1
|
Dong J, Zhang S, Chan YK, Lai S, Deng Y. Vacancies-rich Z-scheme VdW heterojunction as H 2S-sensitized synergistic therapeutic nanoplatform against refractory biofilm infections. Biomaterials 2025; 320:123258. [PMID: 40090255 DOI: 10.1016/j.biomaterials.2025.123258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/02/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Encapsulated in a self-produced negatively charged extracellular polymeric substance (EPS) matrix, the wound infected bacterial biofilms exhibit formidable resistance to conventional positively charged antibiotics and host's immune responses, which can undoubtedly lead to persistent infections and lethal complications. Nevertheless, developing efficacious strategies to root out stubborn biofilm and promote tissue regeneration still remains a challenge. To resolve this dilemma, a versatile vacancies-rich Z-scheme MoSSe Van der Waals heterojunction (MoSSe VdW HJ) is rationally fabricated as nanoplatform for hydrogen sulfide (H2S)-sensitized synergistic therapy of wound bacterial biofilm infection. The rich anion vacancies and Z-scheme heterostructure make the fabricated MoSSe VdW HJ can effectively augment H2S, localized hyperthermia, and reactive oxygen species production under the stimulation of biofilm microenvironments (BME) and irradiation of 808 nm near-infrared (NIR) light. Therefore, MoSSe VdW HJ is capable to integrate H2S gas, chemodynamic, photothermal, and photodynamic therapies to effectively destroy eDNA and polysaccharides in the EPS matrix, thereby breaching the biofilm barrier to eradicate bacteria and facilitate wound healing. The synergistic strategy exhibits superior anti-biofilm and wound repair effects both in vivo and in vitro, thus providing guideline for the development of BME and NIR light activated synergistic therapeutics to fight against refractory biofilm infections.
Collapse
Affiliation(s)
- Jianwen Dong
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Shuting Zhang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077, Hong Kong, China
| | - Shuangquan Lai
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China.
| | - Yi Deng
- School of Chemical Engineering, Sichuan University, Chengdu 610065, China; Department of Mechanical Engineering, The University of Hong Kong, 999077, Hong Kong, China.
| |
Collapse
|
2
|
Mardalisa, Wang R, Sabar MA, Matsuura N, Hara-Yamamura H, Honda R. Different fates between extracellular and intracellular antimicrobial resistome in full-scale activated sludge and membrane bioreactor processes. WATER RESEARCH 2025; 274:123155. [PMID: 39854777 DOI: 10.1016/j.watres.2025.123155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/25/2024] [Accepted: 01/15/2025] [Indexed: 01/26/2025]
Abstract
Treated effluent of wastewater treatment plants (WWTPs) are major sources of extracellular antimicrobial resistance genes (eARGs) into aquatic environments. This study aimed to clarify the fate and origins of eARGs from influent to treated effluent at a full-scale WWTP. The compositions of eARG and intracellular ARG (iARG) were acquired via shotgun metagenomic sequencing in influent wastewater, activated sludge, and treated effluent of the target WWTP, where identical wastewater was treated by conventional activated sludge (CAS) and membrane bioreactor (MBR) processes. The proportion of eARGs to iARGs increased from influent to effluent in both processes, reaching almost half of the total ARG. Most eARGs in influent were associated with clinically important antimicrobials, whereas eARGs in sludge and effluent were dominated by aminoglycoside resistance genes of aadA and APH variants. Although the eARGs composition in influent wastewater mirrored that of iARGs, a substantial shift occurred in activated sludge and effluent, highlighting the presence of distinct dissemination and reduction mechanisms between eARGs and iARGs. Notably, the origin of eARGs in treated effluent was mainly iARGs in the effluent rather than the carryover of eARG from activated sludge, which were substantially reduced in MBR, compared to CAS. Consequently, these differences in selective mechanisms led to different fates between eARG and iARG during wastewater treatment.
Collapse
Affiliation(s)
- Mardalisa
- Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa 920-1192, Japan; Department of Marine Science, Faculty of Fishery and Marine Science, Riau University, Riau 28293, Indonesia
| | - Rongxuan Wang
- Asia-Japan Research Institute, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Muhammad Adnan Sabar
- Department of Environmental and Sustainable Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| | - Norihisa Matsuura
- Faculty of Geosciences and Civil Engineering, Kanazawa University, Kanazawa 920-1192, Japan
| | - Hiroe Hara-Yamamura
- Faculty of Geosciences and Civil Engineering, Kanazawa University, Kanazawa 920-1192, Japan
| | - Ryo Honda
- Faculty of Geosciences and Civil Engineering, Kanazawa University, Kanazawa 920-1192, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, 565-0871, Japan.
| |
Collapse
|
3
|
Ge T, Wu R, Yu T, Hasan MSU, Liu J. Halogen anion modulated metal-organic frameworks with enhanced nanozyme activities for bacterial biofilm disruption. NANOSCALE 2025. [PMID: 40145893 DOI: 10.1039/d5nr00131e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
There is an urgent need to develop new nanozymes with enhanced catalytic activities to combat bacterial infections, which have become increasingly challenging due to the misuse of antibiotics and the difficulties of new antibiotic discovery. Here, we employed a new strategy against bacterial biofilms by introducing halide anions to modulate the crystal facets of ZIF-L metal-organic frameworks (MOFs) and then loading chloroquine to form Ch@ZIF-L. The modulation of crystal facets significantly enhanced the oxidase activities of ZIF-L, which can be significantly changed by modulation of its crystal facets, with the hexagonal ZIF-L (ZIF-L-H-Cl) structure showing the highest oxidase activity. At pH 6.0, over 80% of chloroquine was released from Ch@ZIF-L-H-Cl within 8 hours, altering the DNA conformation of bacterial biofilms and disrupting the extracellular polymeric substances (EPSs). The generation of singlet oxygen catalyzed by ZIF-L-H-Cl can effectively kill bacteria at the infected wound site. The composite nanozyme of Ch@ZIF-L-H-Cl, when treated at 100 μg mL-1, exhibited no adverse effects on normal cell growth or hemolysis. Our in vivo experiments demonstrated an 85% reduction of the wound area by day 8 and a rapid recovery of body weight in mice with wounds infected with Staphylococcus aureus (S. aureus) biofilms. Furthermore, substantial reductions in bacterial counts were observed in both wounds and blood samples in the mice, highlighting the great potential of Ch@ZIF-L-H-Cl in combating bacterial biofilm infections.
Collapse
Affiliation(s)
- Tianjin Ge
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Renfei Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Tianrong Yu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Muhammad Sajjad Ul Hasan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| | - Jian Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou 215123, Jiangsu, P. R. China.
| |
Collapse
|
4
|
Zhang N, Li X, Liu X, Cheng P, Li L, Chai Y, Cao M, Yang Y. Aspirin enhances the antibacterial activity of colistin against multidrug-resistant Pseudomonas aeruginosa. Eur J Pharmacol 2025; 997:177480. [PMID: 40057155 DOI: 10.1016/j.ejphar.2025.177480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Multidrug-resistant (MDR) Pseudomonas aeruginosa (PSA), recently reclassified by the World Health Organization (WHO) as a high-priority antimicrobial-resistant pathogen, continues to impose a substantial global health burden due to escalating resistance and stagnant therapeutic innovation. Colistin retains critical clinical utility against MDR P. aeruginosa infections; however, its dose-limiting nephrotoxicity and neurotoxicity necessitate strategies to optimise therapeutic indices. This study investigated the molecular mechanism underlying the synergistic activity of aspirin in potentiating colistin efficacy against MDR P. aeruginosa. In vitro analyses revealed marked synergistic bactericidal activity (FIC index ≤0.5), with metabolomic profiling demonstrating suppression of key metabolic pathways integral to bacterial membrane biogenesis, including glycerophospholipid metabolism and fatty acid biosynthesis. Ultrastructural imaging confirmed irreversible disruption of membrane integrity via combined treatment. In a rat model of P. aeruginosa-induced pneumonia, colistin-aspirin co-administration demonstrated superior efficacy to monotherapy, significantly reducing pulmonary bacterial load (3 to 4-log CFU/g reduction vs colistin alone; p < 0.01), attenuating histopathological injury, and suppressing pro-inflammatory cytokine levels (IL-6, IL-8, TNF-α) by 30-47%. Critically, this synergy enabled a reduction of colistin dosing to one-sixteenth while maintaining bactericidal potency. These findings provide mechanistic insights into aspirin-mediated colistin sensitisation and evidence supporting combinatorial regimens to circumvent colistin toxicity barriers. This work establishes a rational foundation for clinical translation of repurposed aspirin-colistin therapy against MDR P. aeruginosa infections.
Collapse
Affiliation(s)
- Ning Zhang
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; College of Acupuncture and Massage, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China; First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Xue Li
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; Department of the Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550003, Guiyang, China
| | - Xin Liu
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Ping Cheng
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Lailai Li
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yihui Chai
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Mingle Cao
- People's Hospital of Duyun City, Duyun, 558000, Guizhou, China
| | - Yuqi Yang
- Guizhou University of Traditional Chinese Medicine, 550025, Guiyang, China; School of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
5
|
You J, Ye L, Zhang S, Zhao J, Zhao Y, He Y, Chen J, Kennes C, Chen D. Electrode functional microorganisms in bioelectrochemical systems and its regulation: A review. Biotechnol Adv 2025; 79:108521. [PMID: 39814087 DOI: 10.1016/j.biotechadv.2025.108521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 12/03/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Bioelectrochemical systems (BES) as environmental remediation biotechnologies have boomed in the last two decades. Although BESs combined technologies with electro-chemistry, -biology, and -physics, microorganisms and biofilms remain at their core. In this review, various functional microorganisms in BESs for CO2 reduction, dehalogenation, nitrate, phosphate, and sulfate reduction, metal removal, and volatile organic compound oxidation are summarized and compared in detail. Moreover, interrelationship regulation approaches for functional microorganisms and methods for electroactive biofilm development, such as targeted electrode surface modification, chemical treatment, physical revealing, biological optimization, and genetic programming are pointed out. This review provides promising guidance and suggestions for the selection of microbial inoculants and provides an analysis of the role of individual microorganisms in mixed microbial communities and its metabolisms.
Collapse
Affiliation(s)
- Juping You
- Zhejiang Key Laboratory of Pullution control for Port-Petrochemical Industry, Zhejiang Ocean University, Zhoushan 316022, China; Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Zhejiang Shuren University, Hangzhou 312028, China
| | - Lei Ye
- Zhejiang Key Laboratory of Pullution control for Port-Petrochemical Industry, Zhejiang Ocean University, Zhoushan 316022, China
| | - Shihan Zhang
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jingkai Zhao
- College of Environment, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yan Zhao
- Zhejiang Key Laboratory of Pullution control for Port-Petrochemical Industry, Zhejiang Ocean University, Zhoushan 316022, China
| | - Yaxue He
- Zhejiang Key Laboratory of Pullution control for Port-Petrochemical Industry, Zhejiang Ocean University, Zhoushan 316022, China
| | - Jianmeng Chen
- School of Environment and Natural Resources, Zhejiang University of Science and Technology, Hangzhou 310018, China
| | - Christian Kennes
- Chemical Engineering Laboratory and Center for Advance Scientific Research (CICA), Faculty of Sciences, Universidade da Coruña, Spain
| | - Dongzhi Chen
- Zhejiang Key Laboratory of Pullution control for Port-Petrochemical Industry, Zhejiang Ocean University, Zhoushan 316022, China.
| |
Collapse
|
6
|
Ettadili H, Vural C. Bibliometric Analysis and Network Visualization of Nanozymes for Microbial Theranostics in the Last Decade. Appl Biochem Biotechnol 2025; 197:1923-1945. [PMID: 39625609 DOI: 10.1007/s12010-024-05120-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 03/29/2025]
Abstract
Nanozymes are a class of nanomaterials that are capable of mimicking the activities of natural enzymes. They are currently receiving considerable attention due to their advantageous properties. The objective of this study is to provide a comprehensive analysis of the advancements and trends in nanozymes for microbial theranostics research over the past decade through a detailed bibliometric approach. For this purpose, an effective search query was formulated, and relevant publications from 2013 to 2023 were collected from the Web of Science Core Collection database. Subsequently, the following softwares were employed for analysis: VOSviewer, the Bibliometrix R package, and GraphPad Prism 8.0.2. The findings revealed a statistically significant positive correlation (r = 0.993; p < 0.0001) between publications and citations, in addition to an important growth rate of scientific output of approximately 28.90%. China, India, and the USA were the most productive countries, whereas progress in low- and middle-income countries remained constrained. The Chinese Academy of Sciences was the most productive institution, and remarkably almost the top 10 productive authors were from China. Regarding keywords analysis, current research hotspots are primarily concentrated on the application of nanozymes in anti-biofilm-related research, antibacterial activity and therapy, the development of biosensors for microbial detection and control, and the advancement of wound disinfection and therapy.
