1
|
Habib MB, Batool G, Shah NA, Muhammad T, Akbar NS, Shahid A. Biofilm-mediated infections; novel therapeutic approaches and harnessing artificial intelligence for early detection and treatment of biofilm-associated infections. Microb Pathog 2025; 203:107497. [PMID: 40118297 DOI: 10.1016/j.micpath.2025.107497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 12/04/2024] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
A biofilm is a group of bacteria that have self-produced a matrix and are grouped together in a dense population. By resisting the host's immune system's phagocytosis process and attacking with anti-microbial chemicals such as reactive oxygen and nitrogen species, a biofilm enables pathogenic bacteria to evade elimination. One of the major problems in managing chronic injuries is treating wounds colonized by biofilms. These days, a major issue is the biofilms, which exacerbate infection pathogenesis and severity. Numerous investigators have already discovered cutting-edge methods for biofilm manipulation. Using phytochemicals is a practical tactic to control and prevent the production of biofilms. Numerous studies conducted in the last few years have demonstrated the antibacterial and antibiofilm qualities of nanoparticles (NPs) against bacteria, fungi, and protozoa. Because hydrogel has antibiofilm properties, it has been employed extensively in wound care recently. It may be removed with ease and without causing trauma. Today, artificial intelligence (AI) is being used to improve these tactics by providing customized treatment alternatives and predictive analytics. Artificial intelligence (AI) algorithms have the capability to examine extensive datasets and detect trends in biofilm formation and resistance mechanisms. This can aid in the creation of more potent antimicrobial drugs. AI models analyze complex datasets, predict biofilm formation, and guide the design of personalized treatment strategies by identifying resistance mechanisms and therapeutic targets with exceptional precision. This review provides an integrative perspective on biofilm formation mechanisms and their role in infections, highlighting the innovative applications of AI in this domain. By integrating data from diverse biological systems, AI accelerates drug discovery, optimizes treatment regimens, and enables real-time monitoring of biofilm dynamics. From predictive analytics to personalized care, we explore how AI enhances biofilm diagnostics and introduces precision medicine in biofilm-associated infections. This approach not only addresses the limitations of traditional methods but also paves the way for revolutionary advancements in infection control, antimicrobial resistance management, and improved patient outcomes.
Collapse
Affiliation(s)
| | - Ghanwa Batool
- Department of Computer Science, Comsats University Islamabad, Abbottabad, 22060, Pakistan
| | - Naseer Ali Shah
- Department of Biosciences, COMSATS University, Islamabad, 44000, Pakistan
| | - Taseer Muhammad
- Department of Mathematics, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Noreen Sher Akbar
- Department of Mechanical Engineering, College of Engineering, Prince Mohammad Bin Fahd University, Al Khobar, 31952, Saudi Arabia
| | - Ameera Shahid
- National Institute of Health, Islamabad, 44000, Pakistan
| |
Collapse
|
2
|
Bodine SP, Muir TW. Molecular Mechanisms of Virulence Regulation in Staphylococcus aureus: A Journey into Reconstitutive Biochemistry. Acc Chem Res 2025. [PMID: 40331756 DOI: 10.1021/acs.accounts.5c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
ConspectusMethodological development in the fields of genetics, chemical biology, and biochemistry over the last several decades has provided researchers with a diverse set of powerful tools to investigate biological processes. Leveraging these innovations in concert, scientists can now characterize biological pathways at a level of complexity ranging from systems biology down to molecular and atomic detail.Throughout this Account, we illustrate how discoveries made using these tools build on each other to develop a comprehensive understanding of biological pathways. Advancements in genetic sequencing facilitates association of genotypes and phenotypes, independent of biochemical mechanism. Through the biochemical reconstitution of the interactions between biological macromolecules─including the small molecules (ligands and metabolites) and proteins─that participate in these biological pathways, scientists can characterize the specific molecular features that link genotype and phenotype. This facilitates identification of targets within these pathways that can be manipulated to achieve a greater understanding of the biological process or to develop interventions to improve human health outcomes.Specifically, we describe how this toolbox was leveraged to discover and characterize the molecular biochemistry underlying control of pathogenicity in the Gram-positive bacterium Staphylococcus aureus. Concurrent with advancements in the investigative tools available to the scientific community, we and others reported on the genetic, molecular, and biochemical/biophysical components of this regulatory system. Virulence control in S. aureus is achieved through a chemical system of bacterial cell-to-cell communication indexed to local population density, referred to as quorum sensing (QS). We and our collaborators identified that this QS system is encoded in the accessory gene regulator (agr) operon and functions via the biosynthesis, secretion, and accumulation of a short peptide signaling molecule─the autoinducing peptide (AIP)─in the local environment correlated with the growth of S. aureus in the same biological niche. Above a threshold concentration, these AIPs bind and activate a cell-surface receptor to stimulate an intracellular response resulting in altered gene expression and bacterial group behaviors. We discovered that chemical modification of these AIPs often generates molecules that exhibit potent inhibition of agr QS, with demonstrated therapeutic potential to treat S. aureus infections. We went on to characterize the biochemical mechanism of signaling molecule biosynthesis and receptor activation in controlled systems through in vitro reconstitution of the constituent enzymes and substrates. Biochemical reconstitution enabled quantitative assessment of biophysical parameters. These efforts culminated in the comprehensive characterization and functional in vitro reconstitution of agr QS in a synthetic system in a minimal model at the interface of genotype, mechanism, and phenotype.
