10/2023] Open
Abstract
Our knowledge about the fundamental aspects of biofilm biology, including the mechanisms behind the reduced antimicrobial susceptibility of biofilms, has increased drastically over the last decades. However, this knowledge has so far not been translated into major changes in clinical practice. While the biofilm concept is increasingly on the radar of clinical microbiologists, physicians, and healthcare professionals in general, the standardized tools to study biofilms in the clinical microbiology laboratory are still lacking; one area in which this is particularly obvious is that of antimicrobial susceptibility testing (AST). It is generally accepted that the biofilm lifestyle has a tremendous impact on antibiotic susceptibility, yet AST is typically still carried out with planktonic cells. On top of that, the microenvironment at the site of infection is an important driver for microbial physiology and hence susceptibility; but this is poorly reflected in current AST methods. The goal of this review is to provide an overview of the state of the art concerning biofilm AST and highlight the knowledge gaps in this area. Subsequently, potential ways to improve biofilm-based AST will be discussed. Finally, bottlenecks currently preventing the use of biofilm AST in clinical practice, as well as the steps needed to get past these bottlenecks, will be discussed.
Collapse
Affiliation(s)
- Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
15
|
Prince J, Jones AAD. Heterogenous biofilm mass-transport model replicates periphery sequestration of antibiotics in Pseudomonas aeruginosa PAO1 microcolonies. Proc Natl Acad Sci U S A 2023; 120:e2312995120. [PMID: 37956290 PMCID: PMC10666002 DOI: 10.1073/pnas.2312995120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/04/2023] [Indexed: 11/15/2023] Open
Abstract
A model for antibiotic accumulation in bacterial biofilm microcolonies utilizing heterogenous porosity and attachment site profiles replicated the periphery sequestration reported in prior experimental studies on Pseudomonas aeruginosa PAO1 biofilm cell clusters. These P. aeruginosa cell clusters are in vitro models of the chronic P. aeruginosa infections in cystic fibrosis patients which display recalcitrance to antibiotic treatments, leading to exacerbated morbidity and mortality. This resistance has been partially attributed to periphery sequestration, where antibiotics fail to penetrate biofilm cell clusters. The physical phenomena driving this periphery sequestration have not been definitively established. This paper introduces mathematical models to account for two proposed physical phenomena driving periphery sequestration: biofilm matrix attachment and volume-exclusion due to variable biofilm porosity. An antibiotic accumulation model which incorporated these phenomena better fit observed periphery sequestration data compared to previous models.
Collapse
Affiliation(s)
- Joshua Prince
- Department of Civil and Environmental Engineering, Duke University, Durham, NC27708
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC27708
| | - A-Andrew D. Jones
- Department of Civil and Environmental Engineering, Duke University, Durham, NC27708
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC27708
| |
Collapse
|
16
|
Lichtenberg M, Coenye T, Parsek MR, Bjarnsholt T, Jakobsen TH. What's in a name? Characteristics of clinical biofilms. FEMS Microbiol Rev 2023; 47:fuad050. [PMID: 37656883 PMCID: PMC10503651 DOI: 10.1093/femsre/fuad050] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/06/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023] Open
Abstract
In vitro biofilms are communities of microbes with unique features compared to individual cells. Biofilms are commonly characterized by physical traits like size, adhesion, and a matrix made of extracellular substances. They display distinct phenotypic features, such as metabolic activity and antibiotic tolerance. However, the relative importance of these traits depends on the environment and bacterial species. Various mechanisms enable biofilm-associated bacteria to withstand antibiotics, including physical barriers, physiological adaptations, and changes in gene expression. Gene expression profiles in biofilms differ from individual cells but, there is little consensus among studies and so far, a 'biofilm signature transcriptome' has not been recognized. Additionally, the spatial and temporal variability within biofilms varies greatly depending on the system or environment. Despite all these variable conditions, which produce very diverse structures, they are all noted as biofilms. We discuss that clinical biofilms may differ from those grown in laboratories and found in the environment and discuss whether the characteristics that are commonly used to define and characterize biofilms have been shown in infectious biofilms. We emphasize that there is a need for a comprehensive understanding of the specific traits that are used to define bacteria in infections as clinical biofilms.
Collapse
Affiliation(s)
- Mads Lichtenberg
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Matthew R Parsek
- Department of Microbiology, University of Washington School of Medicine, 1705 NE Pacific St., WA 98195 Seattle, United States
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Ole Maaløes vej 26, 2100 Copenhagen, Denmark
| | - Tim Holm Jakobsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| |
Collapse
|
17
|
Stewart PS, Owkes M. Simulation of catalase-dependent tolerance of microbial biofilm to hydrogen peroxide with a biofilm computer model. NPJ Biofilms Microbiomes 2023; 9:60. [PMID: 37612330 PMCID: PMC10447567 DOI: 10.1038/s41522-023-00426-z] [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: 05/03/2023] [Accepted: 08/07/2023] [Indexed: 08/25/2023] Open
Abstract
Hydrogen peroxide (HP) is a common disinfectant and antiseptic. When applied to a biofilm, it may be expected that the top layer of the biofilm would be killed by HP, the HP would penetrate further, and eventually eradicate the entire biofilm. However, using the Biofilm.jl computer model, we demonstrate a mechanism by which the biofilm can persist, and even become thicker, in the indefinite treatment with an HP solution at concentrations that are lethal to planktonic microorganisms. This surprising result is found to be dependent on the neutralization of HP by dead biomass, which provides protection for living biomass deeper within the biofilm. Practically, to control a biofilm, this result leads to the concept of treating with an HP dose exceeding a critical threshold concentration rather than a sustained, lower-concentration treatment.
Collapse
Affiliation(s)
- Philip S Stewart
- Chemical & Biological Engineering, Montana State University, Bozeman, 59717, MT, USA.
| | - Mark Owkes
- Mechanical & Industrial Engineering, Montana State University, Bozeman, 59717, MT, USA.
| |
Collapse
|
18
|
Roychowdhury S, Roth CM. Pharmacodynamic Model of the Dynamic Response of Pseudomonas aeruginosa Biofilms to Antibacterial Treatments. Biomedicines 2023; 11:2316. [PMID: 37626812 PMCID: PMC10452719 DOI: 10.3390/biomedicines11082316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/03/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Accurate pharmacokinetic-pharmacodynamic (PK-PD) models of biofilm treatment could be used to guide formulation and administration strategies to better control bacterial lung infections. To this end, we developed a detailed pharmacodynamic model of P. aeruginosa treatment with the front-line antibiotics, tobramycin and colistin, and validated it on a detailed dataset of killing dynamics. A compartmental model structure was developed in which the key features are the diffusion of the drug through a boundary layer to the bacteria, concentration-dependent interactions with bacteria, and the passage of the bacteria through successive transit states before death. The number of transit states employed was greater for tobramycin, which is a ribosomal inhibitor, than for colistin, which disrupts bacterial membranes. For both drugs, the experimentally observed delay in the killing of bacteria following drug exposure was consistent with the sum of the diffusion time and the time for passage through the transit states. For each drug, the PD model with a single set of parameters described data across a ten-fold range of concentrations and for both continuous and transient exposure protocols, as well as for combined drug treatments. The ability to predict drug response over a range of administration protocols allows this PD model to be integrated with PK descriptions to describe in vivo antibiotic response dynamics and to predict drug delivery strategies for the improved control of bacterial lung infections.
Collapse
Affiliation(s)
- Swarnima Roychowdhury
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA;
| | - Charles M. Roth
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA;
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
19
|
Prince J, Jones AAD. Heterogenous Biofilm Mass-Transport Model Replicates Periphery Sequestration of Antibiotics in P. aeruginosa PAO1 Microcolonies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551018. [PMID: 37564117 PMCID: PMC10410692 DOI: 10.1101/2023.07.28.551018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
A spatiotemporal model for antibiotic accumulation in bacterial biofilm microcolonies which leverages heterogenous porosity and attachment site profiles replicated the periphery sequestration phenomena reported in prior experimental studies on Pseudomonas aeruginosa PAO1 biofilm cell clusters. These P. aeruginosa cell clusters are in vitro models of the chronic P. aeruginosa infections found in adult cystic fibrosis patients, which display resistance to antibiotic treatments, leading to exacerbated morbidity and mortality. This resistance has been partially attributed to periphery sequestration, where antibiotics are unable to penetrate biofilm cell clusters. The underlying physical phenomena driving this periphery sequestration have not been definitively established. This paper introduces mathematical models to account for two proposed physical phenomena driving periphery sequestration: biofilm matrix attachment and volume-exclusion due to variable biofilm porosity. An antibiotic accumulation model which incorporated these phenomena was able to better fit observed periphery sequestration data compared to previous models.