Collapse
Affiliation(s)
- Hamza Ettadili
- Faculty of Science, Department of Biology, Molecular Biology Section, Pamukkale University, 20160, Denizli, Türkiye
| | - Caner Vural
- Faculty of Science, Department of Biology, Molecular Biology Section, Pamukkale University, 20160, Denizli, Türkiye.
| |
Collapse
|
7
|
Geng RSQ, Sibbald RG, Slomovic J, Toksarka O, Schultz G. Therapeutic Indices of Topical Antiseptics in Wound Care: A Systematic Review. Adv Skin Wound Care 2025; 38:10-18. [PMID: 39355996 DOI: 10.1097/asw.0000000000000233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
GENERAL PURPOSE To review the therapeutic indices of topical antiseptics for bacterial species commonly isolated from chronic wounds. TARGET AUDIENCE This continuing education activity is intended for physicians, physician assistants, nurse practitioners, and registered nurses with an interest in skin and wound care. LEARNING OBJECTIVES/OUTCOMES After participating in this educational activity, the participant will:1. Explain the mechanisms underlying chronic wound physiology and their implications for effective wound healing and management.2. Evaluate the role of therapeutic index values for topical antiseptics in chronic wound management.3. Apply evidence-based treatment strategies for chronic wound management.
Collapse
|
8
|
Agarwal H, Gurnani B, Pippal B, Jain N. Capturing the micro-communities: Insights into biogenesis and architecture of bacterial biofilms. BBA ADVANCES 2024; 7:100133. [PMID: 39839441 PMCID: PMC11750278 DOI: 10.1016/j.bbadva.2024.100133] [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] [Indexed: 01/23/2025] Open
Abstract
Biofilm is an assemblage of microorganisms embedded within the extracellular matrix that provides mechanical stability, nutrient absorption, antimicrobial resistance, cell-cell interactions, and defence against host immune system. Various biomolecules such as lipids, carbohydrates, protein polymers (amyloid), and eDNA are present in the matrix playing significant role in determining the distinctive properties of biofilm. The formation of biofilms contributes to resistance against antimicrobial therapy in most of the human infections and exacerbates existing diseases. Therefore, this field requires several state-of-the-art techniques to fully understand the 3-D organization of biofilms, their cell behaviour and responses to pharmaceutical treatments. Here, we explore the assembly and regulation of biofilm biogenesis in the context of matrix components and highlight the significance of high-resolution imaging and analysing techniques for monitoring complex biofilm architecture. Our review also emphasizes the novelty and advancements in techniques to visualise biofilm structure and composition, providing valuable insights to understand biofilm-related infections.
Collapse
Affiliation(s)
- Harshita Agarwal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Bharat Gurnani
- Centre of Excellence-AyurTech, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Bhumika Pippal
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| | - Neha Jain
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Nagaur Road, Karwar, Rajasthan 342037, India
| |
Collapse
|
9
|
Salikin NH, Keong LC, Azemin WA, Philip N, Yusuf N, Daud SA, Rashid SA. Combating multidrug-resistant (MDR) Staphylococcus aureus infection using terpene and its derivative. World J Microbiol Biotechnol 2024; 40:402. [PMID: 39627623 DOI: 10.1007/s11274-024-04190-w] [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: 07/24/2024] [Accepted: 11/01/2024] [Indexed: 12/13/2024]
Abstract
Multidrug-resistant (MDR) Staphylococcus aureus represents a major global health issue resulting in a wide range of debilitating infections and fatalities. The slow progression of new antibiotics, limited choices for treatment, and scarcity of new drug approvals create immense obstacles in new drug line development. S. aureus poses a significant public health risk, due to the emergence of methicillin-resistant (MRSA) and vancomycin-resistant strains (VRSA), necessitating novel antibiotics for effective control management. Current studies are delving into the terpenes' potential as an antimicrobial agent, indicating positive prospects as promising substitutes or complementary to conventional antibiotics. Concurrent reactions of terpenes with conventional antibiotics create synergistic effects that significantly enhance antibiotic efficacy. Accumulated evidence has shown that while efflux pump (e.g., NorA, TetK, and MepA) is revealed as an essential defense of S. aureus against antibiotics, terpene and its derivative act as its potent inhibitor, suggesting the promising potential of terpenes in combating those infectious pathogens. Furthermore, pronounced cell membrane disruptive activity and antibiofilm properties by terpenes have been exerted, signifying their significance as promising prevention against microbial pathogenesis and antimicrobial resistance. This review provides an overview of the potential of terpenes and their derivatives in combating S. aureus infections, highlighting their potential mechanisms of action (MOA), synergistic effects with conventional antibiotics, and challenges in clinical translation. The unique properties of terpenes offer an opportunity for their use in developing an exceptional defense strategy against antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Nor Hawani Salikin
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Lee Chee Keong
- School of Industrial Technology, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Wan-Atirah Azemin
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Noraini Philip
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Nurhaida Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Syiah Kuala University, Aceh, Indonesia
| | - Siti Aisyah Daud
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia
| | - Syarifah Ab Rashid
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden Pulau Pinang, Malaysia.
| |
Collapse
|
10
|
Burke Ó, Zeden MS, O'Gara JP. The pathogenicity and virulence of the opportunistic pathogen Staphylococcus epidermidis. Virulence 2024; 15:2359483. [PMID: 38868991 DOI: 10.1080/21505594.2024.2359483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/19/2024] [Indexed: 06/14/2024] Open
Abstract
The pervasive presence of Staphylococcus epidermidis and other coagulase-negative staphylococci on the skin and mucous membranes has long underpinned a casual disregard for the infection risk that these organisms pose to vulnerable patients in healthcare settings. Prior to the recognition of biofilm as an important virulence determinant in S. epidermidis, isolation of this microorganism in diagnostic specimens was often overlooked as clinically insignificant with potential delays in diagnosis and onset of appropriate treatment, contributing to the establishment of chronic infection and increased morbidity or mortality. While impressive progress has been made in our understanding of biofilm mechanisms in this important opportunistic pathogen, research into other virulence determinants has lagged S. aureus. In this review, the broader virulence potential of S. epidermidis including biofilm, toxins, proteases, immune evasion strategies and antibiotic resistance mechanisms is surveyed, together with current and future approaches for improved therapeutic interventions.
Collapse
Affiliation(s)
- Órla Burke
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | | | - James P O'Gara
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
11
|
Nielsen SM, Johnsen KK, Hansen LBS, Rikvold PD, Møllebjerg A, Palmén LG, Durhuus T, Schlafer S, Meyer RL. Large-scale screening identifies enzyme combinations that remove in situ grown oral biofilm. Biofilm 2024; 8:100229. [PMID: 39830521 PMCID: PMC11740801 DOI: 10.1016/j.bioflm.2024.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/22/2024] [Accepted: 10/03/2024] [Indexed: 01/22/2025] Open
Abstract
Bacteria in the oral cavity are responsible for the development of dental diseases such as caries and periodontitis, but it is becoming increasingly clear that the oral microbiome also benefits human health. Many oral care products on the market are antimicrobial, killing a large part of the oral microbiome but without removing the disease-causing biofilm. Instead, non-biocidal matrix-degrading enzymes may be used to selectively remove biofilm without harming the overall microbiome. The challenge of using enzymes to degrade biofilms is to match the narrow specificity of enzymes with the large structural diversity of extracellular polymeric substances that hold the biofilm together. In this study, we therefore perform a large-scale screening of single and multi-enzyme formulations to identify combinations of enzymes that most effectively remove dental biofilm. We tested >400 different treatment modalities using 44 different enzymes in combinations with up to six enzymes in each formulation, on in vitro biofilms inoculated with human saliva. Mutanase was the only enzyme capable of removing biofilm on its own. Multi-enzyme formulations removed up to 69 % of the biofilm volume, and the most effective formulations all contained mutanase. We shortlisted 10 enzyme formulations to investigate their efficacy against biofilms formed on glass slabs on dental splints worn by 9 different test subjects. Three of the ten formulations removed more than 50 % of the biofilm volume. If optimal enzyme concentration and exposure time can be reached in vivo, these enzyme combinations have potential to be used in novel non-biocidal oral care products for dental biofilm control.
Collapse
Affiliation(s)
- Signe Maria Nielsen
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
- Novonesis A/S, Biologiens Vej 2, 2800, Kgs. Lyngby, Denmark
| | - Karina Kambourakis Johnsen
- Section for Oral Ecology, Cariology, Department of Dentistry and Oral Health, Aarhus University, Vennelyst Boulevard 9, 8000, Aarhus C, Denmark
| | | | - Pernille Dukanovic Rikvold
- Section for Oral Ecology, Cariology, Department of Dentistry and Oral Health, Aarhus University, Vennelyst Boulevard 9, 8000, Aarhus C, Denmark
- Novonesis A/S, Biologiens Vej 2, 2800, Kgs. Lyngby, Denmark
| | - Andreas Møllebjerg
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
- Novonesis A/S, Biologiens Vej 2, 2800, Kgs. Lyngby, Denmark
| | | | - Thomas Durhuus
- Novonesis A/S, Biologiens Vej 2, 2800, Kgs. Lyngby, Denmark
| | - Sebastian Schlafer
- Section for Oral Ecology, Cariology, Department of Dentistry and Oral Health, Aarhus University, Vennelyst Boulevard 9, 8000, Aarhus C, Denmark
- Department of Biology, Faculty of Natural Sciences, Aarhus University, Ny Munkegade 114, 8000, Aarhus C, Denmark
| | - Rikke Louise Meyer
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
- Department of Biology, Faculty of Natural Sciences, Aarhus University, Ny Munkegade 114, 8000, Aarhus C, Denmark
| |
Collapse
|
12
|
Xi Y, Li X, Liu L, Xiu F, Yi X, Chen H, You X. Sneaky tactics: Ingenious immune evasion mechanisms of Bartonella. Virulence 2024; 15:2322961. [PMID: 38443331 PMCID: PMC10936683 DOI: 10.1080/21505594.2024.2322961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
Gram-negative Bartonella species are facultative intracellular bacteria that can survive in the harsh intracellular milieu of host cells. They have evolved strategies to evade detection and degradation by the host immune system, which ensures their proliferation in the host. Following infection, Bartonella alters the initial immunogenic surface-exposed proteins to evade immune recognition via antigen or phase variation. The diverse lipopolysaccharide structures of certain Bartonella species allow them to escape recognition by the host pattern recognition receptors. Additionally, the survival of mature erythrocytes and their resistance to lysosomal fusion further complicate the immune clearance of this species. Certain Bartonella species also evade immune attacks by producing biofilms and anti-inflammatory cytokines and decreasing endothelial cell apoptosis. Overall, these factors create a challenging landscape for the host immune system to rapidly and effectively eradicate the Bartonella species, thereby facilitating the persistence of Bartonella infections and creating a substantial obstacle for therapeutic interventions. This review focuses on the effects of three human-specific Bartonella species, particularly their mechanisms of host invasion and immune escape, to gain new perspectives in the development of effective diagnostic tools, prophylactic measures, and treatment options for Bartonella infections.
Collapse
Affiliation(s)
- Yixuan Xi
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Xinru Li
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Lu Liu
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Feichen Xiu
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Xinchao Yi
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| | - Hongliang Chen
- Chenzhou NO.1 People’s Hospital, The Affiliated Chenzhou Hospital, Hengyang Medical College, University of South China, ChenZhou, China
| | - Xiaoxing You
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
13
|
Lattar SM, Schneider RP, Eugenio VJ, Padilla G. High release of Candida albicans eDNA as protection for the scaffolding of polymicrobial biofilm formed with Staphylococcus aureus and Streptococcus mutans against the enzymatic activity of DNase I. Braz J Microbiol 2024; 55:3921-3932. [PMID: 39480631 PMCID: PMC11711860 DOI: 10.1007/s42770-024-01550-4] [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] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
This study aimed to determine the protective role of the high release of C. albicans extracellular DNA (eDNA) in a polymicrobial biofilm formed by S. aureus and S. mutans in the course of DNase I treatment. A tube-flow biofilm bioreactor was developed to mimic biofilm formation in the oral cavity. eDNA release was quantified by real-time PCR (qPCR) and confocal microscopy analysis were used to determine the concentration and distribution of eDNA and intracellular DNA (iDNA). The mean amount of eDNA released by each species in the polymicrobial was higher than that in monospecies biofilms (S. aureus: 3.1 × 10-2 ng/μl polymicrobial versus 5.1 × 10-4 ng/μl monospecies; S. mutans: 3 × 10-1 ng/μl polymicrobial versus 2.97 × 10-2 ng/μl monospecies; C. albicans: 8.35 ng/μl polymicrobial versus 4.85 ng/μl monospecies). The large amounts of eDNA released by C. albicans (96%) in polymicrobial biofilms protects the S. aureus and S. mutans cells against the degradation by DNase I and dampens the effect of clindamycin.
Collapse
Affiliation(s)
- Santiago M Lattar
- Cell Biology of Microorganism Laboratory, Department of Microbiology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, BH, Brazil.
| | | | - Vidal Jorge Eugenio
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gabriel Padilla
- Bioproducts Laboratory, Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
14
|
Chen N, Li Y, Liang X, Qin K, Zhang Y, Wang J, Wu Q, Gupta TB, Ding Y. Bacterial extracellular vesicle: A non-negligible component in biofilm life cycle and challenges in biofilm treatments. Biofilm 2024; 8:100216. [PMID: 39184814 PMCID: PMC11341940 DOI: 10.1016/j.bioflm.2024.100216] [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: 04/07/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/27/2024] Open
Abstract
Bacterial biofilms, especially those formed by pathogens, have been increasingly impacting human health. Bacterial extracellular vesicle (bEV), a kind of spherical membranous structure released by bacteria, has not only been reported to be a component of the biofilm matrix but also plays a non-negligible role in the biofilm life cycle. Nevertheless, a comprehensive overview of the bEVs functions in biofilms remains elusive. In this review, we summarize the biogenesis and distinctive features characterizing bEVs, and consolidate the current literature on their functions and proposed mechanisms in the biofilm life cycle. Furthermore, we emphasize the formidable challenges associated with vesicle interference in biofilm treatments. The primary objective of this review is to raise awareness regarding the functions of bEVs in the biofilm life cycle and lay the groundwork for the development of novel therapeutic strategies to control or even eliminate bacterial biofilms.
