1
|
He X, Zhang W, Liu J, Liu J, Chen Y, Luan C, Zhang J, Bao G, Lin X, Muh F, Lin T, Lu F. The global regulatory role of RsbUVW in virulence and biofilm formation in MRSA. Microb Pathog 2025; 203:107508. [PMID: 40158706 DOI: 10.1016/j.micpath.2025.107508] [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: 12/26/2024] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
The widespread prevalence of methicillin-resistant Staphylococcus aureus (MRSA) has caused serious challenges to clinical treatment. This study was designed to explore effective targets for MRSA prevention and control. The key virulence regulator was screened through the correlation analysis between virulence and various regulatory factors in the main clinical epidemic MRSA. The potential key factors were inactivated to further evaluate the inhibitory effect on the virulence of MRSA standard strain S. aureus ATCC43300 and its influence on drug resistance and biofilm formation. Enterobacterial repetitive intergenic consensus-PCR was used to analyze the clinical epidemic genotypes of MRSA. The virulence of MRSA was evaluated mainly by measuring its adhesion and invasion ability to A549 cells, the lethality to Galleria mellonella larvae, and the transcription level of related genes. The biofilm formation was assessed by crystal violet staining on polystyrene microplates. The results showed that most virulence factors of clinical representative MRSA strain were significantly positively correlated with RsbUVW system. After knocking out the rsbV gene, a key component of the rsbUVW system, the virulence of S. aureus ATCC43300 was significantly reduced (P < 0.05), as indicated by a significant decrease in lethality against G. mellonella larvae and invasion against A549 cells, and a significant decrease in the expression of immune escape related virulence factors polysaccharide intercellular adhesin (PIA) and staphyloxanthin. The biomass and stability of protein-dependent biofilm by S. aureus ATCC43300 were significantly increased. This study will provide useful information for the effective prevention and control of MRSA.
Collapse
Affiliation(s)
- Xinlong He
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China; The Key Laboratory of the Jiangsu Higher Education Institutions for Nucleic Acid & Cell Fate Regulation (Yangzhou University), Yangzhou, 225001, China; Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou, 225001, China
| | - Wenwen Zhang
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China; Department of Clinical Laboratory, Changning Maternity and Infant Health Hospital, Affiliated Hospital of East China Normal University, Shanghai, 200050, China
| | - Jie Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Jiali Liu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yinsong Chen
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Changjiao Luan
- Department of Lung, Third People's Hospital of Yangzhou, Yangzhou, China
| | - Jun Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Guangyu Bao
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Xiangfang Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China
| | - Fauzi Muh
- Department of Epidemiology & Tropical Diseases, Faculty of Public Health, Universitas Diponegoro, Tembalang, Semarang, 50275, Indonesia
| | - Tao Lin
- Department of Clinical Laboratory, First Affiliated Hospital of Yangzhou University, Yangzhou, 225000, China.
| | - Feng Lu
- Department of Pathogenic Biology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
2
|
Lee WH, Zygiel EM, Lee CH, Oglesby AG, Nolan EM. Calprotectin-mediated survival of Staphylococcus aureus in coculture with Pseudomonas aeruginosa occurs without nutrient metal sequestration. mBio 2025; 16:e0384624. [PMID: 40152583 PMCID: PMC12077171 DOI: 10.1128/mbio.03846-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/05/2025] [Indexed: 03/29/2025] Open
Abstract
Pseudomonas aeruginosa and Staphylococcus aureus are bacterial pathogens of major clinical concern that cause polymicrobial infections in diverse patient populations. Human calprotectin (CP; S100A8/S100A9 heterooligomer, MRP8/MRP14 heterooligomer) is a host-defense protein that contributes to nutritional immunity by sequestering multiple nutrient metal ions including Mn(II), Fe(II), and Zn(II). Here, we examine the consequences of metal availability and CP treatment on cocultures of P. aeruginosa and S. aureus. We report that CP elicits Fe-starvation responses in both P. aeruginosa and S. aureus in coculture, including the upregulation of genes involved in Fe uptake by both organisms. Moreover, analyses of pseudomonal metabolites in coculture supernatants further demonstrate Fe-starvation responses, showing that CP treatment leads to increased siderophore levels and reduced phenazine levels. Consistent with prior studies, growth under conditions of Fe depletion accelerated P. aeruginosa killing of S. aureus in coculture, but treatment with CP promoted S. aureus survival. Treatment with CP site variants lacking functional transition-metal-binding sites and metalated CP also enhanced S. aureus survival in coculture with P. aeruginosa, revealing that this consequence of CP treatment is independent of its canonical metal-sequestering function. Thus, the protective effects of CP treatment during coculture appear to override the observed Fe-starvation effects that make P. aeruginosa more virulent toward S. aureus. This work highlights an unappreciated facet of how CP contributes to host-pathogen and pathogen-pathogen interactions that are relevant to human infectious disease. IMPORTANCE The current working model that describes how the innate immune protein calprotectin (CP) protects the host against bacterial pathogens focuses on its capacity to sequester multiple essential metal nutrients in a process called nutritional immunity. Our study further explores this function by focusing on the effects of metal availability and CP treatment on the dynamics of Pseudomonas aeruginosa and Staphylococcus aureus grown in coculture. These two bacterial pathogens are of significant clinical concern and colocalize with CP at infection sites. This work reveals that CP modulates P. aeruginosa/S. aureus coculture dynamics in a manner that is independent of its ability to sequester nutrient metal ions. This surprising result is important because it demonstrates that CP has metal-independent function and thus contributes to the host-pathogen and pathogen-pathogen interactions in ways that are not accounted for in the current working model focused on metal sequestration.
Collapse
Affiliation(s)
- Wei H. Lee
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Emily M. Zygiel
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Celis H. Lee
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Amanda G. Oglesby
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Elizabeth M. Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
3
|
He J, Cui Y, Liu Y, Mao J, Dong Y, Yao R, Yang D, Fan P, Xue J. Resveratrol inhibits the formation of Staphylococcus aureus biofilms by reducing PIA, eDNA release, and ROS production. Front Vet Sci 2025; 12:1594239. [PMID: 40370837 PMCID: PMC12076522 DOI: 10.3389/fvets.2025.1594239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 03/31/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Staphylococcus aureus is a zoonotic pathogen that is difficult to control. Resveratrol (RES) has been shown to have significant antibacterial effects. The present study aimed to investigate the inhibitory effect of RES on the formation of Staphylococcus aureus biofilms and their molecular mechanism. Methods First, the minimum inhibitory concentration and inhibitory action curve of RES against Staphylococcus aureus were obtained through testing. Second, we found that RES can inhibit biofilm formation by reducing the release of polysaccharide intercellular adhesion (PIA) and extracellular DNA (eDNA) from Staphylococcus aureus. Results RES treatment significantly reduced the production of reactive oxygen species (ROS) and nicotinamide adenine dinucleotide phosphate (NADPH) in Staphylococcus aureus, indicating that ROS and NADPH are closely related to biofilm formation. Conclusion This study demonstrates that RES inhibits the formation of Staphylococcus aureus biofilms by reducing PIA, eDNA release, and ROS production, and these results provide new ideas for the clinical application of RES in the treatment of Staphylococcus aureus infection.
Collapse
Affiliation(s)
- Jinfei He
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Yilong Cui
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Yan Liu
- College of Life Sciences and Food Engineering, Inner Mongolia MINZU University, Tongliao, China
| | - Jingdong Mao
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Yanxin Dong
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Ruizhi Yao
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Dahan Yang
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Peichao Fan
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| | - Jiangdong Xue
- College of Animal Science and Technology, Inner Mongolia MINZU University, Tongliao, China
| |
Collapse
|
4
|
Abreu-Pereira CA, Gorayb-Pereira AL, Jordão CC, Paro CB, Barbugli PA, Pavarina AC. Zerumbone enhances the photodynamic effect against biofilms of fluconazole-resistant Candida albicans clinical isolates. J Dent 2025; 155:105631. [PMID: 39956310 DOI: 10.1016/j.jdent.2025.105631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/18/2025] Open
Abstract
The aim was the evaluation of Zerumbone (ZER) associated with Photodithazine® (PDZ) mediated antimicrobial photodynamic therapy (aPDT) on fluconazole-resistant (CaR) and -susceptible (CaS) Candida albicans biofilms, and well as two fluconazole-resistant clinical isolates (R70 and R14). Biofilms (48 h) were treated with ZER (256 µg/mL) for 20 min, followed by the absence or presence of the photosensitizer (PDZ; 200 mg/L) and Light Emitting Diode (LED; 600 nm; 50 Jcm2), either alone or in combination, resulting in 8 treatment groups (Control; ZER; aPDT; PDZ; LED; ZER+aPDT; ZER+PDZ; ZER+LED; n = 12). The treatments were evaluated by colony-forming unit (CFU), total and insoluble biomass, extracellular matrix components (ECM) quantification [extracellular DNA (eDNA), proteins, alkali (ASP) and water-soluble polysaccharides (WSP)], and Confocal Laser Scanning Microscopy (CLSM). Data were subjected to three-way ANOVA with Bonferroni post-test (α 5 %). The combination of ZER and aPDT promoted a greater reduction of biofilms' components for all the strains evaluated (CaS, CaR, R14, R70). On average, ZER+aPDT group showed a 2.01 log10 (30 %) reduction in CFU counts, total biomass (30 %), insoluble biomass (33 %), and total protein (15 %). Additionally, there was a reduction in ECM components of the biofilms, such as insoluble proteins (24 %), WSP (68 %), ASP (26 %) and eDNA (60 %) compared to the control group. Images obtained through CLSM showed that the association of ZER+aPDT resulted in a higher content of dead cells. Treatment with ZER+aPDT enhanced the efficacy of photodynamic treatment on fluconazole-resistant C. albicans (including clinical isolates), representing an encouraging antifungal approach.
Collapse
Affiliation(s)
- César Augusto Abreu-Pereira
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil
| | - Ana Luiza Gorayb-Pereira
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil
| | - Cláudia Carolina Jordão
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil
| | - Clara Brait Paro
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil
| | - Paula Aboud Barbugli
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil
| | - Ana Claudia Pavarina
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil.
| |
Collapse
|
5
|
Wang P, Zeng Y, Liu J, Wang L, Yang M, Zhou J. Antimicrobial and anti-biofilm effects of dihydroartemisinin-loaded chitosan nanoparticles against methicillin-resistant Staphylococcus aureus. Microb Pathog 2025; 199:107208. [PMID: 39657894 DOI: 10.1016/j.micpath.2024.107208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
The formation of biofilms enhances bacterial antibiotic resistance, posing significant challenges to clinical treatment. Methicillin-resistant Staphylococcus aureus (MRSA) is a primary pathogen in biofilm-associated infections. Its high antibiotic resistance and incidence rates make it a major clinical challenge, underscoring the urgent need for novel therapeutic strategies. Building on previous research, this study employs nanotechnology to fabricate dihydroartemisinin-chitosan nanoparticles (DHA-CS NPs) and, for the first time, applies them to the treatment of MRSA biofilm infections. The antibacterial and anti-biofilm activities of these compounds were evaluated, and their potential mechanisms of action were preliminarily explored. The results demonstrated that the DHA-CS NPs exhibited a minimum inhibitory concentration (MIC) of15 μg/mLand a minimum bactericidal concentration (MBC) of 30 μg/mL. At 15 μg/mL, the DHA-CS NPs significantly inhibited MRSA biofilm formation (P < 0.001),while at 7.5 μg/mL, they dispersed 67.4 ± 3.77 % of the preformed biofilms (P < 0.001). Scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM) confirmed the disruption of MRSA biofilms. Mechanistic studies, including phenol-sulfuric acid assays, static biofilm microtiter plate assays, and RT-qPCR, revealed that the DHA-CS NPs inhibited the synthesis of extracellular polymeric substances (EPS), suppressed the release of extracellular DNA (eDNA), and downregulated key biofilm-related genes (icaA, sarA, cidA, and agrA). These findings suggest that DHA-CS NPs hold significant promise for inhibiting and eradicating MRSA biofilms, providing a theoretical basis for the development of novel antibiofilm therapies.