Collapse
Affiliation(s)
- Steven P Bodine
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Tom W Muir
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
3
|
Li S, Fan Z, Zheng K, Wu Y, Zhong G, Xu X. Engineered Probiotics with Low Oxygen Targeting Porphyromonas gingivalis and Gingival Fibroblasts for the Treatment of Periodontitis. ACS Biomater Sci Eng 2025. [PMID: 40286317 DOI: 10.1021/acsbiomaterials.5c00111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2025]
Abstract
The overuse of antibiotics has increased the prevalence of drug-resistant bacteria in periodontitis. "Sentinel" gingival fibroblasts, stimulated by pathogenic bacteria, continue to release signaling factors that affect stem cell repair and recruit immune cells, resulting in persistent inflammation in periodontal tissues, eventually leading to the loosening and loss of teeth. Periodontal pathogenic bacteria cause surface hypoxia, and gingival fibroblasts in the inflammatory microenvironment express HIF-1α, promoting hypoxic areas in periodontal pockets. No drug delivery system is available for the hypoxic region of periodontal pockets. We synthesized BI NPs via berberine (BBR) and indocyanine green (ICG) and formed BIP NPs by wrapping BI NPs with polydopamine (PDA), and the BIP NPs were delivered to the hypoxic region of the periodontal pocket by hitchhiking with the anaerobic probiotic Bifidobacterium bifidum (Bif). The BIP NPs released berberin (BBR) under near-infrared (NIR) irradiation, which inhibited the sulfur metabolism of Porphyromonas gingivalis via mild photothermal action and BBR-targeted serine acetyltransferase, resulting in a decrease in resistance to oxidative stress, thus exerting a nonantibiotic bacteriostatic effect. This mild photothermal effect facilitated the uptake of BIP NPs bygingival fibroblasts. Moreover, BBR targeted nuclear factor-erythroid 2-related factor 2 (NRF2) to reduce ferroptosis, and the gingival fibroblast supernatant modulated macrophage polarization through the NF-κB pathway. In the periodontitis rat model, Bif@BIP+NIR treatment carried the drug to deep periodontal pockets, decreasing local gingival ferroptosis and alleviating periodontitis symptoms. To summarize, engineered probiotics target low-oxygen periodontal pockets for drug delivery, P. gingivalis for nonantibiotic bacterial inhibition, and gingival fibroblasts to mitigate ferroptosis, thus alleviating periodontitis to reduce periodontitis.
Collapse
Affiliation(s)
- Shenghong Li
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhibo Fan
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Kaijun Zheng
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yujie Wu
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Guannan Zhong
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiaomei Xu
- Department of Orthodontics, The Affiliated Stomatology Hospital, Luzhou, Sichuan 646000, P.R. China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, The Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- Institute of Stomatology, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
4
|
Pham DT, Thuy NTN, Thao NTP, Nhi LT, Thuy BTP. Naturally derived hydrogels for wound healing. Ther Deliv 2025; 16:349-363. [PMID: 39871586 PMCID: PMC11970767 DOI: 10.1080/20415990.2025.2457928] [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: 10/06/2024] [Accepted: 01/21/2025] [Indexed: 01/29/2025] Open
Abstract
Natural hydrogels have garnered increasing attention due to their natural origins and beneficial roles in wound healing. Hydrogel water-retaining capacity and excellent biocompatibility create an ideal moist environment for wound healing, thereby enhancing cell proliferation and tissue regeneration. For this reason, naturally derived hydrogels formulated from biomaterials such as chitosan, alginate, gelatin, and fibroin are highly promising due to their biodegradability and low immunogenic responses. Recent integrated approaches to utilizing new technologies with bioactive agents have significantly improved the mechanical properties of hydrogels and the controlled release and delivery of active compounds, thereby increasing the efficiency of the treatment processes. Herein, this review highlights the advantages and the challenges of natural hydrogels in wound healing, focusing on their mechanical strength, controlled degradation rates, safety and efficiency validation, and the potential for incorporating advanced technologies such as tissue engineering and gene therapy for utilization in personalized medicine.