Collapse
Affiliation(s)
- Nuo Chen
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yangfu Li
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xinmin Liang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Keyuan Qin
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Ying Zhang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Juan Wang
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qingping Wu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Tanushree B. Gupta
- Food System Integrity Team, AgResearch Ltd., Hopkirk Research Institute, Massey University, Palmerston North, 4474, New Zealand
| | - Yu Ding
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
15
|
Yang J, Xu JF, Liang S. Antibiotic resistance in Pseudomonas aeruginosa: mechanisms and emerging treatment. Crit Rev Microbiol 2024:1-19. [PMID: 39556143 DOI: 10.1080/1040841x.2024.2429599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/22/2024] [Accepted: 11/10/2024] [Indexed: 11/19/2024]
Abstract
Pseudomonas aeruginosa, able to survive on the surfaces of medical devices, is a life-threatening pathogen that mainly leads to nosocomial infection especially in immunodeficient and cystic fibrosis (CF) patients. The antibiotic resistance in P. aeruginosa has become a world-concerning problem, which results in reduced and ineffective therapy efficacy. Besides intrinsic properties to decrease the intracellular content and activity of antibiotics, P. aeruginosa develops acquired resistance by gene mutation and acquisition, as well as adaptive resistance under specific situations. With in-depth research on drug resistance mechanisms and the development of biotechnology, innovative strategies have emerged and yielded benefits such as screening for new antibiotics based on artificial intelligence technology, utilizing drugs synergistically, optimizing administration, and developing biological therapy. This review summarizes the recent advances in the mechanisms of antibiotic resistance and emerging treatments for combating resistance, aiming to provide a reference for the development of therapy against drug-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Jian Yang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuo Liang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Sun J, Wang X, Gao Y, Li S, Hu Z, Huang Y, Fan B, Wang X, Liu M, Qiao C, Zhang W, Wang Y, Ji X. H 2S scavenger as a broad-spectrum strategy to deplete bacteria-derived H 2S for antibacterial sensitization. Nat Commun 2024; 15:9422. [PMID: 39482291 PMCID: PMC11527999 DOI: 10.1038/s41467-024-53764-7] [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: 06/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024] Open
Abstract
Bacteria-derived H2S plays multifunctional protective roles against antibiotics insult, and the H2S biogenesis pathway is emerging as a viable target for the antibacterial adjuvant design. However, the development of a pan-inhibitor against H2S-synthesizing enzymes is challenging and underdeveloped. Herein, we propose an alternative strategy to downregulate the H2S levels in H2S-producing bacteria, which depletes the bacteria-derived H2S chemically by H2S scavengers without acting on the synthesizing enzymes. After the screening of chemically diversified scaffolds and a structural optimization campaign, a potent and specific H2S scavenger is successfully identified, which displays efficient H2S depletion in several H2S-producing bacteria, potentiates both bactericidal agents and photodynamic therapy, enhances the bacterial clearance of macrophages and polymorphonuclear neutrophils, disrupts the formation of bacterial biofilm and increases the sensitivity of bacterial persister cells to antibiotics. Most importantly, such an H2S scavenger exhibits sensitizing effects with gentamicin in Pseudomonas aeruginosa -infected pneumonia and skin wound female mouse models. In aggregate, our results not only provide an effective strategy to deplete bacteria-derived H2S and establish the H2S biogenesis pathway as a viable target for persisters and drug-resistant bacteria, but also deliver a promising antibacterial adjuvant for potential clinical translation.
Collapse
Affiliation(s)
- Jiekai Sun
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Xu Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Ye Gao
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Shuangyu Li
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Ziwei Hu
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Yan Huang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Baoqiang Fan
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Xia Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Miao Liu
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Chunhua Qiao
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Wei Zhang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China.
| | - Yipeng Wang
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China.
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai, China.
| | - Xingyue Ji
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
17
|
Awad R, Marchand S, Couet W, Nasser M, Buyck JM, Tewes F. Assessment of inhaled cationic antibiotics in an in vitro model of Pseudomonas aeruginosa lung biofilm. Microb Pathog 2024; 197:107020. [PMID: 39419459 DOI: 10.1016/j.micpath.2024.107020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVES This study aimed to evaluate the efficacy of inhaled cationic antibiotics, including tobramycin (TOB) and colistin (CST), using an in vitro alginate bead model that simulates Pseudomonas aeruginosa lung biofilms. METHODS Bioluminescent P. aeruginosa were encapsulated within alginate beads and dispersed in either Mueller-Hinton broth (MHB) or artificial sputum medium (ASM). The impact of bead size and culture medium on TOB and CST efficacy was assessed by monitoring bioluminescence kinetics, followed by colony-forming unit (CFU/mL) measurements. Antibiotic efficacy was quantified using a Hill inhibitory model to analyze variations in CFU/mL in response to TOB and CST concentrations. RESULTS The TOB EC50 was found to be 8-fold higher when P. aeruginosa was encapsulated in larger beads (1200 μm) compared to smaller beads (60 μm). TOB efficacy further decreased twofold when larger beads were dispersed in ASM. In contrast, CST demonstrated superior efficacy, being four times more potent than TOB, with corresponding EC50 values of 20.5 ± 2.8 times MIC and 78.4 ± 10.2 times MIC, respectively. No change in MICs was observed for either antibiotic, even after exposing bacteria to 200 times the MIC. CONCLUSIONS This P. aeruginosa biofilm model highlights how alginate and mucus modulated the efficacy of TOB and CST, and suggested the superior efficacy of CST in eradicating pulmonary biofilms.
Collapse
Affiliation(s)
- Rana Awad
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France
| | - Sandrine Marchand
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France; CHU de Poitiers, Laboratoire de Toxicologie et de Pharmacocinétique, Poitiers, France
| | - William Couet
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France; CHU de Poitiers, Laboratoire de Toxicologie et de Pharmacocinétique, Poitiers, France
| | | | - Julien M Buyck
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France
| | - Frédéric Tewes
- Université de Poitiers, PHAR2, Inserm U1070, Poitiers, France.
| |
Collapse
|
18
|
Squyres GR, Newman DK. Real-time high-resolution microscopy reveals how single-cell lysis shapes biofilm matrix morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618105. [PMID: 39463994 PMCID: PMC11507769 DOI: 10.1101/2024.10.13.618105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
During development, multiscale patterning requires that cells organize their behavior in space and time. Bacteria in biofilms must similarly dynamically pattern their behavior with a simpler toolkit. Like in eukaryotes, morphogenesis of the extracellular matrix is essential for biofilm development, but how it is patterned has remained unclear. Here, we explain how the architecture of eDNA, a key matrix component, is controlled by single cell lysis events during Pseudomonas aeruginosa biofilm development. We extend single-cell imaging methods to capture complete biofilm development, characterizing the stages of biofilm development and visualizing eDNA matrix morphogenesis. Mapping the spatiotemporal distribution of single cell lysis events reveals that cell lysis is restricted to a specific biofilm zone. Simulations indicate that this patterning couples cell lysis to growth, more uniformly distributing eDNA throughout the biofilm. Finally, we find that patterning of cell lysis is organized by nutrient gradients that act as positioning cues.
Collapse
Affiliation(s)
- Georgia R. Squyres
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Dianne K. Newman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
19
|
Lander SM, Fisher G, Everett BA, Tran P, Prindle A. Secreted nucleases reclaim extracellular DNA during biofilm development. NPJ Biofilms Microbiomes 2024; 10:103. [PMID: 39375363 PMCID: PMC11458576 DOI: 10.1038/s41522-024-00575-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024] Open
Abstract
DNA is the genetic code found inside all living cells and its molecular stability can also be utilized outside the cell. While extracellular DNA (eDNA) has been identified as a structural polymer in bacterial biofilms, whether it persists stably throughout development remains unclear. Here, we report that eDNA is temporarily invested in the biofilm matrix before being reclaimed later in development. Specifically, by imaging eDNA dynamics within undomesticated Bacillus subtilis biofilms, we found eDNA is produced during biofilm establishment before being globally degraded in a spatiotemporally coordinated pulse. We identified YhcR, a secreted Ca2+-dependent nuclease, as responsible for eDNA degradation in pellicle biofilms. YhcR cooperates with two other nucleases, NucA and NucB, to reclaim eDNA for its phosphate content in colony biofilms. Our results identify extracellular nucleases that are crucial for eDNA reclamation during biofilm development and we therefore propose a new role for eDNA as a dynamic metabolic reservoir.
Collapse
Affiliation(s)
- Stephen M Lander
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Chicago, 60611, IL, USA
- Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, 60611, IL, USA
| | - Garth Fisher
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Blake A Everett
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Chicago, 60611, IL, USA
| | - Peter Tran
- Center for Synthetic Biology, Northwestern University, Evanston, 60208, IL, USA
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, 60208, IL, USA
| | - Arthur Prindle
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Chicago, 60611, IL, USA.
- Center for Synthetic Biology, Northwestern University, Evanston, 60208, IL, USA.
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, 60208, IL, USA.
- Department of Microbiology-Immunology, Feinberg School of Medicine, Chicago, 60611, IL, USA.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, 60642, USA.
| |
Collapse
|
20
|
Tavaddod S, Dawson A, Allen RJ. Bacterial aggregation triggered by low-level antibiotic-mediated lysis. NPJ Biofilms Microbiomes 2024; 10:90. [PMID: 39327479 PMCID: PMC11427687 DOI: 10.1038/s41522-024-00553-1] [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: 11/11/2023] [Accepted: 08/20/2024] [Indexed: 09/28/2024] Open
Abstract
Suspended bacterial aggregates play a central role in ocean biogeochemistry, industrial processes and probably many clinical infections - yet the factors that trigger aggregation remain poorly understood, as does the relationship between suspended aggregates and surface-attached biofilms. Here we show that very low doses of cell-wall targeting antibiotic, far below the minimal inhibitory concentration, can trigger aggregation of Escherichia coli cells. This occurs when a few cells lyse, releasing extracellular DNA - thus, cell-to-cell variability in antibiotic response leads to population-level aggregation. Although lysis-triggered aggregation echoes known trigger mechanisms for surface-attached biofilms, these aggregates may have different ecological implications since they do not show increased biofilm-forming potential or increased antibiotic resistance. Our work contributes to understanding the nature of bacterial aggregates and the factors that trigger their formation, and the possible consequences of widespread low-dose antibiotic exposure in the environment and in the body.
Collapse
Affiliation(s)
- Sharareh Tavaddod
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Angela Dawson
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Rosalind J Allen
- School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK.
- Theoretical Microbial Ecology, Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
21
|
Zhang W, Miller CA, Wilson MJ. Assessment of the In Vitro Phosphatidylinositol Glycan Class A (PIG-A) Gene Mutation Assay Using Human TK6 and Mouse Hepa1c1c7 Cell Lines. J Xenobiot 2024; 14:1293-1311. [PMID: 39311152 PMCID: PMC11417843 DOI: 10.3390/jox14030073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Gene mutations linked to diseases like cancer may be caused by exposure to environmental chemicals. The X-linked phosphatidylinositol glycan class A (PIG-A) gene, required for glycosylphosphatidylinositol (GPI) anchor biosynthesis, is a key target locus for in vitro genetic toxicity assays. Various organisms and cell lines may respond differently to genotoxic agents. Here, we compared the mutagenic potential of directly genotoxic ethyl methane sulfonate (EMS) to metabolically activated pro-mutagenic polycyclic aromatic hydrocarbons (PAHs). The two classes of mutagens were compared in an in vitro PIG-A gene mutation test using the metabolically active murine hepatoma Hepa1c1c7 cell line and the human TK6 cell line, which has limited metabolic capability. Determination of cell viability is required for quantifying mutagenicity. Two common cell viability tests, the MTT assay and propidium iodide (PI) staining measured by flow cytometry, were evaluated. The MTT assay overestimated cell viability in adherent cells at high benzo[a]pyrene (B[a]P) exposure concentrations, so PI-based cytotoxicity was used in calculations. The spontaneous mutation rates for TK6 and Hepa1c1c7 cells were 1.87 and 1.57 per million cells per cell cycle, respectively. TK6 cells exposed to 600 µM and 800 µM EMS showed significantly higher mutation frequencies (36 and 47 per million cells per cell cycle, respectively). Exposure to the pro-mutagen benzo[a]pyrene (B[a]P, 10 µM) did not increase mutation frequency in TK6 cells. In Hepa1c1c7 cells, mutation frequencies varied across exposure groups (50, 50, 29, and 81 per million cells per cell cycle when exposed to 10 µM B[a]P, 5-methylcholanthrene (5-MC), chrysene, or 16,000 µM EMS, respectively). We demonstrate that the choice of cytotoxicity assay and cell line can determine the outcome of the Pig-A mutagenesis assay when assessing a specific mutagen.