Collapse
Affiliation(s)
- Peike Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yali Zeng
- Mianyang 404 Hospital, Mianyang, China.
| | - Jinbo Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| | - Lin Wang
- Mianyang 404 Hospital, Mianyang, China
| | - Min Yang
- School of Public Health, Southwest Medical University, Luzhou, China
| | - Jian Zhou
- School of Public Health, Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Xia Y, Hu Z, Jin Q, Chen Q, Zhao C, Qiang R, Xie Z, Li L, Zhang H. Structural characteristics, functions, and counteracting strategies of biofilms in Staphylococcus aureus. Comput Struct Biotechnol J 2025; 27:488-500. [PMID: 39916696 PMCID: PMC11799891 DOI: 10.1016/j.csbj.2025.01.021] [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: 11/16/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
Background Staphylococcus aureus (S. aureus) is a prevalent pathogen associated with a wide range of infections, exhibiting significant antibiotic resistance and posing therapeutic challenges in clinical settings. The formation of biofilms contributes to the emergence of resistant strains, further diminishing the efficacy of antibiotics. This, in turn, leads to chronic and recurrent infections, ultimately increasing the healthcare burden. Consequently, preventing and eliminating biofilms has become a critical focus in clinical management and research. Aim of review This review systematically examines the mechanisms underlying biofilm formation in S. aureus and its contribution to antibiotic resistance, emphasizing the essential roles biofilms play in maintaining structural integrity and enhancing resistance. It also analyses the protective mechanisms that fortify S. aureus biofilms against antimicrobial treatments. Furthermore, the review provides a comprehensive overview of recent therapeutic innovations, including enzymatic therapy, nanotechnology, gene editing, and phage therapy. Key scientific concepts of review Emerging therapeutic strategies present novel approaches to combat S. aureus biofilm-associated infections through various mechanisms. This review discusses recent advancements in these therapies, their practical challenges in clinical application, and provides an in-depth analysis of each strategy's mechanisms and therapeutic potential. By mapping future research directions, this review aims to refine anti-biofilm strategies to control infection progression and effectively mitigate recurrence.
Collapse
Affiliation(s)
- Yanze Xia
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhenghui Hu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiyuan Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chenhao Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Qiang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zonggang Xie
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liubing Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| |
Collapse
|
7
|
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
|
8
|
Gorayb Pereira AL, Augusto Abreu Pereira C, Dias LM, Jorge JH, Pavarina AC. Zerumbone disrupts mixed biofilms of Candida albicans and Streptococcus mutans on acrylic resin. BIOFOULING 2024:1-16. [PMID: 39704392 DOI: 10.1080/08927014.2024.2441259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/01/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Abstract
The efficacy of Zerumbone (ZER) against mixed biofilms of fluconazole-resistant Candida albicans (ATCC 96901) and Streptococcus mutans (UA159) was evaluated. Biofilms were cultivated on acrylic resin specimens for 48 h, with alternating supplementation of glucose and sucrose. ZER's ability to inhibit biofilm formation (pre-treatment) and eradicate mature biofilms (post-treatment) was assessed. Control groups were treated with Chlorhexidine (CHX), Nystatin (NYS), Penicillin (ATB), and distilled water. The efficacy was measured by colony forming units (CFU/mm2) counts, biomass and biofilm's matrix components quantification (water-soluble polysaccharides [WSP], alkali-soluble polysaccharides [ASPs], proteins, and extracellular DNA [eDNA]). Data were analyzed by one-way ANOVA with Tukey's or Gammes-Howell post-hoc test for normal data and Kruskal-Wallis test for data that did not meet the assumption of normality (α = 0,05). In the biofilm inhibition assay, ZER decreased total microbiota (C. albicans + S. mutans) (2.7 log10; p < 0.005), C. albicans (1.4 log10; p < 0.038) and S. mutans (1.9 log10; p < 0.048) counting (vs control group), and biofilm components [insoluble proteins: 37% (p < 0.001); WSP: 13% (p < 0.042); ASP: 46% (p < 0.001); eDNA: 11% (p < 0.048)]. Post-treatment with ZER reduced total microbiota (3.2 log10; p < 0.001), C. albicans (3 log10; p < 0.001) and S. mutans (2 log10; p < 0.001) counting (vs control group), and biofilm components [soluble proteins: 20% (p < 0.001); WSP: 20% (p < 0.001); ASP: 51% (p < 0.001); and eDNA: 33% (p < 0.001)]. The positive control groups demonstrated similar or lower efficacy than ZER under all experimental conditions. ZER demonstrates efficacy against mixed biofilms by reducing C. albicans and S. mutans counting and disrupting the extracellular matrix in both assays.
Collapse
Affiliation(s)
- Ana Luíza Gorayb Pereira
- Laboratory of Applied Microbiology Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, SP, Brazil
| | - César Augusto Abreu Pereira
- Laboratory of Applied Microbiology Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, SP, Brazil
| | - Luana Mendonça Dias
- Laboratory of Applied Microbiology Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, SP, Brazil
| | - Janaína Habib Jorge
- Laboratory of Applied Microbiology Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, SP, Brazil
| | - Ana Cláudia Pavarina
- Laboratory of Applied Microbiology Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, SP, Brazil
| |
Collapse
|
9
|
Kashi M, Noei M, Chegini Z, Shariati A. Natural compounds in the fight against Staphylococcus aureus biofilms: a review of antibiofilm strategies. Front Pharmacol 2024; 15:1491363. [PMID: 39635434 PMCID: PMC11615405 DOI: 10.3389/fphar.2024.1491363] [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: 09/04/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Staphylococcus aureus is an important pathogen due to its ability to form strong biofilms and antibiotic resistance. Biofilms play an important role in bacterial survival against the host immune system and antibiotics. Natural compounds (NCs) have diverse bioactive properties with a low probability of resistance, making them promising candidates for biofilm control. NC such as curcumin, cinnamaldehyde, carvacrol, eugenol, thymol, citral, linalool, 1,8-cineole, pinene, cymene, terpineol, quercetin, and limonene have been widely utilized for the inhibition and destruction of S. aureus biofilms. NCs influence biofilm formation through several procedures. Some of the antibiofilm mechanisms of NCs are direct bactericidal effect, disrupting the quorum sensing system, preventing bacteria from aggregation and attachment to surfaces, reducing the microbial surface components recognizing adhesive matrix molecules (MSCRAMMs), interfering with sortase A enzyme, and altering the expression of biofilm-associated genes such as icaADBC, agr, and sarA. Furthermore, these compounds affect extracellular polymeric substances (EPS) and their components, such as polysaccharide intercellular adhesin (PIA) and eDNA. However, some disadvantages, such as low water solubility and bioavailability, limit their clinical usage. Therefore, scientists have considered using nanotechnology and drug platforms to improve NC's efficacy. Some NC, such as thymol and curcumin, can also enhance photodynamic therapy against S. aurous biofilm community. This article evaluates the anti-biofilm potential of NC, their mechanisms of action against S. aureus biofilms, and various aspects of their application.
Collapse
Affiliation(s)
- Milad Kashi
- Student Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Milad Noei
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
10
|
AlQadeeb H, Baltazar M, Cazares A, Poonpanichakul T, Kjos M, French N, Kadioglu A, O’Brien M. The Streptococcus agalactiae LytSR two-component regulatory system promotes vaginal colonization and virulence in vivo. Microbiol Spectr 2024; 12:e0197024. [PMID: 39400158 PMCID: PMC11537067 DOI: 10.1128/spectrum.01970-24] [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] [Received: 08/06/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Streptococcus agalactiae (or group B Streptococcus, GBS) is a leading cause of neonatal sepsis and meningitis globally. To sense and respond to variations in its environment, GBS possesses multiple two-component regulatory systems (TCSs), such as LytSR. Here, we aimed to investigate the role of LytSR in GBS pathogenicity. We generated an isogenic lytS knockout mutant in a clinical GBS isolate and used a combination of phenotypic in vitro assays and in vivo murine models to investigate the contribution of lytS to the colonization and invasive properties of GBS. Deletion of the lytS gene in the GBS chromosome resulted in significantly higher survival rates in mice during sepsis, accompanied by reduced bacterial loads in blood, lung, spleen, kidney, and brain tissues compared to infection with the wild-type strain. In a mouse model of GBS vaginal colonization, we also observed that the lytS knockout mutant was cleared more readily from the vaginal tract compared to its wild-type counterpart. Interestingly, lower levels of proinflammatory cytokines were found in the serum of mice infected with the lytS mutant. Our results demonstrate that the LytSR TCS plays a key role in GBS tissue invasion and pathogenesis, and persistence of mucosal colonization.IMPORTANCEStreptococcus agalactiae (group B Streptococcus, or GBS) is a common commensal of the female urogenital tract and one of WHO's priority pathogens. The bacterium has evolved mechanisms to adapt and survive in its host, many of which are regulated via two-component signal transduction systems (TCSs); however, the exact contributions of TCSs toward GBS pathogenicity remain largely obscure. We have constructed a TCS lytS-deficient mutant in a CC-17 hypervirulent GBS clinical isolate. Using murine models, we showed that LytSR regulatory system is essential for vaginal colonization via promoting biofilm production. We also observed that lytS deficiency led to significantly attenuated virulence properties and lower levels of proinflammatory cytokines in blood. Our findings are of significant importance in that they unveil a previously unreported role for LytSR in GBS and pave the way toward a better understanding of its ability to transition from an innocuous commensal to a deadly pathogen.
Collapse
Affiliation(s)
- Hajar AlQadeeb
- Department of Medical Laboratory, College of Applied Medical Sciences in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Murielle Baltazar
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Adrian Cazares
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Tiraput Poonpanichakul
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Nakhon Pathom, Thailand
| | - Morten Kjos
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Neil French
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Aras Kadioglu
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
| | - Marie O’Brien
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, United Kingdom
- ReNewVax Ltd, Liverpool, United Kingdom
| |
Collapse
|
11
|
Pandey S, Kannaujiya VK. Bacterial extracellular biopolymers: Eco-diversification, biosynthesis, technological development and commercial applications. Int J Biol Macromol 2024; 279:135261. [PMID: 39244116 DOI: 10.1016/j.ijbiomac.2024.135261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/22/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Synthetic polymers have been widely thriving as mega industries at a commercial scale in various commercial sectors over the last few decades. The extensive use of synthetic polymers has caused several negative repercussions on the health of humans and the environment. Recently, biopolymers have gained more attention among scientists of different disciplines by their potential therapeutic and commercial applications. Biopolymers are chain-like repeating units of molecules isolated from green sources. They are self-degradable, biocompatible, and non-toxic in nature. Recently, eco-friendly biopolymers such as extracellular polymeric substances (EPSs) have received much attention for their wide applications in the fields of emulsification, flocculation, preservatives, wastewater treatment, nanomaterial functionalization, drug delivery, cosmetics, glycomics, medicinal chemistry, and purification technology. The dynamicity of applications has raised the industrial and consumer demands to cater to the needs of mankind. This review deals with current insights and highlights on database surveys, potential sources, classification, extremophilic EPSs, bioprospecting, patents, microenvironment stability, biosynthesis, and genetic advances for production of high valued ecofriendly polymers. The importance of high valued EPSs in commercial and industrial applications in the global market economy is also summarized. This review concludes with future perspectives and commercial applications for the well-being of humanity.
Collapse
Affiliation(s)
- Saumi Pandey
- Department of Botany, MMV, Banaras Hindu University, Varanasi 221005, India
| | - Vinod K Kannaujiya
- Department of Botany, MMV, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
12
|
Leestemaker-Palmer A, Bermudez LE. Mycobacteroides abscessus ability to interact with the host mucosal cells plays an important role in pathogenesis of the infection. Crit Rev Microbiol 2024:1-13. [PMID: 39460453 DOI: 10.1080/1040841x.2024.2418130] [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: 06/20/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024]
Abstract
Non-tuberculous mycobacteria (NTM) are opportunistic pathogens ubiquitous in the environment. Mycobacteroides abscessus is a relatively new pathogen associated with underlying lung chronic pathologies, accounting for most of the pulmonary infections linked to the rapidly growing mycobacteria group. This includes chronic obstructive pulmonary disease, bronchiectasis, or cystic fibrosis. Patient outcomes from M. abscessus infections are poor due to complicated treatments and other factors. Intrinsic drug resistance plays an important role. The M. abscessus toolbox of resistance is varied leading to complex strategies for treatment. Mechanisms include waxy cell walls, drug export mechanisms, and acquired resistance. Many studies have also shown the impact of extracellular DNA found in the biofilm matrix during early infection and its possible advantage in pathogenicity. In this review, we discuss the current knowledge of early infection focusing on biofilm formation, an environmental strategy, and which treatments prevent its formation improving current antibiotic treatment outcomes in preliminary studies.