Collapse
Affiliation(s)
- Joshua Prince
- Department of Civil and Environmental Engineering, Duke University
- Integrated Toxicology and Environmental Health Program, Duke University
| | - A-Andrew D Jones
- Department of Civil and Environmental Engineering, Duke University
- Integrated Toxicology and Environmental Health Program, Duke University
- Thomas Lord Department of Mechanical Engineering & Materials Science
| |
Collapse
|
20
|
Coppens B, Belpaire TE, Pešek J, Steenackers HP, Ramon H, Smeets B. Anomalous diffusion of nanoparticles in the spatially heterogeneous biofilm environment. iScience 2023; 26:106861. [PMID: 37260744 PMCID: PMC10227381 DOI: 10.1016/j.isci.2023.106861] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/20/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023] Open
Abstract
Biofilms contain extracellular polymeric substances (EPS) that provide structural support and restrict penetration of antimicrobial treatment. To overcome limited penetration, functionalized nanoparticles (NPs) have been suggested as carriers for antimicrobial delivery. Using microscopy, we evaluate the diffusion of nanoparticles in function of the structure of Salmonella biofilms. We observe anomalous diffusion and heterogeneous mobility of NPs resulting in distinct NPs distribution that depended on biofilm structure. Through Brownian dynamics modeling with spatially varying viscosity around bacteria, we demonstrated that spatial gradients in diffusivity generate viscous sinks that trap NPs near bacteria. This model replicates the characteristic diffusion signature and vertical distribution of NPs in the biofilm. From a treatment perspective, our work indicates that both biofilm structure and the level of EPS can impact NP drug delivery, where low levels of EPS might benefit delivery by immobilizing NPs closer to bacteria and higher levels hamper delivery due to shielding effects.
Collapse
Affiliation(s)
- Bart Coppens
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| | - Tom E.R. Belpaire
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| | - Jiří Pešek
- Team SIMBIOTX, Inria Saclay, 91120 Palaiseau, France
| | | | - Herman Ramon
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| | - Bart Smeets
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
21
|
Darby EM, Trampari E, Siasat P, Gaya MS, Alav I, Webber MA, Blair JMA. Molecular mechanisms of antibiotic resistance revisited. Nat Rev Microbiol 2023; 21:280-295. [PMID: 36411397 DOI: 10.1038/s41579-022-00820-y] [Citation(s) in RCA: 459] [Impact Index Per Article: 229.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/22/2022]
Abstract
Antibiotic resistance is a global health emergency, with resistance detected to all antibiotics currently in clinical use and only a few novel drugs in the pipeline. Understanding the molecular mechanisms that bacteria use to resist the action of antimicrobials is critical to recognize global patterns of resistance and to improve the use of current drugs, as well as for the design of new drugs less susceptible to resistance development and novel strategies to combat resistance. In this Review, we explore recent advances in understanding how resistance genes contribute to the biology of the host, new structural details of relevant molecular events underpinning resistance, the identification of new resistance gene families and the interactions between different resistance mechanisms. Finally, we discuss how we can use this information to develop the next generation of antimicrobial therapies.
Collapse
Affiliation(s)
- Elizabeth M Darby
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | - Pauline Siasat
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | - Ilyas Alav
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Mark A Webber
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
- Medical School, University of East Anglia, Norwich Research Park, Norwich, UK.
| | - Jessica M A Blair
- College of Medical and Dental Sciences, Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK.
| |
Collapse
|
22
|
McGlennen M, Dieser M, Foreman CM, Warnat S. Monitoring biofilm growth and dispersal in real-time with impedance biosensors. J Ind Microbiol Biotechnol 2023; 50:kuad022. [PMID: 37653441 PMCID: PMC10485796 DOI: 10.1093/jimb/kuad022] [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: 05/19/2023] [Accepted: 08/22/2023] [Indexed: 09/02/2023]
Abstract
Microbial biofilm contamination is a widespread problem that requires precise and prompt detection techniques to effectively control its growth. Microfabricated electrochemical impedance spectroscopy (EIS) biosensors offer promise as a tool for early biofilm detection and monitoring of elimination. This study utilized a custom flow cell system with integrated sensors to make real-time impedance measurements of biofilm growth under flow conditions, which were correlated with confocal laser scanning microscopy (CLSM) imaging. Biofilm growth on EIS biosensors in basic aqueous growth media (tryptic soy broth, TSB) and an oil-water emulsion (metalworking fluid, MWF) attenuated in a sigmoidal decay pattern, which lead to an ∼22-25% decrease in impedance after 24 Hrs. Subsequent treatment of established biofilms increased the impedance by ∼14% and ∼41% in TSB and MWF, respectively. In the presence of furanone C-30, a quorum-sensing inhibitor (QSI), impedance remained unchanged from the initial time point for 18 Hrs in TSB and 72 Hrs in MWF. Biofilm changes enumerated from CLSM imaging corroborated impedance measurements, with treatment significantly reducing biofilm. Overall, these results support the application of microfabricated EIS biosensors for evaluating the growth and dispersal of biofilm in situ and demonstrate potential for use in industrial settings. ONE-SENTENCE SUMMARY This study demonstrates the use of microfabricated electrochemical impedance spectroscopy (EIS) biosensors for real-time monitoring and treatment evaluation of biofilm growth, offering valuable insights for biofilm control in industrial settings.
Collapse
Affiliation(s)
- Matthew McGlennen
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
- Mechanical and Industrial Engineering, Montana State University, Bozeman, MT 59717, USA
| | - Markus Dieser
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
- Chemical and Biological Engineering, Montana State University, Bozeman, MT 59717, USA
| | - Christine M Foreman
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
- Chemical and Biological Engineering, Montana State University, Bozeman, MT 59717, USA
| | - Stephan Warnat
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
- Mechanical and Industrial Engineering, Montana State University, Bozeman, MT 59717, USA
| |
Collapse
|
23
|
Flemming HC, van Hullebusch ED, Neu TR, Nielsen PH, Seviour T, Stoodley P, Wingender J, Wuertz S. The biofilm matrix: multitasking in a shared space. Nat Rev Microbiol 2023; 21:70-86. [PMID: 36127518 DOI: 10.1038/s41579-022-00791-0] [Citation(s) in RCA: 291] [Impact Index Per Article: 145.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 01/20/2023]
Abstract
The biofilm matrix can be considered to be a shared space for the encased microbial cells, comprising a wide variety of extracellular polymeric substances (EPS), such as polysaccharides, proteins, amyloids, lipids and extracellular DNA (eDNA), as well as membrane vesicles and humic-like microbially derived refractory substances. EPS are dynamic in space and time and their components interact in complex ways, fulfilling various functions: to stabilize the matrix, acquire nutrients, retain and protect eDNA or exoenzymes, or offer sorption sites for ions and hydrophobic substances. The retention of exoenzymes effectively renders the biofilm matrix an external digestion system influencing the global turnover of biopolymers, considering the ubiquitous relevance of biofilms. Physico-chemical and biological interactions and environmental conditions enable biofilm systems to morph into films, microcolonies and macrocolonies, films, ridges, ripples, columns, pellicles, bubbles, mushrooms and suspended aggregates - in response to the very diverse conditions confronting a particular biofilm community. Assembly and dynamics of the matrix are mostly coordinated by secondary messengers, signalling molecules or small RNAs, in both medically relevant and environmental biofilms. Fully deciphering how bacteria provide structure to the matrix, and thus facilitate and benefit from extracellular reactions, remains the challenge for future biofilm research.