Collapse
Affiliation(s)
- Wenhao Zhang
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (C.A.M.); (M.J.W.)
| | - Charles A. Miller
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (C.A.M.); (M.J.W.)
| | - Mark J. Wilson
- Department of Environmental Health Sciences, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (C.A.M.); (M.J.W.)
- Chemical Insights Research Institute of Underwriters Laboratories Research Institutes, Marietta, GA 30067, USA
| |
Collapse
|
22
|
Berking BB, Rijpkema SJ, Zhang BHE, Sait A, Amatdjais-Groenen H, Wilson DA. Biofilm Disruption from within: Light-Activated Molecular Drill-Functionalized Polymersomes Bridge the Gap between Membrane Damage and Quorum Sensing-Mediated Cell Death. ACS Biomater Sci Eng 2024; 10:5881-5891. [PMID: 39176452 PMCID: PMC11388143 DOI: 10.1021/acsbiomaterials.4c01177] [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: 08/24/2024]
Abstract
Bacterial biofilms represent an escalating global health concern with the proliferation of drug resistance and hospital-acquired infections annually. Numerous strategies are under exploration to combat biofilms and preempt the development of antibacterial resistance. Among these, mechanical disruption of biofilms and enclosed bacteria presents a promising avenue, aiming to induce membrane permeabilization and consequent lethal damage. Herein, we introduce a hemithioindigo (HTI) motor activated by visible light, capable of disrupting sessile bacteria when integrated into a polymeric vesicle carrier. Under visible light, bacteria exhibited a notable outer membrane permeability, reduced membrane fluidity, and diminished viability following mechanical drilling. Moreover, various genetic responses pertaining to the cell envelope were examined via qRT-PCR, alongside the activation of a self-lysis mechanism associated with phage stress, which was coupled with increases in quorum sensing, demonstrating a potential self-lysis cascade from within. The multifaceted mechanisms of action, coupled with the energy efficiency of mechanical damage, underscore the potential of this system in addressing the challenges posed by pathogenic biofilms.
Collapse
Affiliation(s)
- Bela B Berking
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University, Nijmegen 6500 HC, The Netherlands
| | - Sjoerd J Rijpkema
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University, Nijmegen 6500 HC, The Netherlands
| | - Bai H E Zhang
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University, Nijmegen 6500 HC, The Netherlands
| | - Arbaaz Sait
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University, Nijmegen 6500 HC, The Netherlands
| | - Helene Amatdjais-Groenen
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University, Nijmegen 6500 HC, The Netherlands
| | - Daniela A Wilson
- Systems Chemistry Department, Institute for Molecules and Materials, Radboud University, Nijmegen 6500 HC, The Netherlands
| |
Collapse
|
23
|
Cui Y, Wang D, Zhang L, Qu X. Research progress on the regulatory mechanism of biofilm formation in probiotic lactic acid bacteria. Crit Rev Food Sci Nutr 2024:1-15. [PMID: 39244761 DOI: 10.1080/10408398.2024.2400593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Probiotic lactic acid bacteria (LAB) must undergo three key stages of testing, including food processing, storage, and gastrointestinal tract environment, their beneficial effects could exert. The biofilm formation of probiotic LAB is helpful for improving their stress resistances, survival rates, and colonization abilities under adverse environmental conditions, laying an important foundation for their probiotic effects. In this review, the formation process, the composition and function of basic components of probiotic LAB biofilm have been summarized. This review focuses on the regulatory mechanism of probiotic LAB biofilm formation. In addition, the characteristics and related mechanisms of probiotics in biofilm state have been analyzed to guide the application of probiotic LAB biofilms in the field of health and food. The biofilm formation of LAB is an extremely complex process involving multiple regulatory factors. Besides quorum sensing (QS), other regulatory factors are not yet fully understood. The probiotic LAB in biofilm state exhibit superior survival rate, adhesion performance, and immunomodulation ability, attribute to various metabolic processes, including stress response, exopolysaccharide (EPS) metabolism, amino acid and protein metabolisms, etc. The understanding about regulatory mechanism of biofilm formation of different probiotic species and strains will accelerate the development and application of probiotics products.
Collapse
Affiliation(s)
- Yanhua Cui
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| | - Dongqi Wang
- Department of Food Nutrition and Health, School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Xiaojun Qu
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin, China
| |
Collapse
|
24
|
Yang N, Song S, Yang X, Nawaz MAH, He D, Han W, Li Y, Yu C. Fabrication of photo-induced molecular superoxide radical generator for highly efficient therapy against bacterial wound infection. Colloids Surf B Biointerfaces 2024; 241:114018. [PMID: 38865868 DOI: 10.1016/j.colsurfb.2024.114018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/18/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
The pressing need for highly efficient antibacterial strategies arises from the prevalence of microbial biofilm infections and the emergence of rapidly evolving antibiotic-resistant strains of pathogenic bacteria. Photodynamic therapy represents a highly efficient and compelling antibacterial approach, offering promising prospects for effective control of the development of bacterial resistance. However, the effectiveness of many photosensitizers is limited due to the reduced generation of reactive oxygen species (ROS) in hypoxic microenvironment, which commonly occur in pathological conditions such as inflammatory and bacteria-infected wounds. Herein, we designed and prepared two phenothiazine-derived photosensitizers (NB-1 and NB-2), which can effectively generate superoxide anion radicals (O2●-) through the type I process. Both photosensitizers demonstrate significant efficacy in vitro for the eradication of broad-spectrum bacteria. Moreover, NB-2 possesses distinct advantages including strong membrane binding and strong generation of O2●-, rendering it an exceptionally efficient antibacterial agent against mature biofilms. In addition, laser activated NB-2 could be applied to treat MRSA-infected wound in vivo, which offers new opportunities for potential practical applications.
Collapse
Affiliation(s)
- Na Yang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Shuang Song
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Xiaofei Yang
- Harbin Center for Disease Control and Prevention, Harbin 150030, PR China
| | - Muhammad Azhar Hayat Nawaz
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Di He
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Wenzhao Han
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Ying Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Cong Yu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China.
| |
Collapse
|
25
|
Schlafer S, Johnsen KK, Kjærbølling I, Schramm A, Meyer RL, Jørgensen MR. The efficacy and safety of an enzyme-containing lozenge for dental biofilm control-a randomized controlled pilot trial. J Dent 2024; 147:105107. [PMID: 38830531 DOI: 10.1016/j.jdent.2024.105107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024] Open
Abstract
OBJECTIVES To evaluate the effect of daily use of a multiple-enzyme lozenge on de novo plaque formation, on gingivitis development, and on the oral microbiome composition. METHODS This trial with two parallel arms included 24 healthy adults allocated to the Active (n = 12) or Placebo (n = 12) group. Subjects consumed one lozenge three times daily for seven days, and no oral hygiene procedures were allowed. Differences in de novo plaque accumulation between a baseline period, and one and seven days of intervention were assessed by the Turesky-modification of the Quigley-and-Hein-Plaque-Index (TM-QHPI). The development of gingivitis after seven days of intervention was assessed by the Gingival Index (GI). Plaque and saliva samples were collected at baseline and after seven days of intervention, and evaluated by 16S rRNA gene sequencing. RESULTS All subjects completed the study, and no adverse events were reported. After one day, the average TM-QHPI was significantly lower in the Active than in the Placebo group, as compared to baseline (p = 0.012). After 7 days, average TM-QHPI values did not differ significantly between groups (p = 0.37). GI values did not increase during the intervention period, with no difference between groups (p = 0.62). Bacterial richness increased in both plaque and saliva samples over a seven-day oral hygiene-free period, with a statistically significant difference for the saliva samples (p = 0.0495) between groups. CONCLUSIONS A multiple-enzymes lozenge decreased the build-up of de novo plaque after one day and slowed down the process of species increment in saliva. The lozenge may be an adjunct to regular mechanical plaque removal. CLINICAL SIGNIFICANCE Dental plaque is the main cause of caries, gingivitis, and periodontitis. The search for therapeutic adjuncts to mechanical plaque removal that have no harmful effects on the oral microbiome is important. Treatment with multiple plaque-matrix degrading enzymes is a promising non-biocidal approach to plaque control.
Collapse
Affiliation(s)
- Sebastian Schlafer
- Department of Dentistry and Oral Health, Section for Oral Ecology, Cariology, Faculty of Health, Aarhus University, Aarhus, Denmark.
| | - Karina K Johnsen
- Department of Dentistry and Oral Health, Section for Oral Ecology, Cariology, Faculty of Health, Aarhus University, Aarhus, Denmark
| | | | - Andreas Schramm
- Department of Biology, Section for Microbiology, Aarhus University, Aarhus, Denmark
| | - Rikke L Meyer
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
26
|
Mohamed H, Marusich E, Divashuk M, Leonov S. A unique combination of natural fatty acids from Hermetia illucens fly larvae fat effectively combats virulence factors and biofilms of MDR hypervirulent mucoviscus Klebsiella pneumoniae strains by increasing Lewis acid-base/van der Waals interactions in bacterial wall membranes. Front Cell Infect Microbiol 2024; 14:1408179. [PMID: 39119288 PMCID: PMC11306206 DOI: 10.3389/fcimb.2024.1408179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Hypervirulent Klebsiella pneumoniae (hvKp) and carbapenem-resistant K. pneumoniae (CR-Kp) are rapidly emerging as opportunistic pathogens that have a global impact leading to a significant increase in mortality rates among clinical patients. Anti-virulence strategies that target bacterial behavior, such as adhesion and biofilm formation, have been proposed as alternatives to biocidal antibiotic treatments to reduce the rapid emergence of bacterial resistance. The main objective of this study was to examine the efficacy of fatty acid-enriched extract (AWME3) derived from the fat of Black Soldier Fly larvae (Hermetia illucens) in fighting against biofilms of multi-drug resistant (MDR) and highly virulent Klebsiella pneumoniae (hvKp) pathogens. Additionally, the study also aimed to investigate the potential mechanisms underlying this effect. Methods Crystal violet (CV) and ethidium bromide (EtBr) assays show how AWME3 affects the formation of mixed and mature biofilms by the KP ATCC BAA-2473, KPi1627, and KPM9 strains. AWME3 has shown exceptional efficacy in combating the hypermucoviscosity (HMV) virulent factors of KPi1627 and KPM9 strains when tested using the string assay. The rudimentary motility of MDR KPM9 and KP ATCC BAA-2473 strains was detected through swimming, swarming, and twitching assays. The cell wall membrane disturbances induced by AWME3 were detected by light and scanning electron microscopy and further validated by an increase in the bacterial cell wall permeability and Lewis acid-base/van der Waals characteristics of K. pneumoniae strains tested by MATS (microbial adhesion to solvents) method. Results After being exposed to 0.5 MIC (0.125 mg/ml) of AWME3, a significant reduction in the rudimentary motility of MDR KPM9 and KP ATCC BAA-2473 strains, whereas the treated bacterial strains exhibited motility between 4.23 ± 0.25 and 4.47 ± 0.25 mm, while the non-treated control groups showed significantly higher motility ranging from 8.5 ± 0.5 to 10.5 ± 0.5 mm. Conclusion In conclusion, this study demonstrates the exceptional capability of the natural AWME3 extract enriched with a unique combination of fatty acids to effectively eliminate the biofilms formed by the highly drug-resistant and highly virulent K. pneumoniae (hvKp) pathogens. Our results highlight the opportunity to control and minimize the rapid emergence of bacterial resistance through the treatment using AWME3 of biofilm-associated infections caused by hvKp and CRKp pathogens.