Collapse
Affiliation(s)
- Duy Toan Pham
- Department of Health Sciences, College of Natural Sciences, Can Tho University, Can Tho, Vietnam
| | - Ngo Thi Ngoc Thuy
- Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Nguyen Thi Phuong Thao
- Institute of Tropical Biology, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Le Thi Nhi
- Faculty of Materials Science, University of Science, Vietnam National University, Ho Chi Minh City, Vietnam
| | - Bui Thi Phuong Thuy
- Faculty of Fundamental Sciences, Van Lang University, Ho Chi Minh City, Vietnam
| |
Collapse
|
5
|
Xiang C, Pu C, Zhong X, Wang Y, Song W, Wang X, Chen K, Li K, Luo Y, Jiang K, Jiang D. Functional hydrogels promote chronic infectious wound healing by re-rousing macrophage M1 and inducing bacterial copper-like death. Mater Today Bio 2025; 31:101571. [PMID: 40051527 PMCID: PMC11883446 DOI: 10.1016/j.mtbio.2025.101571] [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: 12/03/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 03/09/2025] Open
Abstract
Traditional antibiotics are often ineffective against biofilm-associated infections, and biofilm-induced macrophage immune evasion directly halts the wound healing process. Disrupting biofilms and regulating macrophage immune functions are critical to improving wound healing. In this study, we synthesized g-C3N4 with peroxidase (POD) enzyme activity via thermal polymerization and copper alginate microspheres (CAM) via gas cutting. These were co-encapsulated into GelMA hydrogels to form a functionalized wound repair system (GelMA/CAM@g-C3N4) with both anti-biofilm and local immune microenvironment remodeling capabilities. In vitro, this system exhibited excellent biocompatibility and promoted endothelial cell migration, vascular formation, and CD31 expression. It also polarized macrophages toward the M1 phenotype, restoring their pro-inflammatory functions, upregulating inflammatory cytokines (IL-1, IL-6, TNF-α), and inhibiting Staphylococcus aureus and Escherichia coli. In vivo, the system suppressed S. aureus growth, promoted angiogenesis and collagen deposition, and reshaped the pathological microenvironment to achieve wound repair and regeneration. Conclusions: This system offers a new therapeutic strategy for chronic infectious wounds.
Collapse
Affiliation(s)
- Chao Xiang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, 401120, Chongqing, China
- Department of Orthopedics, The Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Chaoyu Pu
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, 401120, Chongqing, China
- Department of Orthopedics, The Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - XueMei Zhong
- School of Clinical Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, 401331, China
| | - Yong Wang
- Department of Orthopedics, The Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Weiyong Song
- Department of Orthopedics, The Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Xingkuan Wang
- Department of Orthopedics, The Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Kemiao Chen
- Chongqing Medical University, 401120, Chongqing, China
| | - Kai Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, 401120, Chongqing, China
| | - Yue Luo
- Department of Orthopedics, The Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Ke Jiang
- Department of Orthopedics, The Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Dianming Jiang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, 401120, Chongqing, China
| |
Collapse
|
6
|
Li S, Ji Y, Xue X, Yang Y, Huang Y, Yang S, Chen X. Hierarchical biofilm models using sodium alginate beads containing bacteria embedded in a cellulose-chitosan hydrogel matrix. J Mater Chem B 2025; 13:3952-3958. [PMID: 40025994 DOI: 10.1039/d4tb02015d] [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: 03/04/2025]
Abstract
In biofilm studies, a stable model is crucial for exploring infection mechanisms, antibiotic resistance, and evaluating materials' antibiofilm performance. Cultured biofilms often face challenges, such as slow maturation or rapid bacteria dispersion. Therefore, developing a stable, mature-stage biofilm model is critical for effective biofilm research. In this study, we report a beads-in-hydrogel biofilm model, in which sodium alginate (SA) hydrogel beads that contain bacteria are embedded within a chitosan-cellulose hydrogel film to simulate natural bacterial biofilms. This model can retain bacteria for a relatively long period of time, preventing their dispersion to the surrounding areas while keeping them viable. The reliability of the model was validated by measuring functional molecules, including extracellular DNA and biofilm-forming related proteins. Overall, this study presents a stable 3D beads-in-hydrogel biofilm model that effectively replicates natural biofilms, providing a reliable platform for exploring infection mechanisms, antibiotic resistance, and evaluating antibiofilm strategies.
Collapse
Affiliation(s)
- Shiwei Li
- School of Engineering, Institute for Bioengineering, University of Edinburgh, The Kings Buildings, EH9 3JL Edinburgh, UK.
| | - Yuxuan Ji
- School of Engineering, Institute for Bioengineering, University of Edinburgh, The Kings Buildings, EH9 3JL Edinburgh, UK.
| | - Xuxiang Xue
- School of Engineering, Institute for Bioengineering, University of Edinburgh, The Kings Buildings, EH9 3JL Edinburgh, UK.
| | - Yaohao Yang
- School of Engineering, Institute for Materials & Processes, University of Edinburgh, The Kings Buildings, EH9 3JL Edinburgh, UK
| | - Yi Huang
- School of Engineering, Institute for Materials & Processes, University of Edinburgh, The Kings Buildings, EH9 3JL Edinburgh, UK
| | - Shuiqing Yang
- Jiangsu Dingying New Materials Co., Ltd. Jiangsu Dingying New Materials Co., Ltd., Changzhou, Jiangsu, 213031, China
| | - Xianfeng Chen
- School of Engineering, Institute for Bioengineering, University of Edinburgh, The Kings Buildings, EH9 3JL Edinburgh, UK.