Collapse
Affiliation(s)
- Amy Leestemaker-Palmer
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Corvallis, OR, USA
| | - Luiz E Bermudez
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Corvallis, OR, USA
- Department of Microbiology, College of Sciences, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
13
|
Yang J, Mao L, Gulfam Y, Zeeshan M, Wang X, Fan T. Effect of Acetic Acid on Biofilm Formation in Paracidovorax citrulli, Causal Agent of Bacterial Fruit Blotch. J Basic Microbiol 2024:e2400188. [PMID: 39428671 DOI: 10.1002/jobm.202400188] [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: 05/06/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024]
Abstract
The unique tissue structure of pathogenic bacteria biofilm plays an important role in its pathogenicity and bactericide resistance. Inhibition or destruction of biofilm formation of pathogenic bacteria is of great significance for the control of plant bacterial diseases. In this study, Paracidovorax citrulli was inoculated into KB medium containing acetic acid, and after shaking at 28°C and 55 r/min for 48 h, it was found that the content of extracellular polysaccharide, extracellular protein and extracellular DNA (eDNA) decreased with the increase of acetic acid concentration, which resulted in the decrease of biofilm formation, it is not even possible to form biofilms on plastic slides. When the final concentration of acetic acid in the culture medium was greater than or equal to 0.5 mg/mL, there was no biofilm on the plastic slides. Therefore, the use of acetic acid as an inhibitor of P. citrulli has a good potential for control of bacterial fruit blotch.
Collapse
Affiliation(s)
- Jincheng Yang
- Key Laboratory for Oasis Agricultural Pest Management and Plant Resource Utilization at Universities of Xinjiang Uygur Autonomous Region, Shihezi, China
- College of Agronomy, Shihezi University, Shihezi, China
| | - Liang Mao
- Xinjiang Turpan Agricultural Technology Promotion Center, Turpan, China
| | - Yousaf Gulfam
- Key Laboratory for Oasis Agricultural Pest Management and Plant Resource Utilization at Universities of Xinjiang Uygur Autonomous Region, Shihezi, China
- College of Agronomy, Shihezi University, Shihezi, China
| | - Muhammad Zeeshan
- Key Laboratory for Oasis Agricultural Pest Management and Plant Resource Utilization at Universities of Xinjiang Uygur Autonomous Region, Shihezi, China
- College of Agronomy, Shihezi University, Shihezi, China
| | - Xiaodong Wang
- Key Laboratory for Oasis Agricultural Pest Management and Plant Resource Utilization at Universities of Xinjiang Uygur Autonomous Region, Shihezi, China
- College of Agronomy, Shihezi University, Shihezi, China
| | - Ting Fan
- College of Sciences, Shihezi University, Shihezi, Xinjiang, China
- Xinjiang Production & Construction Corps Key Laboratory of Advanced Energy Storage Materials and Technologies, Shihezi University, Shihezi, Xinjiang, China
| |
Collapse
|
14
|
Ragupathi H, Pushparaj MM, Gopi SM, Govindarajan DK, Kandaswamy K. Biofilm matrix: a multifaceted layer of biomolecules and a defensive barrier against antimicrobials. Arch Microbiol 2024; 206:432. [PMID: 39402397 DOI: 10.1007/s00203-024-04157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 11/10/2024]
Abstract
Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.
Collapse
Affiliation(s)
- Harini Ragupathi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Mahamahima Muthuswamy Pushparaj
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Sarves Mani Gopi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Drive, 637371, Singapore, Singapore
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India.
| |
Collapse
|
15
|
Yang R, Cui L, Xu S, Zhong Y, Xu T, Liu J, Lan Z, Qin S, Guo Y. Membrane-Targeting Amphiphilic Honokiol Derivatives Containing an Oxazole Moiety as Potential Antibacterials against Methicillin-Resistant Staphylococcus aureus. J Med Chem 2024; 67:16858-16872. [PMID: 39259708 DOI: 10.1021/acs.jmedchem.4c01860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Infections with methicillin-resistant Staphylococcus aureus (MRSA) are becoming increasingly serious, making the development of novel antimicrobials urgent. Here, we synthesized some amphiphilic honokiol derivatives bearing an oxazole moiety and investigated their antibacterial and hemolytic activities. Bioactivity evaluation showed that E17 possessed significant in vitro antibacterial activity against S. aureus and MRSA, along with low hemolytic activity. Moreover, E17 exhibited rapid bactericidal properties and was not susceptible to resistance. Mechanistic studies indicated that E17 interacts with phosphatidylglycerol and cardiolipin of bacterial cell membranes, leading to changes in cell membrane permeability and polarization, increased intracellular ROS, and leakage of DNA and proteins, thus accelerating bacterial death. Transcriptome analysis further demonstrated that E17 has membrane-targeting effects, affecting the expression of genes related to cell membranes and ABC transporter proteins. Notably, in vivo activity showed that E17 has prominent anti-MRSA efficacy, comparable to vancomycin, and is expected to be a new anti-MRSA drug candidate.
Collapse
Affiliation(s)
- Ruige Yang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Liping Cui
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Shengnan Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Yan Zhong
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Ting Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
| | - Jifeng Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Zhenwei Lan
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Shangshang Qin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Yong Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan Province 421001, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| |
Collapse
|
16
|
Ma X, Wu Z, Li J, Yang Y. Functional Study of desKR: a Lineage-Specific Two-Component System Positively Regulating Staphylococcus aureus Biofilm Formation. Infect Drug Resist 2024; 17:4037-4053. [PMID: 39309069 PMCID: PMC11416117 DOI: 10.2147/idr.s485049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Purpose Biofilms significantly contribute to the persistence and antibiotic resistance of Staphylococcus aureus infections. However, the regulatory mechanisms governing biofilm formation of S. aureus remain not fully elucidated. This study aimed to investigate the function of the S. aureus lineage-specific two-component system, desKR, in biofilm regulation and pathogenicity. Methods Bioinformatic analysis was conducted to assess the prevalence of desKR across various S. aureus lineages and to examine its structural features. The impact of desKR on S. aureus pathogenicity was evaluated using in vivo mouse models, including skin abscess, bloodstream infection, and nasal colonization models. Crystal violet staining and confocal laser scanning microscopy were utilized to examine the impact of desKR on S. aureus biofilm formation. Mechanistic insights into desKR-mediated biofilm regulation were investigated by quantifying polysaccharide intercellular adhesin (PIA) production, extracellular DNA (eDNA) release, autolysis assays, and RT-qPCR. Results The prevalence of desKR varied among different S. aureus lineages, with notably low carriage rates in ST398 and ST59 lineages. Deletion of desKR in NCTC8325 strain resulted in decreased susceptibility to β-lactam and glycopeptide antibiotics. Although desKR did not significantly affect acute pathogenicity, the ΔdesKR mutant exhibited significantly reduced nasal colonization and biofilm-forming ability. Overexpression of desKR in naturally desKR-lacking strains (ST398 and ST59) enhanced biofilm formation, suggesting a lineage-independent effect. Phenotypic assays further revealed that the ΔdesKR mutant showed reduced PIA production, decreased eDNA release, and lower autolysis rates. RT-qPCR indicated significant downregulation of icaA, icaD, icaB, and icaC genes, along with upregulation of icaR, whereas autolysis-related genes remained unchanged. Conclusion The desKR two-component system positively regulates S. aureus biofilm formation in a lineage-independent manner, primarily by modulating PIA synthesis via the ica operon. These findings provide new insights into the molecular mechanisms of biofilm formation in S. aureus and highlight desKR as a potential target for therapeutic strategies aimed at combating biofilm-associated infections.
Collapse
Affiliation(s)
- Xinyan Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| | - Ziyan Wu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| | - Junpeng Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| | - Yang Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, People’s Republic of China
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, and Joint Laboratory of International Cooperation on Prevention and Control Technology of Important Animal Diseases and Zoonoses of Jiangsu Higher Education Institutions, Yangzhou, 225009, People’s Republic of China
| |
Collapse
|
17
|
Tian LL, Li Y, Yang R, Jiang Y, He JJ, Wang H, Chen LQ, Zhu WY, Xue T, Li BB. Low concentrations of tetrabromobisphenol A promote the biofilm formation of methicillin-resistant Staphylococcus aureus. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116853. [PMID: 39137468 DOI: 10.1016/j.ecoenv.2024.116853] [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: 03/31/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
The effect and underlying mechanism of tetrabromobisphenol A (TBBPA), a plastic additive, on biofilm formation of methicillin-resistant Staphylococcus aureus (MRSA USA300) remain unknown. This study first investigated the impact of different concentrations of TBBPA on the growth and biofilm formation of USA300. The results indicated that a low concentration (0.5 mg/L) of TBBPA promoted the growth and biofilm formation of USA300, whereas high concentrations (5 mg/L and 10 mg/L) of TBBPA had inhibitory effects. Further exploration revealed that the low concentration of TBBPA enhance biofilm formation by promoting the synthesis of extracellular proteins, release of extracellular DNA (eDNA), and production of staphyloxanthin. RTqPCR analysis demonstrated that the low concentration of TBBPA upregulated genes associated with extracellular protein synthesis (sarA, fnbA, fnbB, aur) and eDNA formation (atlA) and increased the expression of genes involved in staphyloxanthin biosynthesis (crtM), suggesting a potential mechanism for enhanced resistance of USA300 to adverse conditions. These findings shed light on how low concentrations of TBBPA facilitate biofilm formation in USA300 and highlight the indirect impact of plastic additives on pathogenic bacteria in terms of human health. In the future, in-depth studies about effects of plastic additives on pathogenicity of pathogenic bacteria should be conducted. CAPSULE: The protein and eDNA contents in biofilms of methicillin-resistant Staphylococcus aureus are increased by low concentrations of TBBPA.
Collapse
Affiliation(s)
- Lin-Lin Tian
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yun Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Rui Yang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ying Jiang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Jiao-Jiao He
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Hui Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Li-Qi Chen
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Wen-Ya Zhu
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Ting Xue
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China; Food Procession Research Institute, Anhui Agricultural University, Hefei, Anhui 230036, China.
| | - Bing-Bing Li
- School of Life Sciences, Anhui Agricultural University, Hefei, Anhui 230036, China.
| |
Collapse
|
18
|
Rimal B, Chang JD, Liu C, Kim H, Aderotoye O, Zechmann B, Kim SJ. Scanning Electron Microscopy and Energy-Dispersive X-ray Spectroscopy of Staphylococcus aureus Biofilms. ACS OMEGA 2024; 9:37610-37620. [PMID: 39281927 PMCID: PMC11391442 DOI: 10.1021/acsomega.4c01168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 09/18/2024]
Abstract
Understanding the dynamics of biofilm formation and its elemental composition is crucial for developing effective strategies against biofilm-associated infections. In this study, we employed scanning electron microscopy (SEM) and energy-dispersive X-ray spectroscopy (EDS) to investigate the morphological changes and elemental compositions of Staphylococcus aureus biofilms. SEM images revealed distinct stages of biofilm development, from initial aggregation to the formation of mature and aged biofilms. EDS analysis consistently showed elevated levels of sodium (Na), oxygen (O), and phosphorus (P) in the biofilm matrix, indicating its high negative charge and the presence of anionic biopolymers. The incorporation of extracellular DNA (eDNA) into the biofilm matrix, leading to significant retention of sodium ions, underscored the importance of electrostatic interactions in biofilm formation and stability. Our findings highlight the potential of EDS analysis in quantifying elemental compositions and elucidating the role of anionic biopolymers in biofilm development.