Collapse
Affiliation(s)
- Heakal Mohamed
- Agricultural Research Center (ARC), Plant Protection Research Institute (PPRI), Giza, Egypt
- The Laboratory of Personalized Chemoradiotherapy, Institute of Future Biophysics, Moscow, Russia
| | - Elena Marusich
- The Laboratory of Personalized Chemoradiotherapy, Institute of Future Biophysics, Moscow, Russia
| | - Mikhail Divashuk
- All-Russia Research Institute of Agricultural Biotechnology Kurchatov Genomic Center - VNIISB, Moscow, Russia
| | - Sergey Leonov
- The Laboratory of Personalized Chemoradiotherapy, Institute of Future Biophysics, Moscow, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
27
|
David A, Tahrioui A, Tareau AS, Forge A, Gonzalez M, Bouffartigues E, Lesouhaitier O, Chevalier S. Pseudomonas aeruginosa Biofilm Lifecycle: Involvement of Mechanical Constraints and Timeline of Matrix Production. Antibiotics (Basel) 2024; 13:688. [PMID: 39199987 PMCID: PMC11350761 DOI: 10.3390/antibiotics13080688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic infections, especially in immunocompromised patients. Its remarkable adaptability and resistance to various antimicrobial treatments make it difficult to eradicate. Its persistence is enabled by its ability to form a biofilm. Biofilm is a community of sessile micro-organisms in a self-produced extracellular matrix, which forms a scaffold facilitating cohesion, cell attachment, and micro- and macro-colony formation. This lifestyle provides protection against environmental stresses, the immune system, and antimicrobial treatments, and confers the capacity for colonization and long-term persistence, often characterizing chronic infections. In this review, we retrace the events of the life cycle of P. aeruginosa biofilm, from surface perception/contact to cell spreading. We focus on the importance of extracellular appendages, mechanical constraints, and the kinetics of matrix component production in each step of the biofilm life cycle.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sylvie Chevalier
- Univ Rouen Normandie, Univ Caen Normandie, Normandie Univ, CBSA UR 4312, F-76000 Rouen, France
| |
Collapse
|
28
|
Yang S, Meng X, Zhen Y, Baima Q, Wang Y, Jiang X, Xu Z. Strategies and mechanisms targeting Enterococcus faecalis biofilms associated with endodontic infections: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1433313. [PMID: 39091674 PMCID: PMC11291369 DOI: 10.3389/fcimb.2024.1433313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Enterococcus faecalis is one of the main microorganisms that infects root canals, ranking among the most prevalent microorganisms associated with endodontic treatment failure. Given its pervasive presence in persistent endodontic infections, the successful elimination of Enterococcus faecalis is crucial for effective endodontic treatment and retreatment. Furthermore, Enterococcus faecalis can form biofilms - defense structures that microbes use to fight environmental threats. These biofilms confer resistance against host immune system attacks and antibiotic interventions. Consequently, the presence of biofilms poses a significant challenge in the complete eradication of Enterococcus faecalis and its associated disease. In response, numerous scholars have discovered promising outcomes in addressing Enterococcus faecalis biofilms within root canals and undertaken endeavors to explore more efficacious approaches in combating these biofilms. This study provides a comprehensive review of strategies and mechanisms for the removal of Enterococcus faecalis biofilms.
Collapse
Affiliation(s)
- Shipeng Yang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiuping Meng
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuqi Zhen
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Quzhen Baima
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yu Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xinmiao Jiang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhibo Xu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
29
|
Tetz V, Kardava K, Vecherkovskaya M, Khodadadi-Jamayran A, Tsirigos A, Tetz G. Previously unknown regulatory role of extracellular RNA on bacterial directional migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.603110. [PMID: 39026763 PMCID: PMC11257571 DOI: 10.1101/2024.07.11.603110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Bacterial directional migration plays a significant role in bacterial adaptation. However, the regulation of this process, particularly in young biofilms, remains unclear. Here, we demonstrated the critical role of extracellular RNA as part of the Universal Receptive System in bacterial directional migration using a multidisciplinary approach, including bacterial culture, biochemistry, and genetics. We found that the destruction or inactivation of extracellular RNA with RNase or RNA-specific antibodies in the presence of the chemoattractant triggered the formation of bacterial "runner cells» in what we call a "panic state" capable of directional migration. These cells quickly migrated even on the surface of 1.5% agar and formed evolved colonies that were transcriptionally and biochemically different from the ancestral cells. We have also shown that cell-free DNA from blood plasma can act as a potent bacterial chemoattractant. Our data revealed a previously unknown role of bacterial extracellular RNA in the regulation of bacterial migration and have shown that its destruction or inhibition triggered the directional migration of developing and mature biofilms towards the chemoattractant.
Collapse
|
30
|
Mahmud HA, Garcia R, Garcia A, Baishya J, Wakeman CA. Rescue of pyrimidine-defective Pseudomonas aeruginosa through metabolic complementation. Microbiol Spectr 2024; 12:e0422623. [PMID: 38990029 PMCID: PMC11302043 DOI: 10.1128/spectrum.04226-23] [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: 12/18/2023] [Accepted: 06/28/2024] [Indexed: 07/12/2024] Open
Abstract
Chronic infections harbor multiple pathogens where dynamic interactions between members of the polymicrobial community play a major role in determining the infection outcome. For example, in a nutrient-rich polymicrobial infection, bacteria have the potential to undergo evolutionary changes that impair their ability to synthesize essential metabolites. This adaptation may facilitate metabolic interdependencies between neighboring pathogens and lead to difficult-to-treat chronic infections. Our research group previously demonstrated that Pseudomonas aeruginosa (PA) and Staphylococcus aureus (SA), typically considered classical competitors, can adopt a cooperative lifestyle through bi-directional purine exchange medicated by exogenous DNA (eDNA) release. To further validate our initial findings, in this study, we investigated the potential exchange of pyrimidine between PA and other pathogens, which is another constituent of DNA. In our findings, we observed that a pyrimidine-deficient transposon mutant strain of PA showed improved growth when co-cultured with wild-type PA, SA, Acinetobacter baumannii (AB), and Enterococcus faecalis (EF). Additionally, improved fitness of pyrimidine-deficient PA was further observed in chemical complementation with eDNA and uridine-5'-monophosphate. Interestingly, the rescue of PA growth through eDNA complementation is not as effective as in intact cells, such as SA, AB, EF, and wild-type PA, implying that eDNA is a lesser contributor to this metabolic complementation. Also, the exchange mechanism between pathogens involves more active mechanisms beyond simple eDNA or metabolite release. Our data further highlights the importance of cell-to-cell contact for effective and increased metabolic complementation. IMPORTANCE This research holds crucial implications for combating chronic infections, where multiple pathogens coexist and interact within the same environment. By uncovering the dynamic exchange of pyrimidines between Pseudomonas aeruginosa (PA) and Staphylococcus aureus (SA), our study reveals a previously unrecognized aspect of interspecies cooperation. The observed enhanced growth of a pyrimidine-deficient PA strain when co-cultured with SA suggests potential avenues for understanding and disrupting bacterial metabolic interdependencies in chronic infection settings. Furthermore, our findings highlight the mechanisms involved in metabolic exchange, emphasizing the importance of cell-to-cell contact. This research explored essential metabolic interactions to address the challenges posed by difficult-to-treat chronic infections.
Collapse
Affiliation(s)
- Hafij Al Mahmud
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Randy Garcia
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Alexsis Garcia
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Jiwasmika Baishya
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA
- The Assam Royal Global University, Guwahati, Assam, India
| | | |
Collapse
|
31
|
Lin H, Li R, Chen Y, Cheng Y, Yuan Q, Luo Y. Enhanced sensitivity of extracellular antibiotic resistance genes (ARGs) to environmental concentrations of antibiotic. CHEMOSPHERE 2024; 360:142434. [PMID: 38797215 DOI: 10.1016/j.chemosphere.2024.142434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
As emerging contaminants, antibiotics are frequently present in various environments, particularly rivers, albeit often at sublethal concentrations (ng/L∼μg/L). Assessing the risk associated with these low levels, which are far below the lethal threshold for most organisms, remains challenging. In this study, using microcosms containing planktonic bacteria and biofilm, we examined how antibiotic resistance genes (ARGs) in different physical states, including intracellular ARGs (iARGs) and extracellular ARGs (eARGs) responded to these low-level antibiotics. Our findings reveal a positive correlation between sub-lethal antibiotic exposure (ranging from 0.1 to 10 μg/L) and increased prevalence (measured as ARG copies/16s rDNA) of both iARGs and eARGs in planktonic bacteria. Notably, eARGs demonstrated greater sensitivity to antibiotic exposure compared to iARGs, with a lower threshold (0.1 μg/L for eARGs versus 1 μg/L for iARGs) for abundance increase. Moreover, ARGs in biofilms demonstrates higher sensitivity to antibiotic exposure compared to planktonic bacteria. To elucidate the underlying mechanisms, we established an integrated population dynamics-pharmacokinetics-pharmacodynamics (PD-PP) model. This model indicates that the enhanced sensitivity of eARGs is primarily driven by an increased potential for plasmid release from cells under low antibiotic concentrations. Furthermore, the accumulation of antibiotic in biofilms induces a greater sensitivity of ARG compared to the planktonic bacteria. This study provides a fresh perspective on the development of antibiotic resistance and offers an innovative approach for assessing the risk of sublethal antibiotic in the environment.
Collapse
Affiliation(s)
- Huai Lin
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China; Shenzhen Research Institute of Nanjing University, Shen Zhen, 518000, China
| | - Ruiqing Li
- School of Environmental Science and Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Yuying Chen
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China
| | - Yuan Cheng
- School of Environmental Science and Engineering, Nanjing Tech University, Nanjing, 211816, China
| | - Qingbin Yuan
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China; School of Environmental Science and Engineering, Nanjing Tech University, Nanjing, 211816, China.
| | - Yi Luo
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
32
|
Wang S, Tian R, Bi Y, Meng F, Zhang R, Wang C, Wang D, Liu L, Zhang B. A review of distribution and functions of extracellular DNA in the environment and wastewater treatment systems. CHEMOSPHERE 2024; 359:142264. [PMID: 38714248 DOI: 10.1016/j.chemosphere.2024.142264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/23/2024] [Accepted: 05/04/2024] [Indexed: 05/09/2024]
Abstract
Extracellular DNA refers to DNA fragments existing outside the cell, originating from various cell release mechanisms, including active secretion, cell lysis, and phage-mediated processes. Extracellular DNA serves as a vital environmental biomarker, playing crucial ecological and environmental roles in water bodies. This review is summarized the mechanisms of extracellular DNA release, including pathways involving cell lysis, extracellular vesicles, and type IV secretion systems. Then, the extraction and detection methods of extracellular DNA from water, soil, and biofilm are described and analyzed. Finally, we emphasize the role of extracellular DNA in microbial community systems, including its significant contributions to biofilm formation, biodiversity through horizontal gene transfer, and electron transfer processes. This review offers a comprehensive insight into the sources, distribution, functions, and impacts of extracellular DNA within aquatic environments, aiming to foster further exploration and understanding of extracellular DNA dynamics in aquatic environments as well as other environments.
Collapse
Affiliation(s)
- Shaopo Wang
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China
| | - Ruimin Tian
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China
| | - Yanmeng Bi
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China
| | - Fansheng Meng
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China
| | - Rui Zhang
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China
| | - Chenchen Wang
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China
| | - Dong Wang
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China
| | - Lingjie Liu
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Jinjing Road 26, Tianjin, 300384, China; Tianjin Key Laboratory of Aquatic Science and Technology, Jinjing Road 26, Tianjin, China.
| | - Bo Zhang
- Tianjin Eco-City Water Investment and Construction Co. Ltd, Hexu Road 276, Tianjin, 300467, China
| |
Collapse
|
33
|
Li X, Lin S, Wang Y, Chen Y, Zhang W, Shu G, Li H, Xu F, Lin J, Peng G, Fu H. Application of biofilm dispersion-based nanoparticles in cutting off reinfection. Appl Microbiol Biotechnol 2024; 108:386. [PMID: 38896257 PMCID: PMC11186951 DOI: 10.1007/s00253-024-13120-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 06/21/2024]
Abstract
Bacterial biofilms commonly cause chronic and persistent infections in humans. Bacterial biofilms consist of an inner layer of bacteria and an autocrine extracellular polymeric substance (EPS). Biofilm dispersants (abbreviated as dispersants) have proven effective in removing the bacterial physical protection barrier EPS. Dispersants are generally weak or have no bactericidal effect. Bacteria dispersed from within biofilms (abbreviated as dispersed bacteria) may be more invasive, adhesive, and motile than planktonic bacteria, characteristics that increase the probability that dispersed bacteria will recolonize and cause reinfection. The dispersants should be combined with antimicrobials to avoid the risk of severe reinfection. Dispersant-based nanoparticles have the advantage of specific release and intense penetration, providing the prerequisite for further antibacterial agent efficacy and achieving the eradication of biofilms. Dispersant-based nanoparticles delivered antimicrobial agents for the treatment of diseases associated with bacterial biofilm infections are expected to be an effective measure to prevent reinfection caused by dispersed bacteria. KEY POINTS: • Dispersed bacteria harm and the dispersant's dispersion mechanisms are discussed. • The advantages of dispersant-based nanoparticles in bacteria biofilms are discussed. • Dispersant-based nanoparticles for cutting off reinfection in vivo are highlighted.
Collapse
Affiliation(s)
- Xiaojuan Li
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Shiyu Lin
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yueli Wang
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yang Chen
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Wei Zhang
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Gang Shu
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Haohuan Li
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Funeng Xu
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Juchun Lin
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Guangneng Peng
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hualin Fu
- Innovative Engineering Research Center of Veterinary Pharmaceutics, Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
34
|
Palomba E, Chiusano ML, Monticolo F, Langella MC, Sanchez M, Tirelli V, de Alteriis E, Iannaccone M, Termolino P, Capparelli R, Carteni F, Incerti G, Mazzoleni S. Extracellular Self-DNA Effects on Yeast Cell Cycle and Transcriptome during Batch Growth. Biomolecules 2024; 14:663. [PMID: 38927066 PMCID: PMC11201494 DOI: 10.3390/biom14060663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
The cell cycle and the transcriptome dynamics of yeast exposed to extracellular self-DNA during an aerobic batch culture on glucose have been investigated using cytofluorimetric and RNA-seq analyses. In parallel, the same study was conducted on yeast cells growing in the presence of (heterologous) nonself-DNA. The self-DNA treatment determined a reduction in the growth rate and a major elongation of the diauxic lag phase, as well as a significant delay in the achievement of the stationary phase. This was associated with significant changes in the cell cycle dynamics, with slower exit from the G0 phase, followed by an increased level of cell percentage in the S phase, during the cultivation. Comparatively, the exposure to heterologous DNA did not affect the growth curve and the cell cycle dynamics. The transcriptomic analysis showed that self-DNA exposure produced a generalized downregulation of transmembrane transport and an upregulation of genes associated with sulfur compounds and the pentose phosphate pathway. Instead, in the case of the nonself treatment, a clear response to nutrient deprivation was detected. Overall, the presented findings represent further insights into the complex functional mechanisms of self-DNA inhibition.