| |
Collapse
|
7
|
Su SS, An CN, Lin GJ, Li HQ, Yin F, Li XL, Wang KR. Amphiphilic lysine-based glycopeptides exert antibacterial effects on Pseudomonas aeruginosa. J Mater Chem B 2025; 13:3945-3951. [PMID: 40017413 DOI: 10.1039/d4tb02771j] [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: 03/01/2025]
Abstract
The development of new antibiotics with novel antimicrobial mechanisms and strategies has attracted considerable interest. Herein, three amphiphilic lysine-based glycopeptides (SA-l-Gal, OA-l-Gal and LOA-l-Gal) were developed, and their antibacterial activity and inhibition effects on biofilm formation on P. aeruginosa were studied. SA-l-Gal with a stearic acid group modification exhibited better antibacterial effects than the other lysine-based glycopeptides by damaging the bacterial membrane. Furthermore, SA-l-Gal could effectively reduce inflammation factor expression, enhance the formation of new blood vessels, and promote the healing of P. aeruginosa-infected mouse wounds. This result provides insights into the development of glycomimetic drugs against P. aeruginosa infection.
Collapse
Affiliation(s)
- Shan-Shan Su
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Chao-Na An
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Gao-Juan Lin
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Hai-Qing Li
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Fangqian Yin
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
- College of Chemical Engineering & Material, Hebei Key Laboratory of Heterocyclic Compounds, Handan key laboratory of organic small molecule materials, Handan University, Handan 056005, P. R. China
| | - Xiao-Liu Li
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| | - Ke-Rang Wang
- College of Chemistry and Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis (Ministry of Education), State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Chemical Biology of Hebei Province, Hebei University, Baoding, 071002, China.
| |
Collapse
|
8
|
Guo M, Wang H, Zhang H, Bo Z, Zhang C, Zhang X, Wu Y. Identifcation of the genes involved in biofilm formation of Avibacterium paragallinarum using random transposon mutagenesis. Vet Microbiol 2025; 302:110410. [PMID: 39892023 DOI: 10.1016/j.vetmic.2025.110410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/30/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
Infectious coryza (IC) is a respiratory disease in poultry caused by Avibacterium paragallinarum (Av. paragallinarum). The disease caused growth retardation in broilers and reduced egg production in laying hens, resulting in significant economic losses to the global chicken industry. The biofilm is an important virulence factor for many bacterial pathogens, yet there is a paucity of research on the biofilm of Av. paragallinarum. This study aimed to construct a random mutant library of Av. paragallinarum using the Tn5-Kan transposon to identify genes involved in biofilm formation. A total of approximately 3000 mutants were obtained, and 38 of them demonstrated a reduction in biofilm formation of 70-90 % by crystal violet staining. The transposon insertion sites were further determined by chromosome walking, and 17 functional genes related to biofilm formation were identified. According to the functional analysis of the mutated genes, 14 mutants with mutated genes associated with energy metabolism, cell membrane formation, gene transcription and translation, and material transmembrane transport were screened to further explore their biological characteristics and pathogenicity in vivo and in vitro. The results indicated that the growth performance, resistance to disinfectants, adhesion and invasion ability to DF-1 cells and pathogenicity of the 14 mutants were reduced. The 14 mutants displayed increased sensitivity to antibiotics but did not show significant changes in hemagglutination titer or antiserum bactericidal ability. It is noteworthy that the M-76 mutant exhibited a marked reduction in pathogenicity. Following challenge, the experimental chickens did not present any clinical symptoms or pathological changes for a period of seven days, and the respiratory tract bacterial shedding was also the lowest. This indicates that a deficiency in biofilm formation reduces the pathogenicity of Av. paragallinarum. This study will contribute to our understanding of the molecular mechanism of biofilm formation of Av. paragallinarum and further study the pathogenesis of Av. paragallinarum.
Collapse
Affiliation(s)
- Mengjiao Guo
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Haonan Wang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Hao Zhang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zongyi Bo
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Chengcheng Zhang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaorong Zhang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
| | - Yantao Wu
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou 225009, China.
| |
Collapse
|
9
|
Grooters KE, Hayes SL, Richter DM, Ku JC, Sawyer R, Li Y. A novel strategy for eradication of staphylococcal biofilms using blood clots. Front Cell Infect Microbiol 2025; 15:1507486. [PMID: 39958937 PMCID: PMC11827428 DOI: 10.3389/fcimb.2025.1507486] [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: 10/07/2024] [Accepted: 01/09/2025] [Indexed: 02/18/2025] Open
Abstract
Introduction Infections with coagulase negative staphylococcal species (CoNS) are a major cause of mortality and morbidity in joint and heart valve replacement procedures, largely due to biofilm formation. Cells within biofilms have higher rates of antibiotic resistance than their planktonic counterparts; consequently, novel mechanisms are needed to combat these infections. Methods To enhance antibiotic delivery and penetration, this innovative study involved treating CoNS biofilms with murine blood clots impregnated with antibiotics. We then investigated the impact of this treatment on biofilm density, metabolism, and architecture. Results Our pilot study demonstrates that this method of antibiotic delivery results in improved biofilm clearance, relative to conventional exposure methods. Discussion Our results demonstrate that blood clot exposure has an intrinsic impact on biofilm density and potentially reduces colonization, warrenting further investigation into the mechanism.