Collapse
Affiliation(s)
- Binayak Rimal
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76798, United States
| | - James D Chang
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Chengyin Liu
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Haley Kim
- Department of Chemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Oluwatobi Aderotoye
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Bernd Zechmann
- Center for Microscopy and Imaging, Baylor University, Waco, Texas 76798, United States
| | - Sung Joon Kim
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| |
Collapse
|
19
|
Chen L, Wu MY, Chen SL, Hu R, Wang Y, Zeng W, Feng S, Ke M, Wang L, Chen S, Gu M. The Guardian of Vision: Intelligent Bacteriophage-Based Eyedrops for Clinical Multidrug-Resistant Ocular Surface Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407268. [PMID: 39091071 DOI: 10.1002/adma.202407268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Clinical multidrug-resistant Pseudomonas aeruginosa (MDR-PA) is the leading cause of refractory bacterial keratitis (BK). However, the reported BK treatment methods lack biosecurity and bioavailability, which usually causes irreversible visual impairment and even blindness. Herein, for BK caused by clinically isolated MDR-PA infection, armed phages are modularized with the type I photosensitizer (PS) ACR-DMT, and an intelligent phage eyedrop is developed for combined phagotherapy and photodynamic therapy (PDT). These eyedrops maximize the advantages of bacteriophages and ACR-DMT, enabling more robust and specific targeting killing of MDR-PA under low oxygen-dependence, penetrating and disrupting biofilms, and efficiently preventing biofilm reformation. Altering the biofilm and immune microenvironments alleviates inflammation noninvasively, promotes corneal healing without scar formation, protects ocular tissues, restores visual function, and prevents long-term discomfort and pain. This strategy exhibits strong scalability, enables at-home treatment of ocular surface infections with great patient compliance and a favorable prognosis, and has significant potential for clinical application.
Collapse
Affiliation(s)
- Luojia Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ming-Yu Wu
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Si-Ling Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Rui Hu
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yifei Wang
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Weijuan Zeng
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shun Feng
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Min Ke
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lianrong Wang
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, 518026, China
| | - Shi Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Shenzhen Institute of Translational Medicine, Shenzhen University Medical School, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meijia Gu
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
20
|
Kang S, Yang Y, Hou W, Zheng Y. Inhibitory Effects of Lactobionic Acid on Biofilm Formation and Virulence of Staphylococcus aureus. Foods 2024; 13:2781. [PMID: 39272546 PMCID: PMC11395522 DOI: 10.3390/foods13172781] [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: 06/26/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Staphylococcus aureus biofilm is a common bio-contaminant source that leads to food cross-contamination and foodborne disease outbreaks. Hence, there is a need for searching novel antibiofilm agents with potential anti-virulence properties to control S. aureus contamination and infections in food systems. In this study, the antibiofilm effects of lactobionic acid (LBA) against S. aureus and its influence on virulence were explored. The minimum inhibition concentration of LBA on S. aureus was 8 mg/mL. Viable count and crystal violet assays revealed that LBA inhibited and inactivated S. aureus biofilms. Microscopic observations further confirmed the antibiofilm activity of LBA on S. aureus that disrupted the biofilm architecture and inactivated the viable cells in biofilms. Moreover, LBA decreased the release of extracellular DNA (eDNA) and extracellular polysaccharide (EPS) in S. aureus biofilms. LBA suppressed biofilm formation by intervening metabolic activity and reduced virulence secretion by repressing the hemolytic activity of S. aureus. Furthermore, LBA altered the expressions of biofilm- and virulence-related genes in S. aureus, further confirming that LBA suppressed biofilm formation and reduced the virulence secretion of S. aureus. The results suggest that LBA might be useful in preventing and controlling biofilm formation and the virulence of S. aureus to ensure food safety.
Collapse
Affiliation(s)
- Shimo Kang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China
- CAS Engineering Laboratory for Nutrition, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yahui Yang
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China
| | - Wanwan Hou
- Department of Food Science & Technology, School of Agriculture and Biology, State Key Lab of Microbial Metabolism, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yan Zheng
- College of Food Science, Shenyang Agricultural University, Shenyang 110161, China
| |
Collapse
|
21
|
Nguyen A, Roy JJS, Kim JH, Yun KH, Lee W, Kim KK, Kim T, Chaurasia AK. Repeated Exposure of Vancomycin to Vancomycin-Susceptible Staphylococcus aureus (VSSA) Parent Emerged VISA and VRSA Strains with Enhanced Virulence Potentials. J Microbiol 2024; 62:535-553. [PMID: 38814539 DOI: 10.1007/s12275-024-00139-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/31/2024]
Abstract
The emergence of resistance against the last-resort antibiotic vancomycin in staphylococcal infections is a serious concern for human health. Although various drug-resistant pathogens of diverse genetic backgrounds show higher virulence potential, the underlying mechanism behind this is not yet clear due to variability in their genetic dispositions. In this study, we investigated the correlation between resistance and virulence in adaptively evolved isogenic strains. The vancomycin-susceptible Staphylococcus aureus USA300 was exposed to various concentrations of vancomycin repeatedly as a mimic of the clinical regimen to obtain mutation(s)-accrued-clonally-selected (MACS) strains. The phenotypic analyses followed by expression of the representative genes responsible for virulence and resistance of MACS strains were investigated. MACS strains obtained under 2 and 8 µg/ml vancomycin, named Van2 and Van8, respectively; showed enhanced vancomycin minimal inhibitory concentrations (MIC) to 4 and 16 µg/ml, respectively. The cell adhesion and invasion of MACS strains increased in proportion to their MICs. The correlation between resistance and virulence potential was partially explained by the differential expression of genes known to be involved in both virulence and resistance in MACS strains compared to parent S. aureus USA300. Repeated treatment of vancomycin against vancomycin-susceptible S. aureus (VSSA) leads to the emergence of vancomycin-resistant strains with variable levels of enhanced virulence potentials.
Collapse
Affiliation(s)
- An Nguyen
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - J Jean Sophy Roy
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Ji-Hoon Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyung-Hee Yun
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Truc Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| | - Akhilesh Kumar Chaurasia
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Antimicrobial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| |
Collapse
|
22
|
Wei PW, Wang X, Wang C, Chen M, Liu MZ, Liu WX, He YL, Xu GB, Zheng XH, Zhang H, Liu HM, Wang B. Ginkgo biloba L. exocarp petroleum ether extract inhibits methicillin-resistant Staphylococcus aureus by modulating ion transport, virulence, and biofilm formation in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:117957. [PMID: 38493904 DOI: 10.1016/j.jep.2024.117957] [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: 01/04/2024] [Revised: 02/10/2024] [Accepted: 02/19/2024] [Indexed: 03/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As reported in the Ancient Chinese Medicinal Books, Ginkgo biloba L. fruit has been used as a traditional Chinese medicine for the treatment asthma and cough or as a disinfectant. Our previous study demonstrated that G. biloba exocarp extract (GBEE), an extract of a traditional Chinese herb, inhibits the formation of methicillin-resistant Staphylococcus aureus (MRSA) biofilms. However, GBEE is a crude extract that contains many components, and the underlying mechanisms of purified GBEE fractions extracted with solvents of different polarities are unknown. AIM OF THE STUDY This study aimed to investigate the different components in GBEE fractions extracted with solvents of different polarities and their antibacterial effects and mechanisms against MRSA and Staphylococcus haemolyticus biofilms both in vitro and in vivo. METHODS The components in different fractions were detected by high-performance liquid chromatography-high resolution mass spectrometry (HPLC-HRMS). Microbroth dilution assays and time growth curves were used to determine the antibacterial effects of the fractions on 15 clinical bacterial isolates. Crystal violet staining, scanning electron microscopy (SEM) and transmission electron microscopy (TEM) were utilized to identify the fractions that affected bacterial biofilm formation. The potential MRSA targets of the GBEE fraction obtained with petroleum ether (PE), denoted GBEE-PE, were screened by transcriptome sequencing, and the gene expression profile was verified by quantitative polymerase chain reaction (qPCR). RESULTS HPLC-HRMS analysis revealed that the four GBEE fractions (extracted with petroleum ether, ethyl acetate, n-butanol, and water) contained different ginkgo components, and the antibacterial effects decreased as the polarity of the extraction solvent increased. The antibacterial activity of GBEE-PE was greater than that of the GBEE fraction extracted with ethyl acetate (EA). GBEE-PE improved H. illucens survival and reduced MRSA colonization in model mouse organs. Crystal violet staining and SEM and TEM analyses revealed that GBEE-PE inhibited MRSA and S. haemolyticus biofilm formation. Transcriptional analysis revealed that GBEE-PE inhibits MRSA biofilms by altering ion transport, cell wall metabolism and virulence-related gene expression. In addition, the LO2 cell viability and H. illucens toxicity assay data showed that GBEE-PE at 20 mg/kg was nontoxic. CONCLUSION The GBEE fractions contained different components, and their antibacterial effects decreased with increases in the polarity of the extraction solvent. GBEE-PE limited MRSA growth and biofilm formation by affecting ion transport, cell wall synthesis, and virulence-related pathways. This research provides a more detailed overview of the mechanism by which GBEE-PE inhibits MRSA both in vitro and in vivo and suggests that GBEE-PE is a new prospective antimicrobial with the potential to be used in MRSA therapeutics in the future.
Collapse
Affiliation(s)
- Peng-Wei Wei
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Xu Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Cong Wang
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Formulation (R&D) Department, Guiyang, 550001, China
| | - Ming Chen
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Meng-Zhu Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China
| | - Wen-Xia Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China
| | - Yan-Ling He
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Guo-Bo Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants & School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, Guizhou, China.
| | - Xiao-He Zheng
- Zhejiang Hisun Pharmaceutical Co., Ltd., Taizhou, 318000, Zhejiang, China
| | - Hua Zhang
- Department of Laboratory Medicine, Guizhou Provincial People's Hospital, Affiliated Hospital of Guizhou University, Guiyang, 550002, Guizhou, China.
| | - Hong-Mei Liu
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| | - Bing Wang
- Engineering Research Center of Health Medicine Biotechnology of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering (Modern Industry College of Health Medicine), Guizhou Medical University, Guiyang, 561113, Guizhou, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, China Ministry of Education (School of Public Health, Guizhou Medical University), Guiyang, 561113, Guizhou, China; Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, School of Basic Medical Science, Guizhou Medical University, Guiyang, 561113, Guizhou, China.
| |
Collapse
|
23
|
Yu G, Xi H, Sheng T, Lin J, Luo Z, Xu J. Sub-inhibitory concentrations of tetrabromobisphenol A induce the biofilm formation of methicillin-resistant Staphylococcus aureus. Arch Microbiol 2024; 206:301. [PMID: 38874781 DOI: 10.1007/s00203-024-04022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024]
Abstract
Biofilm formation by methicillin-resistant Staphylococcus aureus (MRSA) on indwelling medical devices complicates the treatment of infection. Tetrabromobisphenol A (TBBPA), a synthetic, lipophilic, halogenated aromatic compound widely used as an additive in plastics and electronic products, has raised environmental concerns due to its potential for bioaccumulation. This study investigated the impact of sub-inhibitory concentrations of TBBPA on MRSA biofilm formation. Crystal violet staining and confocal laser scanning microscopy analysis demonstrated that 1/8 MIC (0.5 µg/mL) of TBBPA significantly stimulated MRSA biofilm formation (P < 0.0001). MTT assays indicated that the metabolic activity within the biofilms increased by 15.60-40.85% compared to untreated controls. Dot blot immunoassay, autolysis assay, and extracellular DNA (eDNA) quantification further revealed TBBPA enhanced the production of polysaccharide intercellular adhesin (PIA) and eDNA, which are key biofilm components. Additionally, TBBPA was found to enhance the production of staphyloxanthin, facilitating MRSA survival under oxidative conditions and in human whole blood. RT-qPCR analysis showed that TBBPA significantly upregulated genes associated with biofilm formation (icaA, atlA, sarA), staphyloxanthin biosynthesis (crtM and sigB), and oxidative stress responses (sodA and katA). These findings suggest that TBBPA promotes MRSA biofilm development and enhances bacterial resistance to adverse conditions, thereby potentially exacerbating risks to human health.
Collapse
Affiliation(s)
- Guofang Yu
- Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Huimin Xi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Tianle Sheng
- Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Jin Lin
- Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Zhaoxia Luo
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| | - Jianqing Xu
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
24
|
Bowden LC, Finlinson J, Jones B, Berges BK. Beyond the double helix: the multifaceted landscape of extracellular DNA in Staphylococcus aureus biofilms. Front Cell Infect Microbiol 2024; 14:1400648. [PMID: 38903938 PMCID: PMC11188362 DOI: 10.3389/fcimb.2024.1400648] [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: 03/18/2024] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
Staphylococcus aureus forms biofilms consisting of cells embedded in a matrix made of proteins, polysaccharides, lipids, and extracellular DNA (eDNA). Biofilm-associated infections are difficult to treat and can promote antibiotic resistance, resulting in negative healthcare outcomes. eDNA within the matrix contributes to the stability, growth, and immune-evasive properties of S. aureus biofilms. eDNA is released by autolysis, which is mediated by murein hydrolases that access the cell wall via membrane pores formed by holin-like proteins. The eDNA content of S. aureus biofilms varies among individual strains and is influenced by environmental conditions, including the presence of antibiotics. eDNA plays an important role in biofilm development and structure by acting as an electrostatic net that facilitates protein-cell and cell-cell interactions. Because of eDNA's structural importance in biofilms and its ubiquitous presence among S. aureus isolates, it is a potential target for therapeutics. Treatment of biofilms with DNase can eradicate or drastically reduce them in size. Additionally, antibodies that target DNABII proteins, which bind to and stabilize eDNA, can also disperse biofilms. This review discusses the recent literature on the release, structure, and function of eDNA in S. aureus biofilms, in addition to a discussion of potential avenues for targeting eDNA for biofilm eradication.