Collapse
Affiliation(s)
- Emanuela Palomba
- Institute of Biosciences and Bioresources CNR, Via Università 133, 80055 Portici, Italy; (E.P.); (P.T.)
| | - Maria Luisa Chiusano
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università 100, 80055 Portici, Italy; (M.L.C.); (F.M.); (M.C.L.); (R.C.); (F.C.)
| | - Francesco Monticolo
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università 100, 80055 Portici, Italy; (M.L.C.); (F.M.); (M.C.L.); (R.C.); (F.C.)
- Cutaneous Biology Research Center, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Maria Chiara Langella
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università 100, 80055 Portici, Italy; (M.L.C.); (F.M.); (M.C.L.); (R.C.); (F.C.)
| | - Massimo Sanchez
- Istituto Superiore di Sanità (ISS) Core Facilities, Viale Regina Elena 299, 00161 Rome, Italy; (M.S.); (V.T.)
| | - Valentina Tirelli
- Istituto Superiore di Sanità (ISS) Core Facilities, Viale Regina Elena 299, 00161 Rome, Italy; (M.S.); (V.T.)
| | - Elisabetta de Alteriis
- Department of Biology, University of Naples “Federico II”, Via Cinthia 26, 80126 Naples, Italy;
| | - Marco Iannaccone
- Laboratory of Bioproducts and Bioprocesses ENEA, Piazzale Enrico Fermi 1, 80055 Portici, Italy;
| | - Pasquale Termolino
- Institute of Biosciences and Bioresources CNR, Via Università 133, 80055 Portici, Italy; (E.P.); (P.T.)
| | - Rosanna Capparelli
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università 100, 80055 Portici, Italy; (M.L.C.); (F.M.); (M.C.L.); (R.C.); (F.C.)
| | - Fabrizio Carteni
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università 100, 80055 Portici, Italy; (M.L.C.); (F.M.); (M.C.L.); (R.C.); (F.C.)
| | - Guido Incerti
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Via delle Scienze 206, 33100 Udine, Italy;
| | - Stefano Mazzoleni
- Department of Agricultural Sciences, University of Naples “Federico II”, Via Università 100, 80055 Portici, Italy; (M.L.C.); (F.M.); (M.C.L.); (R.C.); (F.C.)
| |
Collapse
|
35
|
Tian S, Shi L, Ren Y, van der Mei HC, Busscher HJ. A normalized parameter for comparison of biofilm dispersants in vitro. Biofilm 2024; 7:100188. [PMID: 38495770 PMCID: PMC10943042 DOI: 10.1016/j.bioflm.2024.100188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Dispersal of infectious biofilms increases bacterial concentrations in blood. To prevent sepsis, the strength of a dispersant should be limited to allow the immune system to remove dispersed bacteria from blood, preferably without antibiotic administration. Biofilm bacteria are held together by extracellular polymeric substances that can be degraded by dispersants. Currently, comparison of the strength of dispersants is not possible by lack of a suitable comparison parameter. Here, a biofilm dispersal parameter is proposed that accounts for differences in initial biofilm properties, dispersant concentration and exposure time by using PBS as a control and normalizing outcomes with respect to concentration and time. The parameter yielded near-identical values based on dispersant-induced reductions in biomass or biofilm colony-forming-units and appeared strain-dependent across pathogens. The parameter as proposed is largely independent of experimental methods and conditions and suitable for comparing different dispersants with respect to different causative strains in particular types of infection.
Collapse
Affiliation(s)
- Shuang Tian
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713, AV, Groningen, the Netherlands
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, PR China
| | - Yijin Ren
- University of Groningen and University Medical Center Groningen, Department of Orthodontics, Hanzeplein 1, 9700, RB, Groningen, the Netherlands
| | - Henny C. van der Mei
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713, AV, Groningen, the Netherlands
| | - Henk J. Busscher
- University of Groningen and University Medical Center Groningen, Department of Biomedical Engineering, Antonius Deusinglaan 1, 9713, AV, Groningen, the Netherlands
| |
Collapse
|
36
|
Maltz-Matyschsyk M, Melchiorre CK, Knecht DA, Lynes MA. Bacterial metallothionein, PmtA, a novel stress protein found on the bacterial surface of Pseudomonas aeruginosa and involved in management of oxidative stress and phagocytosis. mSphere 2024; 9:e0021024. [PMID: 38712943 PMCID: PMC11237414 DOI: 10.1128/msphere.00210-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Metallothioneins (MTs) are small cysteine-rich proteins that play important roles in homeostasis and protection against heavy metal toxicity and oxidative stress. The opportunistic pathogen, Pseudomonas aeruginosa, expresses a bacterial MT known as PmtA. Utilizing genetically modified P. aeruginosa PAO1 strains (a human clinical wound isolate), we show that inducing pmtA increases levels of pyocyanin and biofilm compared to other PAO1 isogenic strains, supporting previous results that pmtA is important for pyocyanin and biofilm production. We also show that overexpression of pmtA in vitro provides protection for cells exposed to oxidants, which is a characteristic of inflammation, indicating a role for PmtA as an antioxidant in inflammation. We found that a pmtA clean deletion mutant is phagocytized faster than other PAO1 isogenic strains in THP-1 human macrophage cells, indicating that PmtA provides protection from the phagocytic attack. Interestingly, we observed that monoclonal anti-PmtA antibody binds to PmtA, which is accessible on the surface of PAO1 strains using both flow cytometry and enzyme-linked immunosorbent assay techniques. Finally, we investigated intracellular persistence of these PAO1 strains within THP-1 macrophages cells and found that the phagocytic endurance of PAO1 strains is affected by pmtA expression. These data show for the first time that a bacterial MT (pmtA) can play a role in the phagocytic process and can be found on the outer surface of PAO1. Our results suggest that PmtA plays a role both in protection from oxidative stress and in the resistance to the host's innate immune response, identifying PmtA as a potential therapeutic target in P. aeruginosa infection. IMPORTANCE The pathogen Pseudomonas aeruginosa is a highly problematic multidrug-resistant (MDR) pathogen with complex virulence networks. MDR P. aeruginosa infections have been associated with increased clinical visits, very poor healthcare outcomes, and these infections are ranked as critical on priority lists of both the Centers for Disease Control and Prevention and the World Health Organization. Known P. aeruginosa virulence factors have been extensively studied and are implicated in counteracting host defenses, causing direct damage to the host tissues, and increased microbial competitiveness. Targeting virulence factors has emerged as a new line of defense in the battle against MDR P. aeruginosa strains. Bacterial metallothionein is a newly recognized virulence factor that enables evasion of the host immune response. The studies described here identify mechanisms in which bacterial metallothionein (PmtA) plays a part in P. aeruginosa pathogenicity and identifies PmtA as a potential therapeutic target.
Collapse
Affiliation(s)
| | - Clare K Melchiorre
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - David A Knecht
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
37
|
Oves M, Khan MS, Al-Shaeri M, Khan MS. Antifungal potential of multi-drug-resistant Pseudomonas aeruginosa: harnessing pyocyanin for candida growth inhibition. Front Cell Infect Microbiol 2024; 14:1375872. [PMID: 38846355 PMCID: PMC11155300 DOI: 10.3389/fcimb.2024.1375872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/11/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Pseudomonas aeruginosa is notorious for its multidrug resistance and its involvement in hospital-acquired infections. In this study, 20 bacterial strains isolated from soil samples near the Hindan River in Ghaziabad, India, were investigated for their biochemical and morphological characteristics, with a focus on identifying strains with exceptional drug resistance and pyocyanin production. Methods The isolated bacterial strains were subjected to biochemical and morphological analyses to characterize their properties, with a particular emphasis on exopolysaccharide production. Strain GZB16/CEES1, exhibiting remarkable drug resistance and pyocyanin production. Biochemical and molecular analyses, including sequencing of its 16S rRNA gene (accession number LN735036.1), plasmid-curing assays, and estimation of plasmid size, were conducted to elucidate its drug resistance mechanisms and further pyocynin based target the Candida albicans Strain GZB16/CEES1 demonstrated 100% resistance to various antibiotics used in the investigation, with plasmid-curing assays, suggesting plasmid-based resistance gene transmission. The plasmid in GZB16/CEES1 was estimated to be approximately 24 kb in size. The study focused on P. aeruginosa's pyocyanin production, revealing its association with anticandidal activity. The minimum inhibitory concentration (MIC) of the bacterial extract against Candida albicans was 50 μg/ml, with a slightly lower pyocyanin-based MIC of 38.5 μg/ml. Scanning electron microscopy illustrated direct interactions between P. aeruginosa strains and Candida albicans cells, leading to the destruction of the latter. Discussion These findings underscore the potential of P. aeruginosa in understanding microbial interactions and developing strategies to combat fungal infections. The study highlights the importance of investigating bacterial-fungal interactions and the role of pyocyanin in antimicrobial activity. Further research in this area could lead to the development of novel therapeutic approaches for combating multidrug-resistant infections.
Collapse
Affiliation(s)
- Mohammad Oves
- Center of Excellence in Environmental Studies, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Majed Al-Shaeri
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Saghir Khan
- Department of Agricultural Microbiology, Faculty of Agricultural Science, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
38
|
Lopes AA, Vendrell-Fernández S, Deschamps J, Georgeault S, Cokelaer T, Briandet R, Ghigo JM. Bile-induced biofilm formation in Bacteroides thetaiotaomicron requires magnesium efflux by an RND pump. mBio 2024; 15:e0348823. [PMID: 38534200 PMCID: PMC11078008 DOI: 10.1128/mbio.03488-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Bacteroides thetaiotaomicron is a prominent member of the human gut microbiota contributing to nutrient exchange, gut function, and maturation of the host's immune system. This obligate anaerobe symbiont can adopt a biofilm lifestyle, and it was recently shown that B. thetaiotaomicron biofilm formation is promoted by the presence of bile. This process also requires a B. thetaiotaomicron extracellular DNase, which is not, however, regulated by bile. Here, we showed that bile induces the expression of several Resistance-Nodulation-Division (RND) efflux pumps and that inhibiting their activity with a global competitive efflux inhibitor impaired bile-dependent biofilm formation. We then showed that, among the bile-induced RND-efflux pumps, only the tripartite BT3337-BT3338-BT3339 pump, re-named BipABC [for Bile Induced Pump A (BT3337), B (BT3338), and C (BT3339)], is required for biofilm formation. We demonstrated that BipABC is involved in the efflux of magnesium to the biofilm extracellular matrix, which leads to a decrease of extracellular DNA concentration. The release of magnesium in the biofilm matrix also impacts biofilm structure, potentially by modifying the electrostatic repulsion forces within the matrix, reducing interbacterial distance and allowing bacteria to interact more closely and form denser biofilms. Our study therefore, identified a new molecular determinant of B. thetaiotaomicron biofilm formation in response to bile salts and provides a better understanding on how an intestinal chemical cue regulates biofilm formation in a major gut symbiont.IMPORTANCEBacteroides thetaiotaomicron is a prominent member of the human gut microbiota able to degrade dietary and host polysaccharides, altogether contributing to nutrient exchange, gut function, and maturation of the host's immune system. This obligate anaerobe symbiont can adopt a biofilm community lifestyle, providing protection against environmental factors that might, in turn, protect the host from dysbiosis and dysbiosis-related diseases. It was recently shown that B. thetaiotaomicron exposure to intestinal bile promotes biofilm formation. Here, we reveal that a specific B. thetaiotaomicron membrane efflux pump is induced in response to bile, leading to the release of magnesium ions, potentially reducing electrostatic repulsion forces between components of the biofilm matrix. This leads to a reduction of interbacterial distance and strengthens the biofilm structure. Our study, therefore, provides a better understanding of how bile promotes biofilm formation in a major gut symbiont, potentially promoting microbiota resilience to stress and dysbiosis events.