Collapse
Affiliation(s)
- Kayla E. Grooters
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Sheridan L. Hayes
- Division of Medical Engineering, Department of Surgical Science, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, United States
| | - David M. Richter
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Jennifer C. Ku
- Department of Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Robert Sawyer
- Division of Medical Engineering, Department of Surgical Science, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, United States
| | - Yong Li
- Division of Medical Engineering, Department of Surgical Science, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
10
|
Liu Y, Liu K, Lei L, Wang Q, Wang X, Meng X, Liu Q, Du J, Zhang L, Nazaré M, Hu HY. Aminopeptidase-Responsive NIR Photosensitizer for Precision Targeting and Eradication of Pseudomonas aeruginosa Biofilms. ACS APPLIED MATERIALS & INTERFACES 2025; 17:1-12. [PMID: 39711235 DOI: 10.1021/acsami.4c16028] [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: 12/24/2024]
Abstract
The emergence of resistance in Pseudomonas aeruginosa represents a significant global health challenge, particularly due to the hurdle of effectively penetrating biofilms with antimicrobials. Moreover, the rise of antibiotic-resistant pathogens has driven the urgent need for developing innovative therapeutic approaches to overcome antibiotic resistance. Antibacterial phototherapy strategies have shown great potential for combating pathogens due to their broad-spectrum antimicrobial activity, spatiotemporal controllability, and relatively low rate of resistance emergence. However, due to the lack of bacterial specificity and penetration, photosensitizers cause considerable damage to mammalian cells and normal tissues and are less effective against bacterial biofilms. Herein, we developed a novel dual-targeting antibacterial strategy to construct a near-infrared photosensitizer, Cy-NEO-Leu. Cy-NEO-Leu showed great bacterial targeting affinity, penetrating and accumulating in biofilms. At the site of infection, it was specifically activated by P. aeruginosa aminopeptidase (PaAP), producing Cy-NEO-NH2, which demonstrated outstanding photothermal (PTT) and photodynamic (PDT) properties, with a photothermal conversion efficiency of up to 70.34%. Both in vitro and in vivo results demonstrated that Cy-NEO-Leu significantly reduced the biofilm biomass and bacterial viability in P. aeruginosa biofilms. Moreover, phototherapy with Cy-NEO-Leu further activated the immune system, enhancing therapeutic efficacy and promoting wound healing. RNA-seq analysis revealed that the antibacterial mechanism of Cy-NEO-Leu-mediated phototherapy involves disruption of the transcriptional and translational processes of P. aeruginosa under laser irradiation. Overall, our results present a promising therapeutic approach against P. aeruginosa biofilms and inspire the development of next-generation antimicrobials.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Kaixuan Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ling Lei
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qinghua Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiangchuan Meng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qian Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiacheng Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Leilei Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Marc Nazaré
- Medicinal Chemistry, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Hai-Yu Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
11
|
Azeem K, Fatima S, Ali A, Ubaid A, Husain FM, Abid M. Biochemistry of Bacterial Biofilm: Insights into Antibiotic Resistance Mechanisms and Therapeutic Intervention. Life (Basel) 2025; 15:49. [PMID: 39859989 PMCID: PMC11767195 DOI: 10.3390/life15010049] [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: 12/01/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Biofilms, composed of structured communities of bacteria embedded in a self-produced extracellular matrix, pose a significant challenge due to their heightened resistance to antibiotics and immune responses. This review highlights the mechanisms underpinning antibiotic resistance within bacterial biofilms, elucidating the adaptive strategies employed by microorganisms to withstand conventional antimicrobial agents. This encompasses the role of the extracellular matrix, altered gene expression, and the formation of persister cells, contributing to the recalcitrance of biofilms to eradication. A comprehensive understanding of these resistance mechanisms provides a for exploring innovative therapeutic interventions. This study explores promising avenues for future research, emphasizing the necessity of uncovering the specific genetic and phenotypic adaptations occurring within biofilms. The identification of vulnerabilities in biofilm architecture and the elucidation of key biofilm-specific targets emerge as crucial focal points for the development of targeted therapeutic strategies. In addressing the limitations of traditional antibiotics, this review discusses innovative therapeutic approaches. Nanomaterials with inherent antimicrobial properties, quorum-sensing inhibitors disrupting bacterial communication, and bacteriophages as biofilm-specific viral agents are highlighted as potential alternatives. The exploration of combination therapies, involving antimicrobial agents, biofilm-disrupting enzymes, and immunomodulators, is emphasized to enhance the efficacy of existing treatments and overcome biofilm resilience.