Collapse
Affiliation(s)
- Hans-Curt Flemming
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore.
| | | | - Thomas R Neu
- Department of River Ecology, Helmholtz Centre for Environmental Research - UFZ, Magdeburg, Germany
| | - Per H Nielsen
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Thomas Seviour
- Aarhus University Centre for Water Technology, Department of Biological and Chemical Engineering, Aarhus University, Aarhus, Denmark
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA.,Department of Orthopaedics, The Ohio State University, Columbus, OH, USA
| | - Jost Wingender
- University of Duisburg-Essen, Biofilm Centre, Department of Aquatic Microbiology, Essen, Germany
| | - Stefan Wuertz
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
24
|
Williamson KS, Dlakić M, Akiyama T, Franklin MJ. The Pseudomonas aeruginosa RpoH (σ 32) Regulon and Its Role in Essential Cellular Functions, Starvation Survival, and Antibiotic Tolerance. Int J Mol Sci 2023; 24:1513. [PMID: 36675051 PMCID: PMC9866376 DOI: 10.3390/ijms24021513] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The bacterial heat-shock response is regulated by the alternative sigma factor, σ32 (RpoH), which responds to misfolded protein stress and directs the RNA polymerase to the promoters for genes required for protein refolding or degradation. In P. aeruginosa, RpoH is essential for viability under laboratory growth conditions. Here, we used a transcriptomics approach to identify the genes of the RpoH regulon, including RpoH-regulated genes that are essential for P. aeruginosa. We placed the rpoH gene under control of the arabinose-inducible PBAD promoter, then deleted the chromosomal rpoH allele. This allowed transcriptomic analysis of the RpoH (σ32) regulon following a short up-shift in the cellular concentration of RpoH by arabinose addition, in the absence of a sudden change in temperature. The P. aeruginosa ∆rpoH (PBAD-rpoH) strain grew in the absence of arabinose, indicating that some rpoH expression occurred without arabinose induction. When arabinose was added, the rpoH mRNA abundance of P. aeruginosa ∆rpoH (PBAD-rpoH) measured by RT-qPCR increased five-fold within 15 min of arabinose addition. Transcriptome results showed that P. aeruginosa genes required for protein repair or degradation are induced by increased RpoH levels, and that many genes essential for P. aeruginosa growth are induced by RpoH. Other stress response genes induced by RpoH are involved in damaged nucleic acid repair and in amino acid metabolism. Annotation of the hypothetical proteins under RpoH control included proteins that may play a role in antibiotic resistances and in non-ribosomal peptide synthesis. Phenotypic analysis of P. aeruginosa ∆rpoH (PBAD-rpoH) showed that it is impaired in its ability to survive during starvation compared to the wild-type strain. P. aeruginosa ∆rpoH (PBAD-rpoH) also had increased sensitivity to aminoglycoside antibiotics, but not to other classes of antibiotics, whether cultured planktonically or in biofilms. The enhanced aminoglycoside sensitivity of the mutant strain may be due to indirect effects, such as the build-up of toxic misfolded proteins, or to the direct effect of genes, such as aminoglycoside acetyl transferases, that are regulated by RpoH. Overall, the results demonstrate that RpoH regulates genes that are essential for viability of P. aeruginosa, that it protects P. aeruginosa from damage from aminoglycoside antibiotics, and that it is required for survival during nutrient-limiting conditions.
Collapse
Affiliation(s)
- Kerry S. Williamson
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
| | - Mensur Dlakić
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Tatsuya Akiyama
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
| | - Michael J. Franklin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
| |
Collapse
|
25
|
Mohamed A, Raval YS, Gelston S, Tibbits G, Ay SU, Flurin L, Greenwood-Quaintance KE, Patel R, Beyenal H. Anti-Biofilm Activity of a Tunable Hypochlorous Acid-Generating Electrochemical Bandage Controlled By a Wearable Potentiostat. ADVANCED ENGINEERING MATERIALS 2023; 25:2200792. [PMID: 36817722 PMCID: PMC9937732 DOI: 10.1002/adem.202200792] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Indexed: 05/07/2023]
Abstract
Chronic wound biofilm infections represent a major clinical challenge which results in a substantial burden to patients and healthcare systems. Treatment with topical antibiotics is oftentimes ineffective as a result of antibiotic-resistant microorganisms and biofilm-specific antibiotic tolerance. Use of biocides such as hypochlorous acid (HOCl) has gained increasing attention due to the lack of known resistance mechanisms. We designed an HOCl-generating electrochemical bandage (e-bandage) that delivers HOCl continuously at low concentrations targeting infected wound beds in a similar manner to adhesive antimicrobial wound dressings. We developed a battery-operated wearable potentiostat that controls the e-bandage electrodes at potentials suitable for HOCl generation. We demonstrated that e-bandage treatment was tunable by changing the applied potential. HOCl generation on electrode surfaces was verified using microelectrodes. The developed e-bandage showed time-dependent responses against in vitro Acinetobacter baumannii and Staphylococcus aureus biofilms, reducing viable cells to non-detectable levels within 6 and 12 hours of treatment, respectively. The developed e-bandage should be further evaluated as an alternative to topical antibiotics to treat wound biofilm infections.
Collapse
Affiliation(s)
- Abdelrhman Mohamed
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Pullman, WA, USA
| | - Yash S. Raval
- Division of Clinical Microbiology, Mayo Clinic, Rochester, MN, USA
| | - Suzanne Gelston
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Pullman, WA, USA
| | - Gretchen Tibbits
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Pullman, WA, USA
| | - Suat U. Ay
- Department of Electrical and Computer Engineering, University of Idaho, Moscow
| | - Laure Flurin
- Division of Clinical Microbiology, Mayo Clinic, Rochester, MN, USA
- Department of Intensive Care, University Hospital of Guadeloupe, Pointe-à-Pitre, France
| | | | - Robin Patel
- Division of Clinical Microbiology, Mayo Clinic, Rochester, MN, USA
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| | - Haluk Beyenal
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Pullman, WA, USA
| |
Collapse
|
26
|
Lichtenberg M, Kvich L, Larsen SLB, Jakobsen TH, Bjarnsholt T. Inoculum Concentration Influences Pseudomonas aeruginosa Phenotype and Biofilm Architecture. Microbiol Spectr 2022; 10:e0313122. [PMID: 36354337 PMCID: PMC9769529 DOI: 10.1128/spectrum.03131-22] [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: 09/30/2022] [Accepted: 10/21/2022] [Indexed: 11/12/2022] Open
Abstract
In infections, bacterial cells are often found as relatively small multicellular aggregates characterized by a heterogeneous distribution of phenotype, genotype, and growth rates depending on their surrounding microenvironment. Many laboratory models fail to mimic these characteristics, and experiments are often initiated from planktonic bacteria given optimal conditions for rapid growth without concerns about the microenvironmental characteristics during biofilm maturation. Therefore, we investigated how the initial bacterial concentration (henceforth termed the inoculum) influences the microenvironment during initial growth and how this affects the sizes and distribution of developed aggregates in an embedded biofilm model-the alginate bead biofilm model. Following 24 h of incubation, the viable biomass was independent of starting inoculum but with a radically different microenvironment which led to differences in metabolic activity depending on the inoculum. The inoculum also affected the number of cells surviving treatment with the antibiotic tobramycin, where the highest inoculum showed higher survival rates than the lowest inoculum. The change in antibiotic tolerance was correlated with cell-specific RNA content and O2 consumption rates, suggesting a direct role of metabolic activity. Thus, the starting number of bacteria results in different phenotypic trajectories governed by different microenvironmental characteristics, and we demonstrate some of the possible implications of such physiological gradients on the outcome of in vitro experiments. IMPORTANCE Biofilm aggregates grown in the alginate bead biofilm model bear resemblance to features of in vivo biofilms. Here, we show that changing the initial concentration of bacteria in the biofilm model leads to widely different behavior of the bacteria following an incubation period. This difference is influenced by the local conditions experienced by the bacteria during growth, which impact their response to antibiotic treatment. Our study provides a framework for manipulating aggregate sizes in in vitro biofilm models. It underlines the importance of how experiments are initiated, which can profoundly impact the outcomes and interpretation of microbiological experiments.
Collapse
Affiliation(s)
- Mads Lichtenberg
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lasse Kvich
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Sara Louise Borregaard Larsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Tim Holm Jakobsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
27
|
González-Plaza JJ, Furlan C, Rijavec T, Lapanje A, Barros R, Tamayo-Ramos JA, Suarez-Diez M. Advances in experimental and computational methodologies for the study of microbial-surface interactions at different omics levels. Front Microbiol 2022; 13:1006946. [PMID: 36519168 PMCID: PMC9744117 DOI: 10.3389/fmicb.2022.1006946] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/02/2022] [Indexed: 08/31/2023] Open
Abstract
The study of the biological response of microbial cells interacting with natural and synthetic interfaces has acquired a new dimension with the development and constant progress of advanced omics technologies. New methods allow the isolation and analysis of nucleic acids, proteins and metabolites from complex samples, of interest in diverse research areas, such as materials sciences, biomedical sciences, forensic sciences, biotechnology and archeology, among others. The study of the bacterial recognition and response to surface contact or the diagnosis and evolution of ancient pathogens contained in archeological tissues require, in many cases, the availability of specialized methods and tools. The current review describes advances in in vitro and in silico approaches to tackle existing challenges (e.g., low-quality sample, low amount, presence of inhibitors, chelators, etc.) in the isolation of high-quality samples and in the analysis of microbial cells at genomic, transcriptomic, proteomic and metabolomic levels, when present in complex interfaces. From the experimental point of view, tailored manual and automatized methodologies, commercial and in-house developed protocols, are described. The computational level focuses on the discussion of novel tools and approaches designed to solve associated issues, such as sample contamination, low quality reads, low coverage, etc. Finally, approaches to obtain a systems level understanding of these complex interactions by integrating multi omics datasets are presented.