Collapse
Affiliation(s)
| | | | | | - Bradford K. Berges
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| |
Collapse
|
25
|
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
|
26
|
Li J, Lu T, Chu Y, Zhang Y, Zhang J, Fu W, Sun J, Liu Y, Liao X, Zhou Y. Cinnamaldehyde targets SarA to enhance β-lactam antibiotic activity against methicillin-resistant Staphylococcus aureus. MLIFE 2024; 3:291-306. [PMID: 38948140 PMCID: PMC11211666 DOI: 10.1002/mlf2.12121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 07/02/2024]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a current global public health problem due to its increasing resistance to the most recent antibiotic therapies. One critical approach is to develop ways to revitalize existing antibiotics. Here, we show that the phytogenic compound cinnamaldehyde (CIN) and β-lactam antibiotic combinations can functionally synergize and resensitize clinical MRSA isolates to β-lactam therapy and inhibit MRSA biofilm formation. Mechanistic studies indicated that the CIN potentiation effect on β-lactams was primarily the result of inhibition of the mecA expression by targeting the staphylococcal accessory regulator sarA. CIN alone or in combination with β-lactams decreased sarA gene expression and increased SarA protein phosphorylation that impaired SarA binding to the mecA promoter element and downregulated virulence genes such as those encoding biofilm, α-hemolysin, and adhesin. Perturbation of SarA-mecA binding thus interfered with PBP2a biosynthesis and this decreased MRSA resistance to β-lactams. Furthermore, CIN fully restored the anti-MRSA activities of β-lactam antibiotics in vivo in murine models of bacteremia and biofilm infections. Together, our results indicated that CIN acts as a β-lactam adjuvant and can be applied as an alternative therapy to combat multidrug-resistant MRSA infections.
Collapse
Affiliation(s)
- Jianguo Li
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Tingyin Lu
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Yuefei Chu
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Yuejun Zhang
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
- Yantai Fushan Center for Animal Disease Control and PreventionYantaiChina
| | - Wenzhen Fu
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Yahong Liu
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Xiao‐Ping Liao
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| | - Yu‐Feng Zhou
- State Key Laboratory for Animal Disease Control and PreventionSouth China Agricultural UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety EvaluationSouth China Agricultural UniversityGuangzhouChina
| |
Collapse
|
27
|
Wu X, Wang H, Xiong J, Yang GX, Hu JF, Zhu Q, Chen Z. Staphylococcus aureus biofilm: Formulation, regulatory, and emerging natural products-derived therapeutics. Biofilm 2024; 7:100175. [PMID: 38298832 PMCID: PMC10827693 DOI: 10.1016/j.bioflm.2023.100175] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 02/02/2024] Open
Abstract
Staphylococcus aureus can readily form biofilm which enhances the drug-resistance, resulting in life-threatening infections involving different organs. Biofilm formation occurs due to a series of developmental events including bacterial adhesion, aggregation, biofilm maturation, and dispersion, which are controlled by multiple regulatory systems. Rapidly increasing research and development outcomes on natural products targeting S. aureus biofilm formation and/or regulation led to an emergent application of active phytochemicals and combinations. This review aimed at providing an in-depth understanding of biofilm formation and regulation mechanisms for S. aureus, outlining the most important antibiofilm strategies and potential targets of natural products, and summarizing the latest progress in combating S. aureus biofilm with plant-derived natural products. These findings provided further evidence for novel antibiofilm drugs research and clinical therapies.
Collapse
Affiliation(s)
- Xiying Wu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
- Institute of Natural Medicine and Health Products, School of Pharmaceutical Sciences, Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Taizhou University, Zhejiang, 318000, China
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Huan Wang
- School of Pharmacy, Naval Medical University, Shanghai, 200433, China
| | - Juan Xiong
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Guo-Xun Yang
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Jin-Feng Hu
- Institute of Natural Medicine and Health Products, School of Pharmaceutical Sciences, Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Taizhou University, Zhejiang, 318000, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200443, China
| |
Collapse
|
28
|
Maree M, Ushijima Y, Fernandes PB, Higashide M, Morikawa K. SCC mec transformation requires living donor cells in mixed biofilms. Biofilm 2024; 7:100184. [PMID: 38440091 PMCID: PMC10909703 DOI: 10.1016/j.bioflm.2024.100184] [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: 09/15/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an important human pathogen that has emerged through the horizontal acquisition of the staphylococcal cassette chromosome mec (SCCmec). Previously, we showed that SCCmec from heat-killed donors can be transferred via natural transformation in biofilms at frequencies of 10-8-10-7. Here, we show an improved transformation assay of SCCmec with frequencies up to 10-2 using co-cultured biofilms with living donor cells. The Ccr-attB system played an important role in SCCmec transfer, and the deletion of ccrAB recombinase genes reduced the frequency ∼30-fold. SCCmec could be transferred from either MRSA or methicillin-resistant coagulase-negative staphylococci to some methicillin-sensitive S. aureus recipients. In addition, the transformation of other plasmid or chromosomal genes is enhanced by using living donor cells. This study emphasizes the role of natural transformation as an evolutionary ability of S. aureus and in MRSA emergence.
Collapse
Affiliation(s)
- Mais Maree
- Institute of Medicine, University of Tsukuba, Japan
| | | | | | - Masato Higashide
- Kotobiken Medical Laboratories, Inc., Kamiyokoba, Tsukuba, Japan
| | | |
Collapse
|
29
|
Schwartbeck B, Rumpf CH, Hait RJ, Janssen T, Deiwick S, Schwierzeck V, Mellmann A, Kahl BC. Various mutations in icaR, the repressor of the icaADBC locus, occur in mucoid Staphylococcus aureus isolates recovered from the airways of people with cystic fibrosis. Microbes Infect 2024; 26:105306. [PMID: 38316375 DOI: 10.1016/j.micinf.2024.105306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/07/2024]
Abstract
Staphylococcus aureus is one of the major pathogens isolated from the airways of people with cystic fibrosis (pwCF). Recently, we described a mucoid S. aureus phenotype from respiratory specimens of pwCF, which constitutively overproduced biofilm that consisted of polysaccharide intercellular adhesin (PIA) due to a 5bp-deletion (5bp-del) in the intergenic region of the intercellular adhesin (ica) locus. Since we were not able to identify the 5bp-del in mucoid isolates of two pwCF with long-term S. aureus persistence and in a number of mucoid isolates of pwCF from a prospective multicenter study, these strains were (i) characterized phenotypically, (ii) investigated for biofilm formation, and (iii) molecular typed by spa-sequence typing. To screen for mutations responsible for mucoidy, the ica operon of all mucoid isolates was analyzed by Sanger sequencing. Whole genome sequencing was performed for selected isolates. For all mucoid isolates without the 5 bp-del, various mutations in icaR, which is the transcriptional repressor of the icaADBC operon. Mucoid and non-mucoid strains belonged to the same spa-type. Transformation of PIA-overproducing S. aureus with a vector expressing the intact icaR gene restored the non-mucoid phenotype. Altogether, we demonstrated a new mechanism for the emergence of mucoid S. aureus isolates of pwCF.
Collapse
Affiliation(s)
- Bianca Schwartbeck
- Institute of Medical Microbiology, University Hospital Muenster, Germany
| | - Christine H Rumpf
- Institute of Medical Microbiology, University Hospital Muenster, Germany
| | | | - Timo Janssen
- Institute of Medical Microbiology, University Hospital Muenster, Germany
| | - Susanne Deiwick
- Institute of Medical Microbiology, University Hospital Muenster, Germany
| | | | | | - Barbara C Kahl
- Institute of Medical Microbiology, University Hospital Muenster, Germany.
| |
Collapse
|
30
|
Panthi VK, Fairfull-Smith KE, Islam N. Liposomal drug delivery strategies to eradicate bacterial biofilms: Challenges, recent advances, and future perspectives. Int J Pharm 2024; 655:124046. [PMID: 38554739 DOI: 10.1016/j.ijpharm.2024.124046] [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: 12/23/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Typical antibiotic treatments are often ineffectual against biofilm-related infections since bacteria residing within biofilms have developed various mechanisms to resist antibiotics. To overcome these limitations, antimicrobial-loaded liposomal nanoparticles are a promising anti-biofilm strategy as they have demonstrated improved antibiotic delivery and eradication of bacteria residing in biofilms. Antibiotic-loaded liposomal nanoparticles revealed remarkably higher antibacterial and anti-biofilm activities than free drugs in experimental settings. Moreover, liposomal nanoparticles can be used efficaciously for the combinational delivery of antibiotics and other antimicrobial compounds/peptide which facilitate, for instance, significant breakdown of the biofilm matrix, increased bacterial elimination from biofilms and depletion of metabolic activity of various pathogens. Drug-loaded liposomes have mitigated recurrent infections and are considered a promising tool to address challenges associated to antibiotic resistance. Furthermore, it has been demonstrated that surface charge and polyethylene glycol modification of liposomes have a notable impact on their antibacterial biofilm activity. Future investigations should tackle the persistent hurdles associated with development of safe and effective liposomes for clinical application and investigate novel antibacterial treatments, including CRISPR-Cas gene editing, natural compounds, phages, and nano-mediated approaches. Herein, we emphasize the significance of liposomes in inhibition and eradication of various bacterial biofilms, their challenges, recent advances, and future perspectives.
Collapse
Affiliation(s)
- Vijay Kumar Panthi
- Pharmacy Discipline, School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Kathryn E Fairfull-Smith
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, Queensland 4001, Australia; Centre for Materials Science, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Nazrul Islam
- Pharmacy Discipline, School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia; Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Centre for Immunology and Infection Control (CIIC), Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| |
Collapse
|
31
|
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
|
32
|
Yousuf B, Flint A, Weedmark K, Pagotto F, Ramirez-Arcos S. Comparative virulome analysis of four Staphylococcus epidermidis strains from human skin and platelet concentrates using whole genome sequencing. Access Microbiol 2024; 6:000780.v3. [PMID: 38737800 PMCID: PMC11083402 DOI: 10.1099/acmi.0.000780.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 02/29/2024] [Indexed: 05/14/2024] Open
Abstract
Staphylococcus epidermidis is one of the predominant bacterial contaminants in platelet concentrates (PCs), a blood component used to treat bleeding disorders. PCs are a unique niche that triggers biofilm formation, the main pathomechanism of S. epidermidis infections. We performed whole genome sequencing of four S. epidermidis strains isolated from skin of healthy human volunteers (AZ22 and AZ39) and contaminated PCs (ST10002 and ST11003) to unravel phylogenetic relationships and decipher virulence mechanisms compared to 24 complete S. epidermidis genomes in GenBank. AZ39 and ST11003 formed a separate unique lineage with strains 14.1 .R1 and SE95, while AZ22 formed a cluster with 1457 and ST10002 closely grouped with FDAAGOS_161. The four isolates were assigned to sequence types ST1175, ST1174, ST73 and ST16, respectively. All four genomes exhibited biofilm-associated genes ebh, ebp, sdrG, sdrH and atl. Additionally, AZ22 had sdrF and aap, whereas ST10002 had aap and icaABCDR. Notably, AZ39 possesses truncated ebh and sdrG and harbours a toxin-encoding gene. All isolates carry multiple antibiotic resistance genes conferring resistance to fosfomycin (fosB), β-lactams (blaZ) and fluoroquinolones (norA). This study reveales a unique lineage for S. epidermidis and provides insight into the genetic basis of virulence and antibiotic resistance in transfusion-associated S. epidermidis strains.