Collapse
Affiliation(s)
- Kashish Azeem
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Sadaf Fatima
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Asghar Ali
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
- Clinical Biochemistry Laboratory, Department of Biochemistry, School of Chemical and Life Science, Jamia Hamdard, New Delhi 110062, India
| | - Ayesha Ubaid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Abid
- Medicinal Chemistry Laboratory, Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (K.A.); (S.F.); (A.A.); (A.U.)
| |
Collapse
|
12
|
Roncarati D, Vannini A, Scarlato V. Temperature sensing and virulence regulation in pathogenic bacteria. Trends Microbiol 2025; 33:66-79. [PMID: 39164134 DOI: 10.1016/j.tim.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024]
Abstract
Pathogenic bacteria can detect a variety of environmental signals, including temperature changes. While sudden and significant temperature variations act as danger signals that trigger a protective heat-shock response, minor temperature fluctuations typically signal to the pathogen that it has moved from one environment to another, such as entering a specific niche within a host during infection. These latter temperature fluctuations are utilized by pathogens to coordinate the expression of crucial virulence factors. Here, we elucidate the critical role of temperature in governing the expression of virulence factors in bacterial pathogens. Moreover, we outline the molecular mechanisms used by pathogens to detect temperature fluctuations, focusing on systems that employ proteins and nucleic acids as sensory devices. We also discuss the potential implications and the extent of the risk that climate change poses to human pathogenic diseases.
Collapse
Affiliation(s)
- Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| | - Andrea Vannini
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Vincenzo Scarlato
- Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
13
|
Roque‐Borda CA, Primo LMDG, Medina‐Alarcón KP, Campos IC, Nascimento CDF, Saraiva MMS, Berchieri Junior A, Fusco‐Almeida AM, Mendes‐Giannini MJS, Perdigão J, Pavan FR, Albericio F. Antimicrobial Peptides: A Promising Alternative to Conventional Antimicrobials for Combating Polymicrobial Biofilms. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410893. [PMID: 39530703 PMCID: PMC11714181 DOI: 10.1002/advs.202410893] [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: 09/06/2024] [Revised: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Polymicrobial biofilms adhere to surfaces and enhance pathogen resistance to conventional treatments, significantly contributing to chronic infections in the respiratory tract, oral cavity, chronic wounds, and on medical devices. This review examines antimicrobial peptides (AMPs) as a promising alternative to traditional antibiotics for treating biofilm-associated infections. AMPs, which can be produced as part of the innate immune response or synthesized therapeutically, have broad-spectrum antimicrobial activity, often disrupting microbial cell membranes and causing cell death. Many specifically target negatively charged bacterial membranes, unlike host cell membranes. Research shows AMPs effectively inhibit and disrupt polymicrobial biofilms and can enhance conventional antibiotics' efficacy. Preclinical and clinical research is advancing, with animal studies and clinical trials showing promise against multidrug-resistant bacteria and fungi. Numerous patents indicate increasing interest in AMPs. However, challenges such as peptide stability, potential cytotoxicity, and high production costs must be addressed. Ongoing research focuses on optimizing AMP structures, enhancing stability, and developing cost-effective production methods. In summary, AMPs offer a novel approach to combating biofilm-associated infections, with their unique mechanisms and synergistic potential with existing antibiotics positioning them as promising candidates for future treatments.
Collapse
Affiliation(s)
- Cesar Augusto Roque‐Borda
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
- Vicerrectorado de InvestigaciónUniversidad Católica de Santa MaríaArequipa04000Peru
| | - Laura Maria Duran Gleriani Primo
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Kaila Petronila Medina‐Alarcón
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Isabella C. Campos
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Camila de Fátima Nascimento
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Mauro M. S. Saraiva
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Angelo Berchieri Junior
- São Paulo State University (UNESP)School of Agricultural and Veterinarian SciencesJaboticabalSao Paulo14884‐900Brazil
| | - Ana Marisa Fusco‐Almeida
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Maria José Soares Mendes‐Giannini
- Department of Clinical AnalysisSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - João Perdigão
- iMed.ULisboa–Institute for Medicines ResearchFaculty of PharmacyUniversity of LisbonLisbon1649004Portugal
| | - Fernando Rogério Pavan
- Department of Biological SciencesSchool of Pharmaceutical SciencesUniversidade Estadual Paulista (UNESP)AraraquaraSao Paulo14800‐903Brazil
| | - Fernando Albericio
- Peptide Science LaboratorySchool of Chemistry and PhysicsUniversity of KwaZulu‐NatalDurban4001South Africa
- CIBER‐BBNNetworking Centre on BioengineeringBiomaterials and Nanomedicineand Department of Organic ChemistryUniversity of BarcelonaBarcelona08028Spain
| |
Collapse
|
14
|