Collapse
Affiliation(s)
- Juan José González-Plaza
- International Research Centre in Critical Raw Materials-ICCRAM, University of Burgos, Burgos, Spain
| | - Cristina Furlan
- Laboratory of Systems and Synthetic Biology, Wageningen University and Research, Wageningen, Netherlands
| | - Tomaž Rijavec
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Aleš Lapanje
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Rocío Barros
- International Research Centre in Critical Raw Materials-ICCRAM, University of Burgos, Burgos, Spain
| | | | - Maria Suarez-Diez
- Laboratory of Systems and Synthetic Biology, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
28
|
Coenye T, Bové M, Bjarnsholt T. Biofilm antimicrobial susceptibility through an experimental evolutionary lens. NPJ Biofilms Microbiomes 2022; 8:82. [PMID: 36257971 PMCID: PMC9579162 DOI: 10.1038/s41522-022-00346-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/04/2022] [Indexed: 11/19/2022] Open
Abstract
Experimental evolution experiments in which bacterial populations are repeatedly exposed to an antimicrobial treatment, and examination of the genotype and phenotype of the resulting evolved bacteria, can help shed light on mechanisms behind reduced susceptibility. In this review we present an overview of why it is important to include biofilms in experimental evolution, which approaches are available to study experimental evolution in biofilms and what experimental evolution has taught us about tolerance and resistance in biofilms. Finally, we present an emerging consensus view on biofilm antimicrobial susceptibility supported by data obtained during experimental evolution studies.
Collapse
Affiliation(s)
- Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium.
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark.
| | - Mona Bové
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Lichtenberg M, Kragh KN, Fritz B, Kirkegaard JB, Tolker-Nielsen T, Bjarnsholt T. Cyclic-di-GMP signaling controls metabolic activity in Pseudomonas aeruginosa. Cell Rep 2022; 41:111515. [DOI: 10.1016/j.celrep.2022.111515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/13/2022] [Accepted: 09/26/2022] [Indexed: 11/03/2022] Open
|
30
|
NO-Stressed Y. pseudotuberculosis Has Decreased Cell Division Rates in the Mouse Spleen. Infect Immun 2022; 90:e0016722. [PMID: 35862700 PMCID: PMC9387282 DOI: 10.1128/iai.00167-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fluorescence dilution approaches can detect bacterial cell division events and can detect if there are differential rates of cell division across individual cells within a population. This approach typically involves inducing expression of a fluorescent protein and then tracking partitioning of fluorescence into daughter cells. However, fluorescence can be diluted very quickly within a rapidly replicating population, such as pathogenic bacterial populations replicating within host tissues. To overcome this limitation, we have generated two revTetR reporter constructs, where either mCherry or yellow fluorescent protein (YFP) is constitutively expressed and repressed by addition of tetracyclines, resulting in fluorescence dilution within defined time frames. We show that fluorescent signals are diluted in replicating populations and that signal accumulates in growth-inhibited populations, including during nitric oxide (NO) exposure. Furthermore, we show that tetracyclines can be delivered to the mouse spleen during Yersinia pseudotuberculosis infection and defined a drug concentration that results in even exposure of cells to tetracyclines. We then used this system to visualize bacterial cell division within defined time frames postinfection. revTetR-mCherry allowed us to detect slow-growing cells in response to NO in culture; however, this strain had a growth defect within mouse tissues, which complicated results. To address this issue, we constructed revTetR-YFP using the less toxic YFP and showed that heightened NO exposure correlated with heightened YFP signal, indicating decreased cell division rates within this subpopulation in vivo. This revTetR reporter will provide a critical tool for future studies to identify and isolate slowly replicating bacterial subpopulations from host tissues.
Collapse
|
31
|
Harding MW, Marques LLR, Allan N, Olson ME, Buziak B, Nadworny P, Omar A, Howard RJ, Feng J. Bactericidal Efficacy of Oxidized Silver against Biofilms Formed by Curtobacterium flaccumfaciens pv. flaccumfaciens. THE PLANT PATHOLOGY JOURNAL 2022; 38:334-344. [PMID: 35953053 PMCID: PMC9372099 DOI: 10.5423/ppj.oa.04.2022.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
Bacterial wilt is a re-emerging disease on dry bean and can affect many other crop species within the Fabaceae. The causal agent, Curtobacterium flaccumfaciens pv. flaccumfaciens (CFF), is a small, Gram-positive, rodshaped bacterium that is seed-transmitted. Infections in the host become systemic, leading to wilting and economic loss. Clean seed programs and bactericidal seed treatments are two critical management tools. This study characterizes the efficacies of five bactericidal chemicals against CFF. It was hypothesized that this bacterium was capable of forming biofilms, and that the cells within biofilms would be more tolerant to bactericidal treatments. The minimum biocide eradication concentration assay protocol was used to grow CFF biofilms, expose the biofilms to bactericides, and enumerate survivors compared to a non-treated control (water). Streptomycin and oxysilver bisulfate had EC95 values at the lowest concentrations and are likely the best candidates for seed treatment products for controlling seed-borne bacterial wilt of bean. The results showed that CFF formed biofilms during at least two phases of the bacterial wilt disease cycle, and the biofilms were much more difficult to eradicate than their planktonic counterparts. Overall, biofilm formation by CFF is an important part of the bacterial wilt disease cycle in dry edible bean and antibiofilm bactericides such as streptomycin and oxysilver bisulfate may be best suited for use in disease management.
Collapse
Affiliation(s)
- Michael W. Harding
- Alberta Agriculture, Forestry and Rural Economic Development, Crop Diversification Centre South, Brooks, AB, T1R 1E6,
Canada
| | | | - Nick Allan
- Chinook Contract Research, Airdrie, AB, T4A 0C3,
Canada
| | | | | | | | - Amin Omar
- Innovotech Inc., Edmonton, AB, T6N 1H1,
Canada
- Department of Pharmaceutical Sciences and Pharmaceutics, Faculty of Pharmacy, Applied Science Private University, Amman,
Jordan
| | | | - Jie Feng
- Alberta Agriculture, Forestry and Rural Economic Development, Alberta Plant Health Lab, Edmonton, Alberta, T5Y 6H3,
Canada
| |
Collapse
|
32
|
Lanni A, Borroni E, Iacobino A, Russo C, Gentile L, Fattorini L, Giannoni F. Activity of Drug Combinations against Mycobacterium abscessus Grown in Aerobic and Hypoxic Conditions. Microorganisms 2022; 10:microorganisms10071421. [PMID: 35889140 PMCID: PMC9316547 DOI: 10.3390/microorganisms10071421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/30/2022] Open
Abstract
Infections caused by Mycobacterium abscessus (Mab), an environmental non-tuberculous mycobacterium, are difficult to eradicate from patients with pulmonary diseases such as cystic fibrosis and bronchiectasis even after years of antibiotic treatments. In these people, the low oxygen pressure in mucus and biofilm may restrict Mab growth from actively replicating aerobic (A) to non-replicating hypoxic (H) stages, which are known to be extremely drug-tolerant. After the exposure of Mab A and H cells to drugs, killing was monitored by measuring colony-forming units (CFU) and regrowth in liquid medium (MGIT 960) of 1-day-old A cells (A1) and 5-day-old H cells (H5). Mab killing was defined as a lack of regrowth of drug-exposed cells in MGIT tubes after >50 days of incubation. Out of 18 drugs tested, 14-day treatments with bedaquiline-amikacin (BDQ-AMK)-containing three-drug combinations were very active against A1 + H5 cells. However, drug-tolerant cells (persisters) were not killed, as shown by CFU curves with typical bimodal trends. Instead, 56-day treatments with the nitrocompounds containing combinations BDQ-AMK-rifabutin-clarithromycin-nimorazole and BDQ-AMK-rifabutin-clarithromycin-metronidazole-colistin killed all A1 + H5 Mab cells in 42 and 56 days, respectively, as shown by lack of regrowth in agar and MGIT medium. Overall, these data indicated that Mab persisters may be killed by appropriate drug combinations.