Collapse
Affiliation(s)
- Anne-Aurélie Lopes
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Department of Microbiology, Paris, France
- Pediatric Emergency, AP-HP, Necker-Enfants-Malades University Hospital, Paris, France
| | - Sol Vendrell-Fernández
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Department of Microbiology, Paris, France
| | - Julien Deschamps
- INRAE, AgroParisTech, Université Paris-Saclay Institut Micalis, Paris, France
| | - Sonia Georgeault
- Plateforme IBiSA des Microscopies, Université et CHRU de Tours, Tours, France
| | - Thomas Cokelaer
- Institut Pasteur, Université Paris Cité, Plate-forme Technologique Biomics, Center for Technological Resources and Research, Paris, France
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Center for Technological Resources and Research, Paris, France
| | - Romain Briandet
- INRAE, AgroParisTech, Université Paris-Saclay Institut Micalis, Paris, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Université Paris-Cité, UMR CNRS 6047, Genetics of Biofilms Laboratory, Department of Microbiology, Paris, France
| |
Collapse
|
39
|
Zhong S, Yang J, Huang H. The role of single and mixed biofilms in Clostridioides difficile infection and strategies for prevention and inhibition. Crit Rev Microbiol 2024; 50:285-299. [PMID: 36939635 DOI: 10.1080/1040841x.2023.2189950] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/06/2023] [Indexed: 03/21/2023]
Abstract
Clostridioides difficile infection (CDI) is a serious disease with a high recurrence rate. The single and mixed biofilms formed by C. difficile in the gut contribute to the formation of recurrent CDI (rCDI). In parallel, other gut microbes influence the formation and development of C. difficile biofilms, also known as symbiotic biofilms. Interactions between members within the symbiotic biofilm are associated with the worsening or alleviation of CDI. These interactions include effects on C. difficile adhesion and chemotaxis, modulation of LuxS/AI-2 quorum sensing (QS) system activity, promotion of cross-feeding by microbial metabolites, and regulation of intestinal bile acid and pyruvate levels. In the process of C. difficile biofilms control, inhibition of C. difficile initial biofilm formation and killing of C. difficile vegetative cells and spores are the main targets of action. The role of symbiotic biofilms in CDI suggested that targeting interventions of C. difficile-promoting gut microbes could indirectly inhibit the formation of C. difficile mixed biofilms and improved the ultimate therapeutic effect. In summary, this review outlines the mechanisms of C. difficile biofilm formation and summarises the treatment strategies for such single and mixed biofilms, aiming to provide new ideas for the prevention and treatment of CDI.
Collapse
Affiliation(s)
- Saiwei Zhong
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| |
Collapse
|
40
|
Müller ND, Kirtane A, Schefer RB, Mitrano DM. eDNA Adsorption onto Microplastics: Impacts of Water Chemistry and Polymer Physiochemical Properties. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:7588-7599. [PMID: 38624040 DOI: 10.1021/acs.est.3c10825] [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: 04/17/2024]
Abstract
Adsorption of biomacromolecules onto polymer surfaces, including microplastics (MPs), occurs in multiple environmental compartments, forming an ecocorona. Environmental DNA (eDNA), genetic material shed from organisms, can adsorb onto MPs which can potentially either (1) promote long-range transport of antibiotic resistant genes or (2) serve to gain insights into the transport pathways and origins of MPs by analyzing DNA sequences on MPs. However, little is known about the capacity of MPs to adsorb eDNA or the factors that influence sorption, such as polymer and water chemistries. Here we investigated the adsorption of extracellular linear DNA onto a variety of model MP fragments composed of three of the most environmentally prevalent polymers (polyethylene, polyethylene terephthalate, and polystyrene) in their pristine and photochemically weathered states. Batch adsorption experiments in a variety of water chemistries were complemented with nonlinear modeling to quantify the rate and extent of eDNA sorption. Ionic strength was shown to strongly impact DNA adsorption by reducing or inhibiting electrostatic repulsion. Polyethylene terephthalate exhibited the highest adsorption capacity when normalizing for MP specific surface area, likely due to the presence of ester groups. Kinetics experiments showed fast adsorption (majority adsorbed under 30 min) before eventually reaching equilibrium after 1-2 h. Overall, we demonstrated that DNA quickly binds to MPs, with pseudo-first- and -second-order models describing adsorption kinetics and the Freundlich model describing adsorption isotherms most accurately. These insights into DNA sorption onto MPs show that there is potential for MPs to act as vectors for genetic material of interest, especially considering that particle-bound DNA typically persists longer in the environment than dissolved DNA.
Collapse
Affiliation(s)
- Nicolas D Müller
- Department of Environmental Systems Science, ETH Zurich, Universitätstrasse 16, 8092 Zurich, Switzerland
| | - Anish Kirtane
- Department of Environmental Systems Science, ETH Zurich, Universitätstrasse 16, 8092 Zurich, Switzerland
| | - Roman B Schefer
- Department of Environmental Systems Science, ETH Zurich, Universitätstrasse 16, 8092 Zurich, Switzerland
| | - Denise M Mitrano
- Department of Environmental Systems Science, ETH Zurich, Universitätstrasse 16, 8092 Zurich, Switzerland
| |
Collapse
|
41
|
Sharma DK, Rajpurohit YS. Multitasking functions of bacterial extracellular DNA in biofilms. J Bacteriol 2024; 206:e0000624. [PMID: 38445859 PMCID: PMC11025335 DOI: 10.1128/jb.00006-24] [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: 03/07/2024] Open
Abstract
Bacterial biofilms are intricate ecosystems of microbial communities that adhere to various surfaces and are enveloped by an extracellular matrix composed of polymeric substances. Within the context of bacterial biofilms, extracellular DNA (eDNA) originates from cell lysis or is actively secreted, where it exerts a significant influence on the formation, stability, and resistance of biofilms to environmental stressors. The exploration of eDNA within bacterial biofilms holds paramount importance in research, with far-reaching implications for both human health and the environment. An enhanced understanding of the functions of eDNA in biofilm formation and antibiotic resistance could inspire the development of strategies to combat biofilm-related infections and improve the management of antibiotic resistance. This comprehensive review encapsulates the latest discoveries concerning eDNA, encompassing its origins, functions within bacterial biofilms, and significance in bacterial pathogenesis.
Collapse
Affiliation(s)
- Dhirendra Kumar Sharma
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India
- Schools of Life Sciences, Homi Bhabha National Institute (DAE—Deemed University), Mumbai, India
| | - Yogendra Singh Rajpurohit
- Molecular Biology Division, Bhabha Atomic Research Centre, Mumbai, India
- Schools of Life Sciences, Homi Bhabha National Institute (DAE—Deemed University), Mumbai, India
| |
Collapse
|
42
|
Labadie M, Marchal F, Merbahi N, Girbal-Neuhauser E, Fontagné-Faucher C, Marcato-Romain CE. Cell density and extracellular matrix composition mitigate bacterial biofilm sensitivity to UV-C LED irradiation. Appl Microbiol Biotechnol 2024; 108:286. [PMID: 38578301 PMCID: PMC10997551 DOI: 10.1007/s00253-024-13123-4] [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: 05/14/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/06/2024]
Abstract
Ultraviolet-C light-emitting diodes (UV-C LEDs) are an emerging technology for decontamination applications in different sectors. In this study, the inactivation of bacterial biofilms was investigated by applying an UV-C LED emitting at 280 nm and by measuring both the influence of the initial cell density (load) and presence of an extracellular matrix (biofilm). Two bacterial strains exposing diverging matrix structures and biochemical compositions were used: Pseudomonas aeruginosa and Leuconostoc citreum. UV-C LED irradiation was applied at three UV doses (171 to 684 mJ/cm2) on both surface-spread cells and on 24-h biofilms and under controlled cell loads, and bacterial survival was determined. All surface-spread bacteria, between 105 and 109 CFU/cm2, and biofilms at 108 CFU/cm2 showed that bacterial response to irradiation was dose-dependent. The treatment efficacy decreased significantly for L. citreum surface-spread cells when the initial cell load was high, while no load effect was observed for P. aeruginosa. Inactivation was also reduced when bacteria were grown under a biofilm form, especially for P. aeruginosa: a protective effect could be attributed to abundant extracellular DNA and proteins in the matrix of P. aeruginosa biofilms, as revealed by Confocal Laser Scanning Microscopy observations. This study showed that initial cell load and exopolymeric substances are major factors influencing UV-C LED antibiofilm treatment efficacy. KEY POINTS: • Bacterial cell load (CFU/cm2) could impact UV-C LED irradiation efficiency • Characteristics of the biofilm matrix have a paramount importance on inactivation • The dose to be applied can be predicted based on biofilm properties.
Collapse
Affiliation(s)
- Maritxu Labadie
- Université de Toulouse, UPS, IUT Paul Sabatier, LBAE EA 4565 (Laboratoire de Biotechnologies Agroalimentaire Et Environnementale), 24 Rue d'Embaquès, Auch, F-32000, France
| | - Frédéric Marchal
- Université de Toulouse, UPS, INPT, LAPLACE UMR 5223 (Laboratoire Plasma Et Conversion d'Energie), 118 Route de Narbonne, Toulouse, F-31062, France
| | - Nofel Merbahi
- Université de Toulouse, UPS, INPT, LAPLACE UMR 5223 (Laboratoire Plasma Et Conversion d'Energie), 118 Route de Narbonne, Toulouse, F-31062, France
| | - Elisabeth Girbal-Neuhauser
- Université de Toulouse, UPS, IUT Paul Sabatier, LBAE EA 4565 (Laboratoire de Biotechnologies Agroalimentaire Et Environnementale), 24 Rue d'Embaquès, Auch, F-32000, France
| | - Catherine Fontagné-Faucher
- Université de Toulouse, UPS, IUT Paul Sabatier, LBAE EA 4565 (Laboratoire de Biotechnologies Agroalimentaire Et Environnementale), 24 Rue d'Embaquès, Auch, F-32000, France
| | - Claire-Emmanuelle Marcato-Romain
- Université de Toulouse, UPS, IUT Paul Sabatier, LBAE EA 4565 (Laboratoire de Biotechnologies Agroalimentaire Et Environnementale), 24 Rue d'Embaquès, Auch, F-32000, France.
| |
Collapse
|
43
|
Lu L, Zhao Y, Li M, Wang X, Zhu J, Liao L, Wang J. Contemporary strategies and approaches for characterizing composition and enhancing biofilm penetration targeting bacterial extracellular polymeric substances. J Pharm Anal 2024; 14:100906. [PMID: 38634060 PMCID: PMC11022105 DOI: 10.1016/j.jpha.2023.11.013] [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: 08/04/2023] [Revised: 11/08/2023] [Accepted: 11/26/2023] [Indexed: 04/19/2024] Open
Abstract
Extracellular polymeric substances (EPS) constitutes crucial elements within bacterial biofilms, facilitating accelerated antimicrobial resistance and conferring defense against the host's immune cells. Developing precise and effective antibiofilm approaches and strategies, tailored to the specific characteristics of EPS composition, can offer valuable insights for the creation of novel antimicrobial drugs. This, in turn, holds the potential to mitigate the alarming issue of bacterial drug resistance. Current analysis of EPS compositions relies heavily on colorimetric approaches with a significant bias, which is likely due to the selection of a standard compound and the cross-interference of various EPS compounds. Considering the pivotal role of EPS in biofilm functionality, it is imperative for EPS research to delve deeper into the analysis of intricate compositions, moving beyond the current focus on polymeric materials. This necessitates a shift from heavy reliance on colorimetric analytic methods to more comprehensive and nuanced analytical approaches. In this study, we have provided a comprehensive summary of existing analytical methods utilized in the characterization of EPS compositions. Additionally, novel strategies aimed at targeting EPS to enhance biofilm penetration were explored, with a specific focus on highlighting the limitations associated with colorimetric methods. Furthermore, we have outlined the challenges faced in identifying additional components of EPS and propose a prospective research plan to address these challenges. This review has the potential to guide future researchers in the search for novel compounds capable of suppressing EPS, thereby inhibiting biofilm formation. This insight opens up a new avenue for exploration within this research domain.
Collapse
Affiliation(s)
- Lan Lu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Yuting Zhao
- Meishan Pharmaceutical Vocational College, School of Pharmacy, Meishan, Sichuan, 620200, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaobo Wang
- Hepatobiliary Surgery, Langzhong People's Hospital, Langzhong, Sichuan, 646000, China
| | - Jie Zhu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Li Liao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Jingya Wang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| |
Collapse
|
44
|
Hu X, Du X, Li M, Sun J, Li X, Pang X, Lu Y. Preparation and characterization of nisin-loaded chitosan nanoparticles functionalized with DNase I for the removal of Listeria monocytogenes biofilms. J Food Sci 2024; 89:2305-2315. [PMID: 38369953 DOI: 10.1111/1750-3841.16976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 02/20/2024]
Abstract
Listeria monocytogenes biofilms represent a continuous source of contamination, leading to serious food safety concerns and economic losses. This study aims to develop novel nisin-loaded chitosan nanoparticles (CSNPs) functionalized with DNase I and evaluate its antibiofilm activity against L. monocytogenes on food contact surfaces. Nisin-loaded CSNPs (CS-N) were first prepared by ionic cross-linking, and DNase I was covalently grafted on the surface (DNase-CS-N). The NPs were subsequently characterized by Zetasizer Nano, transmission electron microscopy, Fourier transform infrared (FT-IR), and X-ray diffraction (XRD). The antibiofilm activity of NPs was evaluated against L. monocytogenes on polyurethane (PU). The DNase-CS-N was fabricated and characterized with quality attributes (particle size-427.0 ± 15.1 nm, polydispersity [PDI]-0.114 ± 0.034, zeta potential-+52.5 ± 0.2 mV, encapsulation efficiency-46.5% ± 3.6%, DNase conjugate rate-70.4% ± 0.2). FT-IR and XRD verified the loading of nisin and binding of DNase I with chitosan. The DNase-CS-N caused a 3 log colony-forming unit (CFU)/cm2 reduction of L. monocytogenes biofilm cells, significantly higher than those in CSNPs (1.4 log), CS-N (1.8 log), and CS-N in combination with DNase I (2.2 log) treatment groups. In conclusion, nisin-loaded CSNPs functionalized with DNase I were successfully prepared and characterized with smooth surface and nearly spherical shape, high surface positive charge, and good stability, which is effective to eradicate L. monocytogenes biofilm cells on food contact surfaces, exhibiting great potential as antibiofilm agents in food industry. PRACTICAL APPLICATION: Listeria monocytogenes biofilms are a common safety hazard in food processing. In this study, novel nanoparticles were successfully constructed and are expected to be a promising antibiofilm agent in the food industry.