Collapse
Affiliation(s)
- Basit Yousuf
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Annika Flint
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Canada
| | - Kelly Weedmark
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Canada
| | - Franco Pagotto
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
- Bureau of Microbial Hazards, Health Products and Food Branch, Health Canada, Ottawa, Canada
| | - Sandra Ramirez-Arcos
- Medical Affairs and Innovation, Canadian Blood Services, Ottawa, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
33
|
Aboelnaga N, Elsayed SW, Abdelsalam NA, Salem S, Saif NA, Elsayed M, Ayman S, Nasr M, Elhadidy M. Deciphering the dynamics of methicillin-resistant Staphylococcus aureus biofilm formation: from molecular signaling to nanotherapeutic advances. Cell Commun Signal 2024; 22:188. [PMID: 38519959 PMCID: PMC10958940 DOI: 10.1186/s12964-024-01511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/01/2024] [Indexed: 03/25/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) represents a global threat, necessitating the development of effective solutions to combat this emerging superbug. In response to selective pressures within healthcare, community, and livestock settings, MRSA has evolved increased biofilm formation as a multifaceted virulence and defensive mechanism, enabling the bacterium to thrive in harsh conditions. This review discusses the molecular mechanisms contributing to biofilm formation across its developmental stages, hence representing a step forward in developing promising strategies for impeding or eradicating biofilms. During staphylococcal biofilm development, cell wall-anchored proteins attach bacterial cells to biotic or abiotic surfaces; extracellular polymeric substances build scaffolds for biofilm formation; the cidABC operon controls cell lysis within the biofilm, and proteases facilitate dispersal. Beside the three main sequential stages of biofilm formation (attachment, maturation, and dispersal), this review unveils two unique developmental stages in the biofilm formation process for MRSA; multiplication and exodus. We also highlighted the quorum sensing as a cell-to-cell communication process, allowing distant bacterial cells to adapt to the conditions surrounding the bacterial biofilm. In S. aureus, the quorum sensing process is mediated by autoinducing peptides (AIPs) as signaling molecules, with the accessory gene regulator system playing a pivotal role in orchestrating the production of AIPs and various virulence factors. Several quorum inhibitors showed promising anti-virulence and antibiofilm effects that vary in type and function according to the targeted molecule. Disrupting the biofilm architecture and eradicating sessile bacterial cells are crucial steps to prevent colonization on other surfaces or organs. In this context, nanoparticles emerge as efficient carriers for delivering antimicrobial and antibiofilm agents throughout the biofilm architecture. Although metal-based nanoparticles have been previously used in combatting biofilms, its non-degradability and toxicity within the human body presents a real challenge. Therefore, organic nanoparticles in conjunction with quorum inhibitors have been proposed as a promising strategy against biofilms. As nanotherapeutics continue to gain recognition as an antibiofilm strategy, the development of more antibiofilm nanotherapeutics could offer a promising solution to combat biofilm-mediated resistance.
Collapse
Affiliation(s)
- Nirmeen Aboelnaga
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Salma W Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology & Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nehal Adel Abdelsalam
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Salma Salem
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Nehal A Saif
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Manar Elsayed
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Shehab Ayman
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - Maha Nasr
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Elhadidy
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, Egypt.
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt.
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
34
|
Shi L, Lin W, Cai Y, Chen F, Zhang Q, Liang D, Xiu Y, Lin S, He B. Oxidative Stress-Mediated Repression of Virulence Gene Transcription and Biofilm Formation as Antibacterial Action of Cinnamomum burmannii Essential Oil on Staphylococcus aureus. Int J Mol Sci 2024; 25:3078. [PMID: 38474323 DOI: 10.3390/ijms25053078] [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: 01/17/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
This work aimed to identify the chemical compounds of Cinnamomum burmannii leaf essential oil (CBLEO) and to unravel the antibacterial mechanism of CBLEO at the molecular level for developing antimicrobials. CBLEO had 37 volatile compounds with abundant borneol (28.40%) and showed good potential to control foodborne pathogens, of which Staphylococcus aureus had the greatest inhibition zone diameter (28.72 mm) with the lowest values of minimum inhibitory concentration (1.0 μg/mL) and bactericidal concentration (2.0 μg/mL). To unravel the antibacterial action of CBLEO on S. aureus, a dynamic exploration of antibacterial growth, material leakage, ROS formation, protein oxidation, cell morphology, and interaction with genome DNA was conducted on S. aureus exposed to CBLEO at different doses (1/2-2×MIC) and times (0-24 h), indicating that CBLEO acts as an inducer for ROS production and the oxidative stress of S. aureus. To highlight the antibacterial action of CBLEO on S. aureus at the molecular level, we performed a comparative association of ROS accumulation with some key virulence-related gene (sigB/agrA/sarA/icaA/cidA/rsbU) transcription, protease production, and biofilm formation in S. aureus subjected to CBLEO at different levels and times, revealing that CBLEO-induced oxidative stress caused transcript suppression of virulence regulators (RsbU and SigB) and its targeted genes, causing a protease level increase destined for the biofilm formation and growth inhibition of S. aureus, which may be a key bactericidal action. Our findings provide valuable information for studying the antibacterial mechanism of essential oil against pathogens.
Collapse
Affiliation(s)
- Lingling Shi
- College of Biological Sciences and Biotechnology, National Engineering Laboratory for Tree Breeding, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, Ministry of Education, Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, Beijing Forestry University, Beijing 100083, China
| | - Wei Lin
- College of Biological Sciences and Biotechnology, National Engineering Laboratory for Tree Breeding, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, Ministry of Education, Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, Beijing Forestry University, Beijing 100083, China
| | - Yanling Cai
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou 510520, China
| | - Feng Chen
- College of Biological Sciences and Biotechnology, National Engineering Laboratory for Tree Breeding, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, Ministry of Education, Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, Beijing Forestry University, Beijing 100083, China
| | - Qian Zhang
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou 510520, China
| | - Dongcheng Liang
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou 510520, China
| | - Yu Xiu
- College of Biological Sciences and Biotechnology, National Engineering Laboratory for Tree Breeding, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, Ministry of Education, Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, Beijing Forestry University, Beijing 100083, China
| | - Shanzhi Lin
- College of Biological Sciences and Biotechnology, National Engineering Laboratory for Tree Breeding, Key Laboratory of Genetics and Breeding in Forest Trees and Ornamental Plants, Ministry of Education, Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, Beijing Forestry University, Beijing 100083, China
| | - Boxiang He
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou 510520, China
| |
Collapse
|
35
|
Bakalakos M, Ampadiotaki MM, Vlachos C, Sipsas N, Pneumaticos S, Vlamis J. Molecular Mechanisms of Biofilm Formation on Orthopaedic Implants: Review of the Literature. MAEDICA 2024; 19:129-136. [PMID: 38736937 PMCID: PMC11079743 DOI: 10.26574/maedica.2021.19.1.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Orthopaedic implant-associated infections (OIAIs) is one of the most catastrophic complications following joint arthroplasty or fracture fixation. Given the increasing number of orthopaedic implants which are used annually, periprosthetic infections emerge as a global problem. Their diagnosis and consequent therapeutic management remain challenging for clinicians. Biofilm formation is a complex and only partially understood process that has not been extensively studied. Understanding the underlying mechanisms involved in biofilm formation is crucial in the amelioration of both diagnosis and therapeutic management of OIAIs. We performed a literature review of the molecular mechanisms of biofilm formation and discussed the four most common and thoroughly researched microbes of biofilm-related OIAIs.
Collapse
Affiliation(s)
- Matthaios Bakalakos
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| | | | - Christos Vlachos
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| | - Nikolaos Sipsas
- Infectious Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Spiros Pneumaticos
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| | - John Vlamis
- 3rd Orthopaedic Department, National and Kapodistrian University of Athens, KAT General Hospital, Athens, Greece
| |
Collapse
|
36
|
Røder HL, Christidi E, Amador CI, Music S, Olesen AK, Svensson B, Madsen JS, Herschend J, Kreft JU, Burmølle M. Flagellar interference with plasmid uptake in biofilms: a joint experimental and modeling study. Appl Environ Microbiol 2024; 90:e0151023. [PMID: 38095456 PMCID: PMC10807428 DOI: 10.1128/aem.01510-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/30/2023] [Indexed: 01/25/2024] Open
Abstract
Plasmid conjugation is a key facilitator of horizontal gene transfer (HGT), and plasmids encoding antibiotic resistance drive the increasing prevalence of antibiotic resistance. In natural, engineered, and clinical environments, bacteria often grow in protective biofilms. Therefore, a better understanding of plasmid transfer in biofilms is needed. Our aim was to investigate plasmid transfer in a biofilm-adapted wrinkly colony mutant of Xanthomonas retroflexus (XRw) with enhanced matrix production and reduced motility. We found that XRw biofilms had an increased uptake of the broad host-range IncP-1ϵ plasmid pKJK5 compared to the wild type (WT). Proteomics revealed fewer flagellar-associated proteins in XRw, suggesting that flagella were responsible for reducing plasmid uptake. This was confirmed by the higher plasmid uptake of non-flagellated fliM mutants of the X. retroflexus wrinkly mutant as well as the wild type. Moreover, testing several flagellar mutants of Pseudomonas putida suggested that the flagellar effect was more general. We identified seven mechanisms with the potential to explain the flagellar effect and simulated them in an individual-based model. Two mechanisms could thus be eliminated (increased distances between cells and increased lag times due to flagella). Another mechanism identified as viable in the modeling was eliminated by further experiments. The possibility of steric hindrance of pilus movement and binding by flagella, reducing the frequency of contact and thus plasmid uptake, proved viable, and the three other viable mechanisms had a reduced probability of plasmid transfer in common. Our findings highlight the important yet complex effects of flagella during bacterial conjugation in biofilms.IMPORTANCEBiofilms are the dominant form of microbial life and bacteria living in biofilms are markedly different from their planktonic counterparts, yet the impact of the biofilm lifestyle on horizontal gene transfer (HGT) is still poorly understood. Horizontal gene transfer by conjugative plasmids is a major driver in bacterial evolution and adaptation, as exemplified by the troubling spread of antibiotic resistance. To either limit or promote plasmid prevalence and dissemination, we need a better understanding of plasmid transfer between bacterial cells, especially in biofilms. Here, we identified a new factor impacting the transfer of plasmids, flagella, which are required for many types of bacterial motility. We show that their absence or altered activity can lead to enhanced plasmid uptake in two bacterial species, Xanthomonas retroflexus and Pseudomonas putida. Moreover, we demonstrate the utility of mathematical modeling to eliminate hypothetical mechanisms.
Collapse
Affiliation(s)
- Henriette Lyng Røder
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Eleni Christidi
- School of Biosciences & Institute of Microbiology and Infection & Centre for Computational Biology, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | | | - Samra Music
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Birte Svensson
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Jakob Herschend
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jan-Ulrich Kreft
- School of Biosciences & Institute of Microbiology and Infection & Centre for Computational Biology, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | - Mette Burmølle
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Campbell MJ, Beenken KE, Spencer HJ, Jayana B, Hester H, Sahukhal GS, Elasri MO, Smeltzer MS. Comparative evaluation of small molecules reported to be inhibitors of Staphylococcus aureus biofilm formation. Microbiol Spectr 2024; 12:e0314723. [PMID: 38059629 PMCID: PMC10782960 DOI: 10.1128/spectrum.03147-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023] Open
Abstract
IMPORTANCE Because biofilm formation is such a problematic feature of Staphylococcus aureus infections, much effort has been put into identifying biofilm inhibitors. However, the results observed with these compounds are often reported in isolation, and the methods used to assess biofilm formation vary between labs, making it impossible to assess relative efficacy and prioritize among these putative inhibitors for further study. The studies we report address this issue by directly comparing putative biofilm inhibitors using a consistent in vitro assay. This assay was previously shown to maximize biofilm formation, and the results observed with this assay have been proven to be relevant in vivo. Of the 19 compounds compared using this method, many had no impact on biofilm formation under these conditions. Indeed, only one proved effective at limiting biofilm formation without also inhibiting growth.