Collapse
Affiliation(s)
- Alessio Lanni
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.L.); (A.I.); (L.F.)
| | - Emanuele Borroni
- Emerging Bacterial Pathogens Unit, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Angelo Iacobino
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.L.); (A.I.); (L.F.)
| | - Cristina Russo
- Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (C.R.); (L.G.)
| | - Leonarda Gentile
- Bambino Gesù Children’s Hospital, 00165 Rome, Italy; (C.R.); (L.G.)
| | - Lanfranco Fattorini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.L.); (A.I.); (L.F.)
| | - Federico Giannoni
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.L.); (A.I.); (L.F.)
- Correspondence: ; Tel.: +39-06-49902318; Fax: +39-06-49387112
| |
Collapse
|
33
|
Nitrate respiration occurs throughout the depth of mucoid and non-mucoid Pseudomonas aeruginosa submerged agar colony biofilms including the oxic zone. Sci Rep 2022; 12:8557. [PMID: 35595796 PMCID: PMC9123002 DOI: 10.1038/s41598-022-11957-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen and well characterized biofilm former. P. aeruginosa forms strong oxygen gradients inside biofilms due to rapid oxygen respiration in the top layers and the poor solubility of oxygen coupled with diffusion limited transport. Transcriptomic evidence from in vitro and ex vivo sampling suggests that denitrification is occurring in biofilms in ostensibly oxic environments. It is hypothesized that in the presence of nitrate there is stratification with aerobic respiration occurring in the outer oxic layer and denitrification in the lower anoxic zone. We used submerged agar colony biofilms grown from mucoid (FRD1) and non-mucoid (PAO1) strains to simultaneously measure depth microprofiles of oxygen and nitrous oxide in the same colony with microelectrodes. Oxygen respiration occurred at the top of the colony as expected but denitrification occurred throughout the entire depth, even in the oxic region. Local denitrification rates were highly variable suggesting heterogenous metabolic activity within the colony. We also assessed the short-term influence of tobramycin on aerobic respiration within a PAO1 colony. Although there was an immediate reduction in respiration it was never completely arrested over a 2 h period. On tobramycin removal the oxygen gradient steadily reestablished, demonstrating immediate recovery of metabolic activity.
Collapse
|
34
|
Malone M, Schultz G. Challenges in the diagnosis and management of wound infection. Br J Dermatol 2022; 187:159-166. [PMID: 35587707 DOI: 10.1111/bjd.21612] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2022] [Indexed: 12/16/2022]
Abstract
Human epithelia are constantly exposed to microorganisms present in the environment or residing as part of commensal flora. Despite this exposure, infections involving the skin and subcutaneous tissue in healthy individuals are, fortunately, quite rare. Many of the wounds that afflict the human body occur in individuals of ill health and/or where the mechanism of wounding is impeded by host immunological, physiological or regenerative dysfunction. The interplay between microorganisms and host immunity is complex and remains ill defined; however, the interpretation of downstream manifestations of the host response to invading microorganisms is still based largely on the clinical signs and symptoms of an active infectious process. In this review article we will provide a brief overview of the current challenges clinicians face in diagnosing wound infections, how chronic infections caused by biofilms are a major challenge, and how there have been minimal advancements in developing new diagnostics or therapeutics in the identification and management of wound infections.
Collapse
Affiliation(s)
- Matthew Malone
- South West Sydney Limb Preservation and Wound Research, South Western Sydney LHD, Sydney, NSW, Australia.,Infectious Diseases and Microbiology, School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Gregory Schultz
- Institute for Wound Research, University of Florida, Gainesville, FL, USA
| |
Collapse
|
35
|
Stevanovic M, Boukéké-Lesplulier T, Hupe L, Hasty J, Bittihn P, Schultz D. Nutrient Gradients Mediate Complex Colony-Level Antibiotic Responses in Structured Microbial Populations. Front Microbiol 2022; 13:740259. [PMID: 35572643 PMCID: PMC9093743 DOI: 10.3389/fmicb.2022.740259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Antibiotic treatments often fail to eliminate bacterial populations due to heterogeneity in how individual cells respond to the drug. In structured bacterial populations such as biofilms, bacterial metabolism and environmental transport processes lead to an emergent phenotypic structure and self-generated nutrient gradients toward the interior of the colony, which can affect cell growth, gene expression and susceptibility to the drug. Even in single cells, survival depends on a dynamic interplay between the drug's action and the expression of resistance genes. How expression of resistance is coordinated across populations in the presence of such spatiotemporal environmental coupling remains elusive. Using a custom microfluidic device, we observe the response of spatially extended microcolonies of tetracycline-resistant E. coli to precisely defined dynamic drug regimens. We find an intricate interplay between drug-induced changes in cell growth and growth-dependent expression of resistance genes, resulting in the redistribution of metabolites and the reorganization of growth patterns. This dynamic environmental feedback affects the regulation of drug resistance differently across the colony, generating dynamic phenotypic structures that maintain colony growth during exposure to high drug concentrations and increase population-level resistance to subsequent exposures. A mathematical model linking metabolism and the regulation of gene expression is able to capture the main features of spatiotemporal colony dynamics. Uncovering the fundamental principles that govern collective mechanisms of antibiotic resistance in spatially extended populations will allow the design of optimal drug regimens to counteract them.
Collapse
Affiliation(s)
- Mirjana Stevanovic
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Thomas Boukéké-Lesplulier
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Lukas Hupe
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Jeff Hasty
- BioCircuits Institute, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States.,Department of Bioengineering, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States.,Molecular Biology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Philip Bittihn
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany.,BioCircuits Institute, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, United States
| | - Daniel Schultz
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| |
Collapse
|
36
|
Hurlow J, Bowler PG. Acute and chronic wound infections: microbiological, immunological, clinical and therapeutic distinctions. J Wound Care 2022; 31:436-445. [PMID: 35579319 DOI: 10.12968/jowc.2022.31.5.436] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Wound infection is a complex pathology that may manifest either as a rapid onset acute condition, or as a prolonged chronic condition. Although systemic antibiotic therapy is often appropriate and necessary for acute wound infections, it is often used inappropriately, excessively and unsuccessfully in chronic wound infections. Overuse of antibiotics in chronic (hard-to-heal) wound management contributes to antibiotic resistance. This literature review confirms that acute and chronic wound infections are significantly differentiated by their cause (microbial phenotype), the subsequent host immune response and by the resulting clinical manifestations. Consequently, recognition of the type of wound infection followed by appropriate and timely therapy is required to improve wound healing outcomes while encouraging more judicious and responsible use of antibiotics.
Collapse
|
37
|
Stewart PS, Williamson KS, Boegli L, Hamerly T, White B, Scott L, Hu X, Mumey BM, Franklin MJ, Bothner B, Vital-Lopez FG, Wallqvist A, James GA. Search for a Shared Genetic or Biochemical Basis for Biofilm Tolerance to Antibiotics across Bacterial Species. Antimicrob Agents Chemother 2022; 66:e0002122. [PMID: 35266829 PMCID: PMC9017379 DOI: 10.1128/aac.00021-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/29/2022] [Indexed: 12/19/2022] Open
Abstract
Is there a universal genetically programmed defense providing tolerance to antibiotics when bacteria grow as biofilms? A comparison between biofilms of three different bacterial species by transcriptomic and metabolomic approaches uncovered no evidence of one. Single-species biofilms of three bacterial species (Pseudomonas aeruginosa, Staphylococcus aureus, and Acinetobacter baumannii) were grown in vitro for 3 days and then challenged with respective antibiotics (ciprofloxacin, daptomycin, and tigecycline) for an additional 24 h. All three microorganisms displayed reduced susceptibility in biofilms compared to planktonic cultures. Global transcriptomic profiling of gene expression comparing biofilm to planktonic and antibiotic-treated biofilm to untreated biofilm was performed. Extracellular metabolites were measured to characterize the utilization of carbon sources between biofilms, treated biofilms, and planktonic cells. While all three bacteria exhibited a species-specific signature of stationary phase, no conserved gene, gene set, or common functional pathway could be identified that changed consistently across the three microorganisms. Across the three species, glucose consumption was increased in biofilms compared to planktonic cells, and alanine and aspartic acid utilization were decreased in biofilms compared to planktonic cells. The reasons for these changes were not readily apparent in the transcriptomes. No common shift in the utilization pattern of carbon sources was discerned when comparing untreated to antibiotic-exposed biofilms. Overall, our measurements do not support the existence of a common genetic or biochemical basis for biofilm tolerance against antibiotics. Rather, there are likely myriad genes, proteins, and metabolic pathways that influence the physiological state of individual microorganisms in biofilms and contribute to antibiotic tolerance.