Collapse
Affiliation(s)
- Xin Hu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Xueying Du
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Mingwei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| |
Collapse
|
45
|
Jeong GJ, Khan F, Tabassum N, Cho KJ, Kim YM. Bacterial extracellular vesicles: Modulation of biofilm and virulence properties. Acta Biomater 2024; 178:13-23. [PMID: 38417645 DOI: 10.1016/j.actbio.2024.02.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/01/2024]
Abstract
Microbial pathogens cause persistent infections by forming biofilms and producing numerous virulence factors. Bacterial extracellular vesicles (BEVs) are nanostructures produced by various bacterial species vital for molecular transport. BEVs include various components, including lipids (glycolipids, LPS, and phospholipids), nucleic acids (genomic DNA, plasmids, and short RNA), proteins (membrane proteins, enzymes, and toxins), and quorum-sensing signaling molecules. BEVs play a major role in forming extracellular polymeric substances (EPS) in biofilms by transporting EPS components such as extracellular polysaccharides, proteins, and extracellular DNA. BEVs have been observed to carry various secretory virulence factors. Thus, BEVs play critical roles in cell-to-cell communication, biofilm formation, virulence, disease progression, and resistance to antimicrobial treatment. In contrast, BEVs have been shown to impede early-stage biofilm formation, disseminate mature biofilms, and reduce virulence. This review summarizes the current status in the literature regarding the composition and role of BEVs in microbial infections. Furthermore, the dual functions of BEVs in eliciting and suppressing biofilm formation and virulence in various microbial pathogens are thoroughly discussed. This review is expected to improve our understanding of the use of BEVs in determining the mechanism of biofilm development in pathogenic bacteria and in developing drugs to inhibit biofilm formation by microbial pathogens. STATEMENT OF SIGNIFICANCE: Bacterial extracellular vesicles (BEVs) are nanostructures formed by membrane blebbing and explosive cell lysis. It is essential for transporting lipids, nucleic acids, proteins, and quorum-sensing signaling molecules. BEVs play an important role in the formation of the biofilm's extracellular polymeric substances (EPS) by transporting its components, such as extracellular polysaccharides, proteins, and extracellular DNA. Furthermore, BEVs shield genetic material from nucleases and thermodegradation by packaging it during horizontal gene transfer, contributing to the transmission of bacterial adaptation determinants like antibiotic resistance. Thus, BEVs play a critical role in cell-to-cell communication, biofilm formation, virulence enhancement, disease progression, and drug resistance. In contrast, BEVs have been shown to prevent early-stage biofilm, disperse mature biofilm, and reduce virulence characteristics.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Institute of Fisheries Sciences, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Kyung-Jin Cho
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
46
|
Zhang J, Huang L, Wang Y. Changes in the level of biofilm development significantly affect the persistence of environmental DNA in flowing water. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 917:170162. [PMID: 38244634 DOI: 10.1016/j.scitotenv.2024.170162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/27/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
As one of the powerful tools of species biomonitoring, the utilization of environmental DNA (eDNA) technology is progressively expanding in both scope and frequency within the field of ecology. Nonetheless, the growing dissemination of this technology has brought to light a multitude of intricate issues. The complex effects of environmental factors on the persistence of eDNA in water have brought many challenges to the interpretation of eDNA information. In this study, the primary objective was to examine how variations in the presence and development of biofilms impact the persistence of grass carp eDNA under different sediment types and flow conditions. This investigation encompassed the processes of eDNA removal and resuspension in water, shedding light on the complex interactions involved. The findings reveal that with an elevated biofilm development level, the total removal rate of eDNA gradually rose, resulting in a corresponding decrease in its residence time within the mesocosms. The influence of biofilms on the persistence of grass carp eDNA is more pronounced under flowing water conditions. However, changes in bottom sediment types did not significantly interact with biofilms. Lastly, in treatments involving alternating flow conditions between flowing and still water, significant resuspension of grass carp eDNA was not observed due to interference from multiple factors, including the effect of biofilms. Our study offers preliminary insights into the biofilm-mediated mechanisms of aquatic eDNA removal, emphasizing the need for careful consideration of environmental factors in the practical application of eDNA technology for biomonitoring in natural aquatic environments.
Collapse
Affiliation(s)
- Jianmin Zhang
- State Key Laboratory of Hydraulics and Mountain River Engineering, Sichuan University, First Ring Road 24#, Chengdu 610065, People's Republic of China.
| | - Lei Huang
- State Key Laboratory of Hydraulics and Mountain River Engineering, Sichuan University, First Ring Road 24#, Chengdu 610065, People's Republic of China.
| | - Yurong Wang
- State Key Laboratory of Hydraulics and Mountain River Engineering, Sichuan University, First Ring Road 24#, Chengdu 610065, People's Republic of China.
| |
Collapse
|
47
|
Song M, Tang Q, Ding Y, Tan P, Zhang Y, Wang T, Zhou C, Xu S, Lyu M, Bai Y, Ma X. Staphylococcus aureus and biofilms: transmission, threats, and promising strategies in animal husbandry. J Anim Sci Biotechnol 2024; 15:44. [PMID: 38475886 PMCID: PMC10936095 DOI: 10.1186/s40104-024-01007-6] [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: 11/04/2023] [Accepted: 02/03/2024] [Indexed: 03/14/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is a common pathogenic bacterium in animal husbandry that can cause diseases such as mastitis, skin infections, arthritis, and other ailments. The formation of biofilms threatens and exacerbates S. aureus infection by allowing the bacteria to adhere to pathological areas and livestock product surfaces, thus triggering animal health crises and safety issues with livestock products. To solve this problem, in this review, we provide a brief overview of the harm caused by S. aureus and its biofilms on livestock and animal byproducts (meat and dairy products). We also describe the ways in which S. aureus spreads in animals and the threats it poses to the livestock industry. The processes and molecular mechanisms involved in biofilm formation are then explained. Finally, we discuss strategies for the removal and eradication of S. aureus and biofilms in animal husbandry, including the use of antimicrobial peptides, plant extracts, nanoparticles, phages, and antibodies. These strategies to reduce the spread of S. aureus in animal husbandry help maintain livestock health and improve productivity to ensure the ecologically sustainable development of animal husbandry and the safety of livestock products.
Collapse
Affiliation(s)
- Mengda Song
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Key Laboratory of Innovative Utilization of Local Cattle and Sheep Germplasm Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Qi Tang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yakun Ding
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Peng Tan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yucheng Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Tao Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chenlong Zhou
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shenrui Xu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Mengwei Lyu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yueyu Bai
- Key Laboratory of Innovative Utilization of Local Cattle and Sheep Germplasm Resources (Co-Construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Xi Ma
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
48
|
Jiang F, Wang J, Ren Z, Hu Y, Wang B, Li M, Yu J, Tang J, Guo G, Cheng Y, Han P, Shen H. Targeted Light-Induced Immunomodulatory Strategy for Implant-Associated Infections via Reversing Biofilm-Mediated Immunosuppression. ACS NANO 2024; 18:6990-7010. [PMID: 38385433 DOI: 10.1021/acsnano.3c10172] [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: 02/23/2024]
Abstract
The clinical treatment efficacy for implant-associated infections (IAIs), particularly those caused by Methicillin-resistant Staphylococcus aureus (MRSA), remains unsatisfactory, primarily due to the formation of biofilm barriers and the resulting immunosuppressive microenvironment, leading to the chronicity and recurrence of IAIs. To address this challenge, we propose a light-induced immune enhancement strategy, synthesizing BSA@MnO2@Ce6@Van (BMCV). The BMCV exhibits precise targeting and adhesion to the S. aureus biofilm-infected region, coupled with its capacity to catalyze oxygen generation from H2O2 in the hypoxic and acidic biofilm microenvironment (BME), promoting oxygen-dependent photodynamic therapy efficacy while ensuring continuous release of manganese ions. Notably, targeted BMCV can penetrate biofilms, producing ROS that degrade extracellular DNA, disrupting the biofilm structure and impairing its barrier function, making it vulnerable to infiltration and elimination by the immune system. Furthermore, light-induced reactive oxygen species (ROS) around the biofilm can lyse S. aureus, triggering bacterium-like immunogenic cell death (ICD), releasing abundant immune costimulatory factors, facilitating the recognition and maturation of antigen-presenting cells (APCs), and activating adaptive immunity. Additionally, manganese ions in the BME act as immunoadjuvants, further amplifying macrophage-mediated innate and adaptive immune responses and reversing the immunologically cold BME to an immunologically hot BME. We prove that our synthesized BMCV elicits a robust adaptive immune response in vivo, effectively clearing primary IAIs and inducing long-term immune memory to prevent recurrence. Our study introduces a potent light-induced immunomodulatory nanoplatform capable of reversing the biofilm-induced immunosuppressive microenvironment and disrupting biofilm-mediated protective barriers, offering a promising immunotherapeutic strategy for addressing challenging S. aureus IAIs.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jian Wang
- Department of Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Zun Ren
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yujie Hu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Boyong Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Mingzhang Li
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jinlong Yu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jin Tang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Geyong Guo
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yingsheng Cheng
- Department of Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Department of Imaging Medicine and Nuclear Medicine, Tongji Hospital, Shanghai 200065, China
| | - Pei Han
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hao Shen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| |
Collapse
|
49
|
Cui Y, Zhang W, Shan J, He J, Niu Q, Zhu C, Wang W, Chen XL, Wang X. Copper Nanodots-Based Hybrid Hydrogels with Multiple Enzyme Activities for Acute and Infected Wound Repair. Adv Healthc Mater 2024; 13:e2302566. [PMID: 37931140 DOI: 10.1002/adhm.202302566] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/18/2023] [Indexed: 11/08/2023]
Abstract
Effectively controlling bacterial infection, reducing the inflammation and promoting vascular regeneration are all essential strategies for wound repair. Nanozyme technology has potential applications in the treatment of infections because its non-antibiotic dependent, topical and noninvasive nature. In wound management, copper-based nanozymes have emerged as viable alternatives to antibiotics. In this study, an ultrasmall cupric enzyme with high enzymatic activity is synthesized and added to a nontoxic, self-healing, injectable cationic guar gum (CG) hydrogel network. The nanozyme exhibits remarkable antioxidant properties under neutral conditions, effectively scavenging reactive nitrogen and oxygen species (RNOS). Under acidic conditions, Cu NDs have peroxide (POD) enzyme-like activity, which allows them to eliminate hydrogen peroxides and produce free radicals locally. Antibacterial experiments show that they can kill bacteria and remove biofilms. It reveals that low concentrations of Cu ND/CG decrease the expression of the inflammatory factors in cells and tissues, effectively controlling inflammatory responses. Cu ND/CG hydrogels also inhibit HIF-1α and promote VEGF expression in the wound with the ability to promote vascular regeneration. In vivo safety assessments reveal a favorable biosafety profile. Cu ND/CG hydrogels offer a promising solution for treating acute and infected wounds, highlighting the potential of innovative nanomaterials in wound healing.
Collapse
Affiliation(s)
- Yuyu Cui
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Wei Zhang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China
| | - Jie Shan
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Jia He
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Qiang Niu
- Department of Clinical Medicine, The First School of Clinical Medicine, Anhui Medical University, Hefei, 230032, China
| | - Can Zhu
- Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, Hefei, 230032, China
| | - Wenqi Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, 230032, China
- College and Hospital of Stomatology, Key Lab. of Oral Diseases Research of Anhui Province, Anhui Medical University, Hefei, 230032, P. R. China
| |
Collapse
|
50
|
Wang X, Wang D, Lu H, Wang X, Wang X, Su J, Xia G. Strategies to Promote the Journey of Nanoparticles Against Biofilm-Associated Infections. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305988. [PMID: 38178276 DOI: 10.1002/smll.202305988] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/08/2023] [Indexed: 01/06/2024]
Abstract
Biofilm-associated infections are one of the most challenging healthcare threats for humans, accounting for 80% of bacterial infections, leading to persistent and chronic infections. The conventional antibiotics still face their dilemma of poor therapeutic effects due to the high tolerance and resistance led by bacterial biofilm barriers. Nanotechnology-based antimicrobials, nanoparticles (NPs), are paid attention extensively and considered as promising alternative. This review focuses on the whole journey of NPs against biofilm-associated infections, and to clarify it clearly, the journey is divided into four processes in sequence as 1) Targeting biofilms, 2) Penetrating biofilm barrier, 3) Attaching to bacterial cells, and 4) Translocating through bacterial cell envelope. Through outlining the compositions and properties of biofilms and bacteria cells, recent advances and present the strategies of each process are comprehensively discussed to combat biofilm-associated infections, as well as the combined strategies against these infections with drug resistance, aiming to guide the rational design and facilitate wide application of NPs in biofilm-associated infections.
Collapse
Affiliation(s)
- Xiaobo Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Dan Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Hongwei Lu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Xiaowei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Xuelei Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Jiayi Su
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| | - Guimin Xia
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, P. R. China
| |
Collapse
|