Collapse
Affiliation(s)
- Mara J. Campbell
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Karen E. Beenken
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Horace J. Spencer
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Bina Jayana
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Hana Hester
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gyan S. Sahukhal
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Mohamed O. Elasri
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Mark S. Smeltzer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
38
|
Jordão CC, Klein MI, Barbugli PA, Mima EGDO, de Sousa TV, Ferrisse TM, Pavarina AC. DNase improves the efficacy of antimicrobial photodynamic therapy in the treatment of candidiasis induced with Candida albicans. Front Microbiol 2023; 14:1274201. [PMID: 38188579 PMCID: PMC10766804 DOI: 10.3389/fmicb.2023.1274201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
The study evaluated the association of DNase I enzyme with antimicrobial photodynamic therapy (aPDT) in the treatment of oral candidiasis in mice infected with fluconazole-susceptible (CaS) and -resistant (CaR) Candida albicans strains. Mice were inoculated with C. albicans, and after the infection had been established, the tongues were exposed to DNase for 5 min, followed by photosensitizer [Photodithazine®(PDZ)] and light (LED), either singly or combined. The treatments were performed for 5 consecutive days. Treatment efficacy was evaluated by assessing the tongues via fungal viable population, clinical evaluation, histopathological and fluorescence microscopy methods immediately after finishing treatments, and 7 days of follow-up. The combination of DNase with PDZ-aPDT reduced the fungal viability in mice tongues immediately after the treatments by around 4.26 and 2.89 log10 for CaS and CaR, respectively (versus animals only inoculated). In the fluorescence microscopy, the polysaccharides produced by C. albicans and fungal cells were less labeled in animals treated with the combination of DNase with PDZ-aPDT, similar to the healthy animals. After 7 days of the treatment, DNase associated with PDZ-aPDT maintained a lower count, but not as pronounced as immediately after the intervention. For both strains, mice treated with the combination of DNase with PDZ-aPDT showed remission of oral lesions and mild inflammatory infiltrate in both periods assessed, while animals treated only with PDZ-aPDT presented partial remission of oral lesions. DNase I enzyme improved the efficacy of photodynamic treatment.
Collapse
Affiliation(s)
- Cláudia Carolina Jordão
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Marlise Inêz Klein
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Paula Aboud Barbugli
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Ewerton Garcia de Oliveira Mima
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Tábata Viana de Sousa
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Túlio Morandin Ferrisse
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Ana Claudia Pavarina
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
39
|
Luo ZX, Li Y, Liu MF, Zhao R. Ciprofloxacin enhances the biofilm formation of Staphylococcus aureus via an agrC-dependent mechanism. Front Microbiol 2023; 14:1328947. [PMID: 38179460 PMCID: PMC10764545 DOI: 10.3389/fmicb.2023.1328947] [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: 10/27/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024] Open
Abstract
Staphylococcus aureus readily forms biofilms on host tissues and medical devices, enabling its persistence in chronic infections and resistance to antibiotic therapy. The accessory gene regulator (Agr) quorum sensing system plays a key role in regulating S. aureus biofilm formation. This study reveals the widely used fluoroquinolone antibiotic, ciprofloxacin, strongly stimulates biofilm formation in methicillin-resistant S. aureus, methicillin-sensitive S. aureus, and clinical isolates with diverse genetic backgrounds. Crystal violet staining indicated that ciprofloxacin induced a remarkable 12.46- to 15.19-fold increase in biofilm biomass. Confocal laser scanning microscopy revealed that ciprofloxacin induced denser biofilms. Phenotypic assays suggest that ciprofloxacin may enhance polysaccharide intercellular adhesin production, inhibit autolysis, and reduce proteolysis during the biofilm development, thus promoting initial adhesion and enhancing biofilm stability. Mechanistically, ciprofloxacin significantly alters the expression of various biofilm-related genes (icaA, icaD, fnbA, fnbB, eap, emp) and regulators (agrA, saeR). Gene knockout experiments revealed that deletion of agrC, rather than saeRS, abolishes the ciprofloxacin-induced enhancement of biofilm formation, underscoring the key role of agrC. Thermal shift assays showed ciprofloxacin binds purified AgrC protein, thereby inhibiting the Agr system. Molecular docking results further support the potential interaction between ciprofloxacin and AgrC. In summary, subinhibitory concentrations of ciprofloxacin stimulate S. aureus biofilm formation via an agrC-dependent pathway. This inductive effect may facilitate local infection establishment and bacterial persistence, ultimately leading to therapeutic failure.
Collapse
Affiliation(s)
- Zhao-xia Luo
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Yuting Li
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, China
| | - Mei-fang Liu
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Zhao
- Department of Clinical Laboratory, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
40
|
Liu J, Huang T, Xu Z, Mao Y, Soteyome T, Liu G, Qu C, Yuan L, Ma Q, Zhou F, Seneviratne G. Sub-MIC streptomycin and tetracycline enhanced Staphylococcus aureus Guangzhou-SAU749 biofilm formation, an in-depth study on transcriptomics. Biofilm 2023; 6:100156. [PMID: 37779859 PMCID: PMC10539642 DOI: 10.1016/j.bioflm.2023.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023] Open
Abstract
Staphylococcus aureus is a major human pathogen, a potential "Super-bug" and a typical biofilm forming bacteria. With usage of large amount of antibiotics, the residual antibiotics in clinical settings further complicate the colonization, pathogenesis and resistance of S. aureus. This study aimed at investigating the phenotypical and global gene expression changes on biofilm formation of a clinical S. aureus isolate treated under different types of antibiotics. Firstly, an isolate Guangzhou-SAU749 was selected from a large sale of previously identified S. aureus isolates, which exhibited weak biofilm formation in terms of biomass and viability. Secondly, 9 commonly prescribed antibiotics for S. aureus infections treatment, together with 10 concentrations ranging from 1/128 to 4 minimum inhibitory concentration (MIC) with 2-fold serial dilution, were used as different antibiotic stress conditions. Then, biofilm formation of S. aureus Guangzhou-SAU749 at different stages including 8 h, 16 h, 24 h, and 48 h, was tested by crystal violet and MTS assays. Thirdly, the whole genome of S. aureus Guangzhou-SAU749 was investigated by genome sequencing on PacBio platform. Fourthly, since enhancement of biofilm formation occurred when treated with 1/2 MIC tetracycline (TCY) and 1/4 MIC streptomycin (STR) since 5 h, the relevant biofilm samples were selected and subjected to RNA-seq and bioinformatics analysis. Last, expression of two component system (TCS) and biofilm associated genes in 4 h, 8 h, 16 h, 24 h, and 48 h sub-MIC TCY and STR treated biofilm samples were performed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Although most antibiotics lowered the biomass and cell viability of Guangzhou-SAU749 biofilm at concentrations higher than MIC, certain antibiotics including TCY and STR promoted biofilm formation at sub-MICs. Additionally, upon genome sequencing, RNA-seq and RT-qPCR on biofilm samples treated with sub-MIC of TCY and STR at key time points, genes lytR, arlR, hssR, tagA, clfB, atlA and cidA related to TCS and biofilm formation were identified to contribute to the enhanced biofilm formation, providing a theoretical basis for further controlling on S. aureus biofilm formation.
Collapse
Affiliation(s)
- Junyan Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Tengyi Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhenbo Xu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuzhu Mao
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, South China University of Technology, Guangzhou, 510640, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Gongliang Liu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Chunyun Qu
- College of Light Industry and Food Science, Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, 510225, China
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, 225127, PR China
| | - Qin Ma
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture /Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, 510610, China
| | - Fang Zhou
- The First Affiliated Hospital, Sun Yan-Sen University, Guangzhou, 510080, China
| | - Gamini Seneviratne
- National Institute of Fundamental Studies, Hantana road, Kandy, Sri Lanka
| |
Collapse
|
41
|
Heredia-Ponce Z, Secchi E, Toyofuku M, Marinova G, Savorana G, Eberl L. Genotoxic stress stimulates eDNA release via explosive cell lysis and thereby promotes streamer formation of Burkholderia cenocepacia H111 cultured in a microfluidic device. NPJ Biofilms Microbiomes 2023; 9:96. [PMID: 38071361 PMCID: PMC10710452 DOI: 10.1038/s41522-023-00464-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
DNA is a component of biofilms, but the triggers of DNA release during biofilm formation and how DNA contributes to biofilm development are poorly investigated. One key mechanism involved in DNA release is explosive cell lysis, which is a consequence of prophage induction. In this article, the role of explosive cell lysis in biofilm formation was investigated in the opportunistic human pathogen Burkholderia cenocepacia H111 (H111). Biofilm streamers, flow-suspended biofilm filaments, were used as a biofilm model in this study, as DNA is an essential component of their matrix. H111 contains three prophages on chromosome 1 of its genome, and the involvement of each prophage in causing explosive cell lysis of the host and subsequent DNA and membrane vesicle (MV) release, as well as their contribution to streamer formation, were studied in the presence and absence of genotoxic stress. The results show that two of the three prophages of H111 encode functional lytic prophages that can be induced by genotoxic stress and their activation causes DNA and MVs release by explosive cell lysis. Furthermore, it is shown that the released DNA enables the strain to develop biofilm streamers, and streamer formation can be enhanced by genotoxic stress. Overall, this study demonstrates the involvement of prophages in streamer formation and uncovers an often-overlooked problem with the use of antibiotics that trigger the bacterial SOS response for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Zaira Heredia-Ponce
- Department of Plant and Microbial Biology, University of Zürich, 8008, Zürich, Switzerland
| | - Eleonora Secchi
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH Zürich, 8093, Zürich, Switzerland
| | - Masanori Toyofuku
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, Japan
| | - Gabriela Marinova
- Department of Plant and Microbial Biology, University of Zürich, 8008, Zürich, Switzerland
| | - Giovanni Savorana
- Institute of Environmental Engineering, Department of Civil, Environmental and Geomatic Engineering, ETH Zürich, 8093, Zürich, Switzerland
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zürich, 8008, Zürich, Switzerland.
| |
Collapse
|
42
|
Li JG, Chen XF, Lu TY, Zhang J, Dai SH, Sun J, Liu YH, Liao XP, Zhou YF. Increased Activity of β-Lactam Antibiotics in Combination with Carvacrol against MRSA Bacteremia and Catheter-Associated Biofilm Infections. ACS Infect Dis 2023; 9:2482-2493. [PMID: 38019707 DOI: 10.1021/acsinfecdis.3c00338] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
β-Lactam antibiotics are the mainstay for the treatment of staphylococcal infections, but their utility is greatly limited by the emergence and rapid dissemination of methicillin-resistant Staphylococcus aureus (MRSA). Herein, we evaluated the ability of the plant-derived monoterpene carvacrol to act as an antibiotic adjuvant, revitalizing the anti-MRSA activity of β-lactam antibiotics. Increased susceptibility of MRSA to β-lactam antibiotics and significant synergistic activities were observed with carvacrol-based combinations. Carvacrol significantly inhibited MRSA biofilms and reduced the production of exopolysaccharide, polysaccharide intercellular adhesin, and extracellular DNA and showed synergistic biofilm inhibition in combination with β-lactams. Transcriptome analysis revealed profound downregulation in the expression of genes involved in two-component systems and S. aureus infection. Mechanistic studies indicate that carvacrol inhibits the expression of staphylococcal accessory regulator sarA and interferes with SarA-mecA promoter binding that decreases mecA-mediated β-lactam resistance. Consistently, the in vivo experiment also supported that carvacrol restored MRSA sensitivity to β-lactam antibiotic treatments in both murine models of bacteremia and biofilm-associated infection. Our results indicated that carvacrol has a potential role as a combinatorial partner with β-lactam antibiotics to address MRSA infections.
Collapse
Affiliation(s)
- Jian-Guo Li
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Feng Chen
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Ting-Yin Lu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Yantai Fushan Center for Animal Disease Control and Prevention, Fushan, Yantai, Shandong 265500, China
| | - Shu-He Dai
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Ya-Hong Liu
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiao-Ping Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| | - Yu-Feng Zhou
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
43
|
Hernández-Cuellar E, Tsuchiya K, Valle-Ríos R, Medina-Contreras O. Differences in Biofilm Formation by Methicillin-Resistant and Methicillin-Susceptible Staphylococcus aureus Strains. Diseases 2023; 11:160. [PMID: 37987271 PMCID: PMC10660471 DOI: 10.3390/diseases11040160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a common pathogen involved in community- and hospital-acquired infections. Its biofilm formation ability predisposes it to device-related infections. Methicillin-resistant S. aureus (MRSA) strains are associated with more serious infections and higher mortality rates and are more complex in terms of antibiotic resistance. It is still controversial whether MRSA are indeed more virulent than methicillin-susceptible S. aureus (MSSA) strains. A difference in biofilm formation by both types of bacteria has been suggested, but how only the presence of the SCCmec cassette or mecA influences this phenotype remains unclear. In this review, we have searched for literature studying the difference in biofilm formation by MRSA and MSSA. We highlighted the relevance of the icaADBC operon in the PIA-dependent biofilms generated by MSSA under osmotic stress conditions, and the role of extracellular DNA and surface proteins in the PIA-independent biofilms generated by MRSA. We described the prominent role of surface proteins with the LPXTG motif and hydrolases for the release of extracellular DNA in the MRSA biofilm formation. Finally, we explained the main regulatory systems in S. aureus involved in virulence and biofilm formation, such as the SarA and Agr systems. As most of the studies were in vitro using inert surfaces, it will be necessary in the future to focus on biofilm formation on extracellular matrix components and its relevance in the pathogenesis of infection by both types of strains using in vivo animal models.