Collapse
Affiliation(s)
- Philip S. Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, Montana, USA
| | - Kerry S. Williamson
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Laura Boegli
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
| | - Timothy Hamerly
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Ben White
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Liam Scott
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Xiao Hu
- Gianforte School of Computing, Montana State University, Bozeman, Montana, USA
| | - Brendan M. Mumey
- Gianforte School of Computing, Montana State University, Bozeman, Montana, USA
| | - Michael J. Franklin
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, USA
| | - Brian Bothner
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Francisco G. Vital-Lopez
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, Maryland, USA
| | - Garth A. James
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
38
|
Thomas-Moore BA, Del Valle CA, Field RA, Marín MJ. Recent advances in nanoparticle-based targeting tactics for antibacterial photodynamic therapy. Photochem Photobiol Sci 2022; 21:1111-1131. [PMID: 35384638 PMCID: PMC9287206 DOI: 10.1007/s43630-022-00194-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 02/23/2022] [Indexed: 12/21/2022]
Abstract
Abstract The rise of antibacterial drug resistance means treatment options are becoming increasingly limited. We must find ways to tackle these hard-to-treat drug-resistant and biofilm infections. With the lack of new antibacterial drugs (such as antibiotics) reaching the clinics, research has switched focus to exploring alternative strategies. One such strategy is antibacterial photodynamic therapy (aPDT), a system that relies on light, oxygen, and a non-toxic dye (photosensitiser) to generate cytotoxic reactive oxygen species. This technique has already been shown capable of handling both drug-resistant and biofilm infections but has limited clinical approval to date, which is in part due to the low bioavailability and selectivity of hydrophobic photosensitisers. Nanotechnology-based techniques have the potential to address the limitations of current aPDT, as already well-documented in anti-cancer PDT. Here, we review recent advances in nanoparticle-based targeting tactics for aPDT. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Brydie A Thomas-Moore
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
- Norwich Research Park Innovation Centre, Iceni Glycoscience Ltd, Colney Lane, Norwich, NR4 7GJ, UK.
| | - Carla Arnau Del Valle
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Robert A Field
- Norwich Research Park Innovation Centre, Iceni Glycoscience Ltd, Colney Lane, Norwich, NR4 7GJ, UK
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| | - María J Marín
- School of Chemistry, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
39
|
Biofilm Survival Strategies in Chronic Wounds. Microorganisms 2022; 10:microorganisms10040775. [PMID: 35456825 PMCID: PMC9025119 DOI: 10.3390/microorganisms10040775] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 01/22/2023] Open
Abstract
Bacterial biofilms residing in chronic wounds are thought to have numerous survival strategies, making them extremely difficult to eradicate and resulting in long-term infections. However, much of our knowledge regarding biofilm persistence stems from in vitro models and experiments performed in vivo in animal models. While the knowledge obtained from such experiments is highly valuable, its direct translation to the human clinical setting should be undertaken with caution. In this review, we highlight knowledge obtained from human clinical samples in different aspects of biofilm survival strategies. These strategies have been divided into segments of the following attributes: altered transcriptomic profiles, spatial distribution, the production of extracellular polymeric substances, an altered microenvironment, inter-and intra-species interactions, and heterogeneity in the bacterial population. While all these attributes are speculated to contribute to the enhanced persistence of biofilms in chronic wounds, only some of them have been demonstrated to exist in human wounds. Some of the attributes have been observed in other clinical diseases while others have only been observed in vitro. Here, we have strived to clarify the limitations of the current knowledge in regard to this specific topic, without ignoring important in vitro and in vivo observations.
Collapse
|
40
|
Fernandes S, Gomes IB, Sousa SF, Simões M. Antimicrobial Susceptibility of Persister Biofilm Cells of Bacillus cereus and Pseudomonas fluorescens. Microorganisms 2022; 10:160. [PMID: 35056610 PMCID: PMC8779418 DOI: 10.3390/microorganisms10010160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/04/2023] Open
Abstract
The present study evaluates the antimicrobial susceptibility of persister cells of Bacillus cereus and Pseudomonas fluorescens after their regrowth in suspension and as biofilms. Two conventional (benzalkonium chloride-BAC and peracetic acid-PAA) and two emerging biocides (glycolic acid-GA and glyoxal-GO) were selected for this study. Persister cells resulted from biofilms subjected to a critical treatment using the selected biocides. All biocide treatments developed B. cereus persister cells, except PAA that effectively reduced the levels of vegetative cells and endospores. P. fluorescens persister cells comprise viable and viable but non-culturable cells. Afterwards, persister cells were regrown in suspension and in biofilms and were subjected to a second biocide treatment. In general, planktonic cultures of regrown persister cells in suspension lost their antimicrobial tolerance, for both bacteria. Regrown biofilms of persister cells had antimicrobial susceptibility close to those regrown biofilms of biocide-untreated cells, except for regrown biofilms of persister P. fluorescens after BAC treatment, which demonstrated increased antimicrobial tolerance. The most active biocide against persister cells was PAA, which did not promote changes in susceptibility after their regrowth. In conclusion, persister cells are ubiquitous within biofilms and survive after critical biocide treatment. The descendant planktonic and biofilms populations showed similar properties as the original ones.
Collapse
Affiliation(s)
- Susana Fernandes
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (S.F.); (I.B.G.)
| | - Inês B. Gomes
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (S.F.); (I.B.G.)
| | - Sérgio F. Sousa
- UCIBIO/REQUIMTE, BioSIM, Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal;
| | - Manuel Simões
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; (S.F.); (I.B.G.)
| |
Collapse
|
41
|
Metabolic adaption to extracellular pyruvate triggers biofilm formation in Clostridioides difficile. THE ISME JOURNAL 2021; 15:3623-3635. [PMID: 34155333 PMCID: PMC8630010 DOI: 10.1038/s41396-021-01042-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 02/06/2023]
Abstract
Clostridioides difficile infections are associated with gut microbiome dysbiosis and are the leading cause of hospital-acquired diarrhoea. The infectious process is strongly influenced by the microbiota and successful infection relies on the absence of specific microbiota-produced metabolites. Deoxycholate and short-chain fatty acids are microbiota-produced metabolites that limit the growth of C. difficile and protect the host against this infection. In a previous study, we showed that deoxycholate causes C. difficile to form strongly adherent biofilms after 48 h. Here, our objectives were to identify and characterize key molecules and events required for biofilm formation in the presence of deoxycholate. We applied time-course transcriptomics and genetics to identify sigma factors, metabolic processes and type IV pili that drive biofilm formation. These analyses revealed that extracellular pyruvate induces biofilm formation in the presence of deoxycholate. In the absence of deoxycholate, pyruvate supplementation was sufficient to induce biofilm formation in a process that was dependent on pyruvate uptake by the membrane protein CstA. In the context of the human gut, microbiota-generated pyruvate is a metabolite that limits pathogen colonization. Taken together our results suggest that pyruvate-induced biofilm formation might act as a key process driving C. difficile persistence in the gut.
Collapse
|
42
|
Rilstone V, Vignale L, Craddock J, Cushing A, Filion Y, Champagne P. The role of antibiotics and heavy metals on the development, promotion, and dissemination of antimicrobial resistance in drinking water biofilms. CHEMOSPHERE 2021; 282:131048. [PMID: 34470147 DOI: 10.1016/j.chemosphere.2021.131048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 06/13/2023]
Abstract
Antimicrobial resistance (AMR), as well as the development of biofilms in drinking water distribution systems (DWDSs), have become an increasing concern for public health and management. As bulk water travels from source to tap, it may accumulate contaminants of emerging concern (CECs) such as antibiotics and heavy metals. When these CECs and other selective pressures, such as disinfection, pipe material, temperature, pH, and nutrient availability interact with planktonic cells and, consequently, DWDS biofilms, AMR is promoted. The purpose of this review is to highlight the mechanisms by which AMR develops and is disseminated within DWDS biofilms. First, this review will lay a foundation by describing how DWDS biofilms form, as well as their basic intrinsic and acquired resistance mechanisms. Next, the selective pressures that further induce AMR in DWDS biofilms will be elaborated. Then, the pressures by which antibiotic and heavy metal CECs accumulate in DWDS biofilms, their individual resistance mechanisms, and co-selection are described and discussed. Finally, the known human health risks and current management strategies to mitigate AMR in DWDSs will be presented. Overall, this review provides critical connections between several biotic and abiotic factors that influence and induce AMR in DWDS biofilms. Implications are made regarding the importance of monitoring and managing the development, promotion, and dissemination of AMR in DWDS biofilms.