Hofmeisterová L, Bajer T, Walczak M, Šilha D. Chemical Composition and Antibacterial Effect of Clove and Thyme Essential Oils on Growth Inhibition and Biofilm Formation of Arcobacter spp. and Other Bacteria. Antibiotics (Basel) 2024; 13:1232. [PMID: 39766622 PMCID: PMC11672449 DOI: 10.3390/antibiotics13121232] [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: 11/28/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: In recent years, significant resistance of microorganisms to antibiotics has been observed. A biofilm is a structure that significantly aids the survival of the microbial population and also significantly affects its resistance. Methods: Thyme and clove essential oils (EOs) were subjected to chemical analysis using gas chromatography coupled to mass spectrometry (GC-MS) and gas chromatography with a flame ionization detector (GC-FID). Furthermore, the antimicrobial effect of these EOs was tested in both the liquid and vapor phases using the volatilization method. The effect of the EOs on growth parameters was monitored using an RTS-8 bioreactor. However, the effect of the EOs on the biofilm formation of commonly occurring bacteria with pathogenic potential was also monitored, but for less described and yet clinically important strains of Arcobacter spp. Results: In total, 37 and 28 compounds were identified in the thyme and clove EO samples, respectively. The most common were terpenes and also derivatives of phenolic substances. Both EOs exhibited antimicrobial activity in the liquid and/or vapor phase against at least some strains. The determined antimicrobial activity of thyme and clove oil was in the range of 32-1024 µg/mL in the liquid phase and 512-1024 µg/mL in the vapor phase, respectively. The results of the antimicrobial effect are also supported by similar conclusions from monitoring growth curves using the RTS bioreactor. The effect of EOs on biofilm formation differed between strains. Biofilm formation of Pseudomonas aeruginosa was completely suppressed in an environment with a thyme EO concentration of 1024 µg/mL. On the other hand, increased biofilm formation was found, e.g., in an environment of low concentration (1-32 µg/mL). Conclusions: The potential of using natural matrices as antimicrobials or preservatives is evident. The effect of these EOs on biofilm formation, especially Arcobacter strains, is described for the first time.
Collapse
Affiliation(s)
- Leona Hofmeisterová
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic;
| | - Tomáš Bajer
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic;
| | - Maciej Walczak
- Department of Environmental Microbiology and Biotechnology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Lwowska 1, 87-100 Toruń, Poland;
| | - David Šilha
- Department of Biological and Biochemical Sciences, Faculty of Chemical Technology, University of Pardubice, Studentská 573, 532 10 Pardubice, Czech Republic;
| |
Collapse
|
15
|
Sathishkumar P, Khan F. Leveraging bacteria-inspired nanomaterials for targeted controlling biofilm and virulence properties of Pseudomonas aeruginosa. Microb Pathog 2024; 197:107103. [PMID: 39505089 DOI: 10.1016/j.micpath.2024.107103] [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: 09/12/2024] [Revised: 10/24/2024] [Accepted: 11/03/2024] [Indexed: 11/08/2024]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen designated as a high-priority pathogen because of its role in major healthcare-associated and nosocomial infections. Biofilm production by these bacteria is one of the adaptive resistance mechanisms to traditional antibiotics, making treatment challenging, especially for immunocompromised patients. P. aeruginosa also produces a variety of virulence factors, which aid in invasion, adhesion, persistence, and immune system protection. Recent advances in nanotechnology-based therapy, notably the application of bioinspired metal and metal-oxide nanomaterials, have been seen as a viable way to control P. aeruginosa biofilm and virulence. Because of its ease of growth and culture, synthesizing metal and metal-oxide nanomaterials using bacterial species has become one of the most environmentally benign green synthesis options. The application of bacterial-inspired nanomaterials is particularly successful for targeted control of P. aeruginosa infection due to interactions with cell membrane components and transport systems. This paper delves into and provides a complete overview of the application of bacterial-inspired metal and metal-oxide nanomaterials to treat P. aeruginosa infection by targeting biofilm and virulence characteristics. The review focused on synthesizing and applying gold, silver, copper, iron, magnetite, and zinc oxide nanomaterials to mitigate P. aeruginosa biofilm and virulence. The underlying mechanism of these metal and metal-oxide nanoparticles in relation to biofilm and virulence features has also been thoroughly discussed. The current review introduces novel approaches to treating and controlling drug-resistant P. aeruginosa using bacterial-inspired nanomaterials as a targeted therapeutic strategy.
Collapse
Affiliation(s)
- Palanivel Sathishkumar
- Green Lab, Department of Prosthodontics, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| | - Fazlurrahman Khan
- Ocean and Fisheries Development International Cooperation Institute, Pukyong National University. Busan 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
16
|
Zhang Y, Jiao F, Zeng D, Yu X, Zhou Y, Xue J, Yang W, Guo J. Synergistic Effects of Pyrrosia lingua Caffeoylquinic Acid Compounds with Levofloxacin Against Uropathogenic Escherichia coli: Insights from Molecular Dynamics Simulations, Antibiofilm, and Antimicrobial Assessments. Molecules 2024; 29:5679. [PMID: 39683837 DOI: 10.3390/molecules29235679] [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: 09/15/2024] [Revised: 11/23/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Urinary tract infections (UTIs), primarily caused by uropathogenic Escherichia coli (UPEC), have high morbidity and recurrence rates. Resistance to levofloxacin hydrochloride (LEV), a commonly used treatment for UTIs, is increasingly problematic, exacerbated by biofilm formation mediated by interactions between cyclic di-GMP (c-di-GMP or CDG) and YcgR. In this study, we identified three caffeoylquinic acid compounds from Pyrrosia lingua-chlorogenic acid (CGA), sibiricose A5 (Si-A5), and 3-O-caffeoylquinic acid methyl ester (CAM)-that target YcgR through molecular docking. Biological assays revealed that combining these compounds with levofloxacin hydrochloride significantly enhanced antibacterial activity against standard UPEC strains in a concentration-dependent manner and clinically isolated UPEC strains. Notably, chlorogenic acid and sibiricose A5, when used with levofloxacin hydrochloride, enhanced intracellular c-di-GMP levels and swimming motility, significantly reduced YcgR gene expression, and effectively inhibited biofilm formation of UPEC at multiple time points. Additionally, molecular dynamics simulations elucidated the strong binding of these compounds to YcgR, underscoring the critical roles of residues, such as Arg118 and Asp145. This research serves as a foundation for tackling antibiotic resistance and developing innovative therapeutics for UTIs.