Collapse
Affiliation(s)
- Eduardo Hernández-Cuellar
- Laboratorio de Biología Celular y Tisular, Departamento de Morfología, Universidad Autónoma de Aguascalientes, Aguascalientes 20100, C.P., México
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan;
| | - Ricardo Valle-Ríos
- Research Division, Faculty of Medicine, Universidad Nacional Autónoma de México (UNAM), Mexico City 04360, C.P., México;
- Laboratory of Research in Immunology and Proteomics, Federico Gómez Children’s Hospital of Mexico, Mexico City 06720, C.P., México
| | - Oscar Medina-Contreras
- Epidemiology, Endocrinology & Nutrition Research Unit, Mexico Children’s Hospital (HIMFG), Mexico City 06720, C.P., México;
| |
Collapse
|
44
|
Wang B, Zhan Q, Xiao Y, Xu Y, Zhao H, Rao L, Wang X, Zhang J, Shen L, Zhou Y, Guo Y, Wu X, Yu J, Yu F. Mupirocin enhances the biofilm formation of Staphylococcus epidermidis in an atlE-dependent manner. Int J Antimicrob Agents 2023; 62:106904. [PMID: 37385560 DOI: 10.1016/j.ijantimicag.2023.106904] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/27/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023]
Abstract
The pathogenicity of Staphylococcus epidermidis is largely attributed to its exceptional ability to form biofilms. Here, we report that mupirocin, an antimicrobial agent widely used for staphylococcal decolonization and anti-infection, strongly stimulates the biofilm formation of S. epidermidis. Although the polysaccharide intercellular adhesin (PIA) production was unaffected, mupirocin significantly facilitated extracellular DNA (eDNA) release by accelerating autolysis, thereby positively triggering cell surface attachment and intercellular agglomeration during biofilm development. Mechanistically, mupirocin regulated the expression of genes encoding for the autolysin AtlE as well as the programmed cell death system CidA-LrgAB. Critically, through gene knockout, we found out that deletion of atlE, but not cidA or lrgA, abolished the enhancement of biofilm formation and eDNA release in response to mupirocin treatment, indicating that atlE is required for this effect. In Triton X-100 induced autolysis assay, mupirocin treated atlE mutant displayed a slower autolysis rate compared with the wild-type strain and complementary strain. Therefore, we concluded that subinhibitory concentrations of mupirocin enhance the biofilm formation of S. epidermidis in an atlE dependent manner. This induction effect could conceivably be responsible for some of the more unfavourable outcomes of infectious diseases.
Collapse
Affiliation(s)
- Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qing Zhan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Infection Control Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanghua Xiao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanlei Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huilin Zhao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lulin Rao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyi Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ying Zhou
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaocui Wu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jingyi Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
45
|
Peng Q, Tang X, Dong W, Zhi Z, Zhong T, Lin S, Ye J, Qian X, Chen F, Yuan W. Carvacrol inhibits bacterial polysaccharide intracellular adhesin synthesis and biofilm formation of mucoid Staphylococcus aureus: an in vitro and in vivo study. RSC Adv 2023; 13:28743-28752. [PMID: 37807974 PMCID: PMC10552078 DOI: 10.1039/d3ra02711b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is one of the important human pathogens and causes both superficial and systemic infections. More importantly, the formation of S. aureus biofilms, a main cause of its pathogenicity and drug resistance, has been a critical challenge in clinical treatment. Carvacrol, a plant-based natural product, has gained great interest for therapeutic purposes due to its effective biological activity with low cytotoxicity. The present study aimed to investigate the effect of carvacrol on anti-biofilm activity. Growth curve analysis showed that applying a sub-inhibitory concentration of carvacrol (4 μg mL-1) was not lethal to S. aureus SYN; however, the inhibition rate of biofilm formation was as high as 63.6%, and the clearance rate of mature biofilms was as high as 30.7%. In addition, carvacrol effectively reduced the production of biofilm-associated extracellular polysaccharides and showed no effect on eDNA release. Furthermore, qPCR analysis revealed that carvacrol significantly down-regulated the expression of icaA, icaB, icaC, agrA, and sarA (P < 0.05). The in vivo efficacy of carvacrol against biofilm infection was further verified with a biological model of G. mellonella larvae. The results showed that carvacrol was non-toxic to the larvae and can effectively increase the survival rate of the larvae infected with S. aureus strain SYN.
Collapse
Affiliation(s)
- Qi Peng
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| | - Xiaohua Tang
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University Guangzhou 510150 PR China
| | - Wanyang Dong
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| | - Ziling Zhi
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| | - Tian Zhong
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| | - Shunan Lin
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| | - Jingyi Ye
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| | - Xiping Qian
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| | - Fu Chen
- Panyu District Health Management Center Guangzhou 511450 PR China
| | - Wenchang Yuan
- Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University Guangzhou 510180 PR China
| |
Collapse
|
46
|
Gadar K, McCarthy RR. Using next generation antimicrobials to target the mechanisms of infection. NPJ ANTIMICROBIALS AND RESISTANCE 2023; 1:11. [PMID: 38686217 PMCID: PMC11057201 DOI: 10.1038/s44259-023-00011-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/28/2023] [Indexed: 05/02/2024]
Abstract
The remarkable impact of antibiotics on human health is being eroded at an alarming rate by the emergence of multidrug resistant pathogens. There is a recognised consensus that new strategies to tackle infection are urgently needed to limit the devasting impact of antibiotic resistance on our global healthcare infrastructure. Next generation antimicrobials (NGAs) are compounds that target bacterial virulence factors to disrupt pathogenic potential without impacting bacterial viability. By disabling the key virulence factors required to establish and maintain infection, NGAs make pathogens more vulnerable to clearance by the immune system and can potentially render them more susceptible to traditional antibiotics. In this review, we discuss the developing field of NGAs and how advancements in this area could offer a viable standalone alternative to traditional antibiotics or an effective means to prolong antibiotic efficacy when used in combination.
Collapse
Affiliation(s)
- Kavita Gadar
- Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH United Kingdom
| | - Ronan R. McCarthy
- Division of Biosciences, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH United Kingdom
| |
Collapse
|
47
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
48
|
Das A, Patro S, Simnani FZ, Singh D, Sinha A, Kumari K, Rao PV, Singh S, Kaushik NK, Panda PK, Suar M, Verma SK. Biofilm modifiers: The disparity in paradigm of oral biofilm ecosystem. Biomed Pharmacother 2023; 164:114966. [PMID: 37269809 DOI: 10.1016/j.biopha.2023.114966] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 05/29/2023] [Indexed: 06/05/2023] Open
Abstract
A biofilm is a population of sessile microorganisms that has a distinct organized structure and characteristics like channels and projections. Good oral hygiene and reduction in the prevalence of periodontal diseases arise from minimal biofilm accumulation in the mouth, however, studies focusing on modifying the ecology of oral biofilms have not yet been consistently effective. The self-produced matrix of extracellular polymeric substances and greater antibiotic resistance make it difficult to target and eliminate biofilm infections, which lead to serious clinical consequences that are often lethal. Therefore, a better understanding is required to target and modify the ecology of biofilms in order to eradicate the infection, not only in instances of oral disorders but also in terms of nosocomial infections. The review focuses on several biofilm ecology modifiers to prevent biofilm infections, as well as the involvement of biofilm in antibiotic resistance, implants or in-dwelling device contamination, dental caries, and other periodontal disorders. It also discusses recent advances in nanotechnology that may lead to novel strategies for preventing and treating infections caused by biofilms as well as a novel outlook to infection control.
Collapse
Affiliation(s)
- Antarikshya Das
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Swadheena Patro
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India.
| | | | - Dibyangshee Singh
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Adrija Sinha
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Khushbu Kumari
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Patnala Vedika Rao
- KIIT School of Medical Sciences, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Sarita Singh
- BVG Life Sciences Limited, Sagar Complex, Old Pune-Mumbai Road, Chinchwad, Pune 411034, India
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, Republic of Korea.
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India.
| | - Suresh K Verma
- KIIT School of Dental Sciences, KIIT University, Bhubaneswar 751024, Odisha, India.
| |
Collapse
|
49
|
Van Roy Z, Shi W, Kak G, Duan B, Kielian T. Epigenetic Regulation of Leukocyte Inflammatory Mediator Production Dictates Staphylococcus aureus Craniotomy Infection Outcome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:414-428. [PMID: 37314520 PMCID: PMC10524781 DOI: 10.4049/jimmunol.2300050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/18/2023] [Indexed: 06/15/2023]
Abstract
Staphylococcus aureus is a common cause of surgical-site infections, including those arising after craniotomy, which is performed to access the brain for the treatment of tumors, epilepsy, or hemorrhage. Craniotomy infection is characterized by complex spatial and temporal dynamics of leukocyte recruitment and microglial activation. We recently identified unique transcriptional profiles of these immune populations during S. aureus craniotomy infection. Epigenetic processes allow rapid and reversible control over gene transcription; however, little is known about how epigenetic pathways influence immunity to live S. aureus. An epigenetic compound library screen identified bromodomain and extraterminal domain-containing (BET) proteins and histone deacetylases (HDACs) as critical for regulating TNF, IL-6, IL-10, and CCL2 production by primary mouse microglia, macrophages, neutrophils, and granulocytic myeloid-derived suppressor cells in response to live S. aureus. Class I HDACs (c1HDACs) were increased in these cell types in vitro and in vivo during acute disease in a mouse model of S. aureus craniotomy infection. However, substantial reductions in c1HDACs were observed during chronic infection, highlighting temporal regulation and the importance of the tissue microenvironment for dictating c1HDAC expression. Microparticle delivery of HDAC and BET inhibitors in vivo caused widespread decreases in inflammatory mediator production, which significantly increased bacterial burden in the brain, galea, and bone flap. These findings identify histone acetylation as an important mechanism for regulating cytokine and chemokine production across diverse immune cell lineages that is critical for bacterial containment. Accordingly, aberrant epigenetic regulation may be important for promoting S. aureus persistence during craniotomy infection.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198
| | - Gunjan Kak
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
50
|
Zhang A, Wu H, Chen X, Chen Z, Pan Y, Qu W, Hao H, Chen D, Xie S. Targeting and arginine-driven synergizing photodynamic therapy with nutritional immunotherapy nanosystems for combating MRSA biofilms. SCIENCE ADVANCES 2023; 9:eadg9116. [PMID: 37450586 PMCID: PMC10348676 DOI: 10.1126/sciadv.adg9116] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023]
Abstract
The resistance and immune escape of methicillin-resistant Staphylococcus aureus (MRSA) biofilms cause recalcitrant infections. Here, we design a targeting and synergizing cascade PDT with nutritional immunotherapy nanosystems (Arg-PCN@Gel) containing PCN-224 as PDT platform for providing reactive oxygen species (ROS), incorporating arginine (Arg) as nitric oxide (NO) donor to cascade with ROS to produce more lethal ONOO- and promote immune response, and coating with gelatin as targeting agent and persistent Arg provider. The nanosystems adhered to the autolysin of MRSA and inhibited Arg metabolism by down-regulating icdA and icaA. It suppressed polysaccharide intercellular adhesin and extracellular DNA synthesis to prevent biofilm formation. The NO broke mature biofilms and helped ROS and ONOO- penetrate into biofilms to inactivate internal MRSA. Arg-PCN@Gel drove Arg to enhance immunity via inducible NO synthase/NO axis and arginase/polyamine axis and achieve efficient target treatment in MRSA biofilm infections. The targeting and cascading PDT synergized with nutritional immunotherapy provide an effective promising strategy for biofilm-associated infections.
Collapse
Affiliation(s)
- Aoxue Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Hao Wu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Xin Chen
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhen Chen
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Yuanhu Pan
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Wei Qu
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Haihong Hao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dongmei Chen
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| | - Shuyu Xie
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
- Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs,Wuhan, Hubei 430070, China
| |
Collapse
|