Collapse
Affiliation(s)
- Victoria Rilstone
- Beaty Water Research Centre, Department of Civil Engineering, Union Street, Queen's University, Kingston, K7L 3Z6, Canada
| | - Leah Vignale
- Beaty Water Research Centre, Department of Civil Engineering, Union Street, Queen's University, Kingston, K7L 3Z6, Canada
| | - Justine Craddock
- Beaty Water Research Centre, Department of Civil Engineering, Union Street, Queen's University, Kingston, K7L 3Z6, Canada
| | - Alexandria Cushing
- Beaty Water Research Centre, Department of Civil Engineering, Union Street, Queen's University, Kingston, K7L 3Z6, Canada
| | - Yves Filion
- Beaty Water Research Centre, Department of Civil Engineering, Union Street, Queen's University, Kingston, K7L 3Z6, Canada.
| | - Pascale Champagne
- Beaty Water Research Centre, Department of Civil Engineering, Union Street, Queen's University, Kingston, K7L 3Z6, Canada; Institut National de la Recherche Scientifique (INRS), 490 rue de la Couronne, Québec City, Québec, G1K 9A9, Canada
| |
Collapse
|
43
|
Bjarnsholt T, Whiteley M, Rumbaugh KP, Stewart PS, Jensen PØ, Frimodt-Møller N. The importance of understanding the infectious microenvironment. THE LANCET. INFECTIOUS DISEASES 2021; 22:e88-e92. [PMID: 34506737 DOI: 10.1016/s1473-3099(21)00122-5] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 01/25/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Standard doses of antibiotics do not efficiently treat chronic infections of the soft tissue and bone. In this Personal View, we advocate for improving treatment of these infections by taking the infectious microenvironment into account. The infectious microenvironment can cause sensitive bacteria to lose their susceptibility to antibiotics that are effective in standard laboratory susceptibility testing. We propose that bacteria behave substantially different in standard laboratory conditions than they do in actual infections. The infectious microenvironment could impose changes in growth and metabolic activity that result in increased protection against antibiotics. Therefore, we advocate that improved antibiotic treatment of chronic infection is achievable when antibiotics are recommended on the basis of susceptibility testing in relevant in vitro conditions that resemble actual infectious microenvironments. We recommend establishing knowledge of the relevant conditions of the chemical and physical composition of the infectious microenvironment. Recent advances in RNA sequencing, metabolomics, and microscopy have made it possible for the characterisation of the microenvironment of infections and to validate the clinical relevance of in vitro conditions to actual infections.
Collapse
Affiliation(s)
- Thomas Bjarnsholt
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark; Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| | - Marvin Whiteley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA; Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA, USA; Emory-Children's Cystic Fibrosis Center, Atlanta, GA, USA
| | - Kendra P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Peter Ø Jensen
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark; Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Frimodt-Møller
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
44
|
Kosztołowicz T, Metzler R. Diffusion of antibiotics through a biofilm in the presence of diffusion and absorption barriers. Phys Rev E 2021; 102:032408. [PMID: 33075880 DOI: 10.1103/physreve.102.032408] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022]
Abstract
We propose a model of antibiotic diffusion through a bacterial biofilm when diffusion and/or absorption barriers develop in the biofilm. The idea of this model is: We deduce details of the diffusion process in a medium in which direct experimental study is difficult, based on probing diffusion in external regions. Since a biofilm has a gel-like consistency, we suppose that subdiffusion of particles in the biofilm may occur. To describe this process we use a fractional subdiffusion-absorption equation with an adjustable anomalous diffusion exponent. The boundary conditions at the boundaries of the biofilm are derived by means of a particle random walk model on a discrete lattice leading to an expression involving a fractional time derivative. We show that the temporal evolution of the total amount of substance that has diffused through the biofilm explicitly depends on whether there is antibiotic absorption in the biofilm. This fact is used to experimentally check for antibiotic absorption in the biofilm and if subdiffusion and absorption parameters of the biofilm change over time. We propose a four-stage model of antibiotic diffusion in biofilm based on the following physical characteristics: whether there is absorption of the antibiotic in the biofilm and whether all biofilm parameters remain unchanged over time. The biological interpretation of the stages, in particular their relation with the bacterial defense mechanisms, is discussed. Theoretical results are compared with empirical results of ciprofloxacin diffusion through Pseudomonas aeruginosa biofilm, and ciprofloxacin and gentamicin diffusion through Proteus mirabilis biofilm.
Collapse
Affiliation(s)
- Tadeusz Kosztołowicz
- Institute of Physics, Jan Kochanowski University, Uniwersytecka 7, 25-406 Kielce, Poland
| | - Ralf Metzler
- Institute for Physics and Astronomy, University of Potsdam, D-14476, Potsdam-Golm, Germany
| |
Collapse
|
45
|
Modifying TIMER to generate a slow-folding DsRed derivative for optimal use in quickly-dividing bacteria. PLoS Pathog 2021; 17:e1009284. [PMID: 34214139 PMCID: PMC8291646 DOI: 10.1371/journal.ppat.1009284] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 07/20/2021] [Accepted: 06/14/2021] [Indexed: 11/29/2022] Open
Abstract
It is now well appreciated that members of pathogenic bacterial populations exhibit heterogeneity in growth rates and metabolic activity, and it is known this can impact the ability to eliminate all members of the bacterial population during antibiotic treatment. It remains unclear which pathways promote slowed bacterial growth within host tissues, primarily because it has been difficult to identify and isolate slow growing bacteria from host tissues for downstream analyses. To overcome this limitation, we have developed a novel variant of TIMER, a slow-folding fluorescent protein, named DsRed42, to identify subsets of slowly dividing bacteria within host tissues. The original TIMER folds too slowly for fluorescence accumulation in quickly replicating bacterial species (Escherichia coli, Yersinia pseudotuberculosis), however DsRed42 accumulates red fluorescence in late stationary phase cultures of E. coli and Y. pseudotuberculosis. We show DsRed42 signal also accumulates during exposure to sources of nitric oxide (NO), suggesting DsRed42 signal detects growth-arrested bacterial cells. In a mouse model of Y. pseudotuberculosis deep tissue infection, DsRed42 signal was detected, and primarily accumulates in bacteria expressing markers of stationary phase growth. There was no significant overlap between DsRed42 signal and NO-exposed subpopulations of bacteria within host tissues, suggesting NO stress was transient, allowing bacteria to recover from this stress and resume replication. This novel DsRed42 variant represents a tool that will enable additional studies of slow-growing subpopulations of bacteria, specifically within bacterial species that quickly divide. We have generated a variant of TIMER that can be used to mark slow-growing subsets of Yersinia pseudotuberculosis, which has a relatively short division time, similar to E. coli. We used a combination of site-directed and random mutagenesis to generate DsRed42, which has red fluorescent signal accumulation in post-exponential or stationary phase cells. Since this variant accumulates only red fluorescence, it is no longer a TIMER protein, and is more appropriately termed DsRed42. We found that nitric oxide (NO) stress is sufficient to promote DsRed42 signal accumulation in culture, however within host tissues, DsRed42 signal correlates with a stationary phase reporter (dps). These results suggest NO may cause an immediate arrest in bacterial cell division, but during growth in host tissues exposure to NO is transient, allowing bacteria to recover from this stress and resume cell division. Thus instead of indicating a response to host stressors, DsRed42 signal accumulation within host tissues appears to identify slow-growing cells that are experiencing nutrient limitation.
Collapse
|
46
|
Di Bonaventura G, Pompilio A. In Vitro Antimicrobial Susceptibility Testing of Biofilm-Growing Bacteria: Current and Emerging Methods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1369:33-51. [PMID: 33963526 DOI: 10.1007/5584_2021_641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The antibiotic susceptibility of bacterial pathogens is typically determined based on planktonic cells, as recommended by several international guidelines. However, most of chronic infections - such as those established in wounds, cystic fibrosis lung, and onto indwelling devices - are associated to the formation of biofilms, communities of clustered bacteria attached onto a surface, abiotic or biotic, and embedded in an extracellular matrix produced by the bacteria and complexed with molecules from the host. Sessile microorganisms show significantly increased tolerance/resistance to antibiotics compared with planktonic counterparts. Consequently, antibiotic concentrations used in standard antimicrobial susceptibility tests, although effective against planktonic bacteria in vitro, are not predictive of the concentrations required to eradicate biofilm-related infections, thus leading to treatment failure, chronicization and removal of material in patients with indwelling medical devices.Meeting the need for the in vitro evaluation of biofilm susceptibility to antibiotics, here we reviewed several methods proposed in literature highlighting their advantages and limitations to guide scientists towards an appropriate choice.
Collapse
Affiliation(s)
- Giovanni Di Bonaventura
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy. .,Laboratory of Clinical Microbiology, Chieti, Italy.
| | - Arianna Pompilio
- Department of Medical, Oral and Biotechnological Sciences, and Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Laboratory of Clinical Microbiology, Chieti, Italy
| |
Collapse
|
47
|
Thaarup IC, Bjarnsholt T. Current In Vitro Biofilm-Infected Chronic Wound Models for Developing New Treatment Possibilities. Adv Wound Care (New Rochelle) 2021; 10:91-102. [PMID: 32496982 DOI: 10.1089/wound.2020.1176] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Significance: The prevalence of chronic wounds is increasing worldwide. The most recent estimates suggest that up to 2% of the population in the industrialized countries is affected. Recent Advances: During the past few decades, bacterial biofilms have been elucidated as one of the primary reasons why chronic wounds fail to heal. Critical Issues: There is a lack of direct causation and evidence of the role that biofilms play in persistent wounds, which complicates research on new treatment options, since it is still unknown which factors dominate. For this reason, several different in vitro wound models that mimic the biofilm infections observed in chronic wounds and other chronic infections have been created. These different models are, among other purposes, used to test a variety of wound care products. However, chronic wounds are highly complex, and several different factors must be taken into consideration along with the infection, including physiochemical and human-supplemented factors. Furthermore, the limitations of using in vitro models, such as the lack of a responsive immune system should always be given due consideration. Future Directions: Present understandings of all the elements and interactions that take place within chronic wounds are incomplete. As our insight of in vivo chronic wounds continues to expand, so too must the in vitro models used to mimic these infections evolve and adapt to new knowledge.
Collapse
Affiliation(s)
- Ida C. Thaarup
- Department of Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
48
|
Pharmacokinetic and Pharmacodynamic Optimization of Antibiotic Therapy in Cystic Fibrosis Patients: Current Evidences, Gaps in Knowledge and Future Directions. Clin Pharmacokinet 2021; 60:409-445. [PMID: 33486720 DOI: 10.1007/s40262-020-00981-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
Antibiotic therapy is one of the main treatments for cystic fibrosis (CF). It aims to eradicate bacteria during early infection, calms down the inflammatory process, and leads to symptom resolution of pulmonary exacerbations. CF can modify both the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of antibiotics, therefore specific PK/PD endpoints should be determined in the context of CF. Currently available data suggest that optimal PK/PD targets cannot be attained in sputum with intravenous aminoglycosides. Continuous infusion appears preferable for β-lactam antibiotics, but optimal concentrations in sputum are unlikely to be reached, with some possible exceptions such as meropenem and ceftolozane. Usual doses are likely suboptimal for fluoroquinolones and linezolid, whereas daily doses of 45-60 mg/kg and 200 mg could be convenient for vancomycin and doxycycline, respectively. Weekly azithromycin doses of 22-30 mg/kg could also be appropriate for its anti-inflammatory effect. The difficulty with achieving optimal concentrations supports the use of combined treatments and the inhaled administration route, as very high local concentrations, concomitantly with low systemic exposure, can be obtained with the inhaled route for aminoglycosides, colistin, and fluoroquinolones, thus minimizing the risk of toxicity.
Collapse
|
49
|
Dostert M, Trimble MJ, Hancock REW. Antibiofilm peptides: overcoming biofilm-related treatment failure. RSC Adv 2021; 11:2718-2728. [PMID: 35424252 PMCID: PMC8694000 DOI: 10.1039/d0ra09739j] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022] Open
Abstract
Health leaders and scientists worldwide consider antibiotic resistance among the world's most dangerous pathogens as one of the biggest threats to global health. Antibiotic resistance has largely been attributed to genetic changes, but the role and recalcitrance of biofilms, largely due to growth state dependent adaptive resistance, is becoming increasingly appreciated. Biofilms are mono- and multi-species microbial communities embedded in an extracellular, protective matrix. In this growth state, bacteria are transcriptionally primed to survive extracellular stresses. Adaptations, affecting metabolism, regulation, surface charge, immune recognition and clearance, allow bacteria to thrive in the human body and withstand antibiotics and the host immune system. Biofilms resist clearance by multiple antibiotics and have a major role in chronic infections, causing more than 65% of all infections. No specific antibiofilm agents have been developed. Thus, there is a pressing need for alternatives to traditional antibiotics that directly inhibit and/or eradicate biofilms. Host defence peptides (HDPs) are small cationic peptides that are part of the innate immune system to both directly kill microbes but also function to modulate the immune response. Specific HDPs and their derivatives demonstrate broad-spectrum activity against biofilms. In vivo biofilm assays show efficacy in abscess, respiratory, in-dwelling device, contact lens and skin infection models. Further progress has been made through the study of ex vivo organoid and air-liquid interface models to better understand human infections and treatment while relieving the burden and complex nature of animal models. These avenues pave the way for a better understanding and treatment of the underlying cause of chronic infections that challenge the healthcare system.
Collapse
Affiliation(s)
- Melanie Dostert
- Department of Microbiology and Immunology, University of British Columbia Vancouver British Columbia Canada
| | - Michael J Trimble
- Department of Microbiology and Immunology, University of British Columbia Vancouver British Columbia Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
50
|
Brilhante RSN, Fernandes MR, Pereira VS, Costa ADC, Oliveira JSD, de Aguiar L, Rodrigues AM, de Camargo ZP, Pereira-Neto WA, Sidrim JJC, Rocha MFG. Biofilm formation on cat claws by Sporothrix species: An ex vivo model. Microb Pathog 2020; 150:104670. [PMID: 33285221 DOI: 10.1016/j.micpath.2020.104670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022]
Abstract
This work aimed to evaluate the ability of Sporothrix species to attach and form biofilm on the surface of cat claws as an ex vivo model. A total of 14 strains (5 Sporothrix brasiliensis, 3 Sporothrix schenckii s. str., 3 Sporothrix globosa and 3 Sporothrix mexicana) were used. The biofilms were incubated for periods of 01, 03, 07, 10 and fifteenth 15 days. Their metabolic activities were evaluated by the XTT reduction assay and the morphology and structure were investigated by scanning electron microscopy (SEM). The analysis of the SEM images revealed that all the species can form biofilms on cat claws. The metabolic activity in the ex vivo biofilms was similar to that found in in vitro biofilms when incubated for the same period. This is the first report of an ex vivo biofilm model involving cat claws. The ability to form biofilms on cat claws can increase the viable period of the fungus and consequently the number of possibly infected animals and people.
Collapse
Affiliation(s)
- Raimunda Sâmia Nogueira Brilhante
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil
| | - Mirele Rodrigues Fernandes
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil
| | - Vandbergue Santos Pereira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil
| | - Anderson da Cunha Costa
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil
| | - Jonathas Sales de Oliveira
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil
| | - Lara de Aguiar
- Postgraduate in Veterinary Sciences, Faculty of Veterinary, State University of Ceará. Dr. Silas Munguba Avenue, 1700, Itaperi Campus, 60714-903, Fortaleza, Ceará, Brazil
| | - Anderson Messias Rodrigues
- Cellular Biology Division, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Botucatu Street, 862 - 04023-062, Sao Paulo, Sao Paulo, Brazil
| | - Zoilo Pires de Camargo
- Cellular Biology Division, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, Botucatu Street, 862 - 04023-062, Sao Paulo, Sao Paulo, Brazil
| | - Waldemiro Aquino Pereira-Neto
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil
| | - José Júlio Costa Sidrim
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil.
| | - Marcos Fábio Gadelha Rocha
- Specialized Medical Mycology Center, Postgraduate Program in Medical Microbiology, Department of Pathology and Forensic Medicine, Federal University of Ceará, Coronel Nunes de Melo Street, 1315 - Rodolfo Teófilo - 60430-275, Fortaleza, Ceará, Brazil; Postgraduate in Veterinary Sciences, Faculty of Veterinary, State University of Ceará. Dr. Silas Munguba Avenue, 1700, Itaperi Campus, 60714-903, Fortaleza, Ceará, Brazil
| |
Collapse
|