Collapse
Affiliation(s)
- Yan Zhang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Fangfang Jiao
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Derong Zeng
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Xiang Yu
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Yongqiang Zhou
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Juan Xue
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Wude Yang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Jingjing Guo
- Centre in Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| |
Collapse
|
17
|
Zhao Y, Xu H, Wang H, Wang P, Chen S. Multidrug resistance in Pseudomonas aeruginosa: genetic control mechanisms and therapeutic advances. MOLECULAR BIOMEDICINE 2024; 5:62. [PMID: 39592545 PMCID: PMC11599538 DOI: 10.1186/s43556-024-00221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Pseudomonas aeruginosa is a significant opportunistic pathogen, and its complex mechanisms of antibiotic resistance pose a challenge to modern medicine. This literature review explores the advancements made from 1979 to 2024 in understanding the regulatory networks of antibiotic resistance genes in Pseudomonas aeruginosa, with a particular focus on the molecular underpinnings of these resistance mechanisms. The review highlights four main pathways involved in drug resistance: reducing outer membrane permeability, enhancing active efflux systems, producing antibiotic-inactivating enzymes, and forming biofilms. These pathways are intricately regulated by a combination of genetic regulation, transcriptional regulators, two-component signal transduction, DNA methylation, and small RNA molecules. Through an in-depth analysis and synthesis of existing literature, we identify key regulatory elements mexT, ampR, and argR as potential targets for novel antimicrobial strategies. A profound understanding of the core control nodes of drug resistance offers a new perspective for therapeutic intervention, suggesting that modulating these elements could potentially reverse resistance and restore bacterial susceptibility to antibiotics. The review looks forward to future research directions, proposing the use of gene editing and systems biology to further understand resistance mechanisms and to develop effective antimicrobial strategies against Pseudomonas aeruginosa. This review is expected to provide innovative solutions to the problem of drug resistance in infectious diseases.
Collapse
Affiliation(s)
- Yuanjing Zhao
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Haoran Xu
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Hui Wang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ping Wang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| | - Simin Chen
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
18
|
Vilas Boas D, Lima CMG, Margalho LP, Amorim-Neto DP, Canales HDS, Lemos Junior WJF, Ramos AC, Saraiva G, Sant'Ana AS. Impact of hydrophobic and hydrophilic surface properties on Pseudomonas aeruginosa adhesion in materials used in mineral water wells. BIOFOULING 2024; 40:735-742. [PMID: 39380146 DOI: 10.1080/08927014.2024.2410771] [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: 08/01/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024]
Abstract
Microbiologically contaminated water is a significant source of infections in humans and animals, with Pseudomonas aeruginosa (PSA) being particularly concerning due to its ability to thrive in water environments and its resistance to many disinfectants. Therefore, this study investigates the adhesion potential of PSA strains on various materials used in mineral water extraction wells, focusing on hydrophobic and hydrophilic properties. Mineral water samples were collected from three wells (P-01, P-07, and P-08) within the Guarani Aquifer System and Fractured Aquifer System (SAF) in Brazil. The physicochemical properties of the water, including concentrations of Sr (strontium), Fe (iron), Si (silicon), SO42- (sulfate ions), Cl- (chloride ions), and ORP (oxidation-reduction potential), were analyzed. Results indicated higher PSA adhesion on hydrophobic materials, particularly high-density polyethylene (HDPE) and geomechanically plasticized polyvinyl chloride (PVC). Multiple correlation analyses revealed positive correlations between PSA adhesion on hydrophilic materials and Sr, Fe, Si, SO42-, and Cl- concentrations. Conversely, ORP negatively correlated with bacterial adhesion on PVC surfaces, suggesting higher ORP values reduced PSA attachment. These findings highlight the importance of water composition and material properties in influencing bacterial adhesion and potential biofilm formation in mineral water extraction systems.
Collapse
Affiliation(s)
- Danilo Vilas Boas
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Clara M G Lima
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Larissa P Margalho
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Dionísio P Amorim-Neto
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | - Héctor D S Canales
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| | | | | | | | - Anderson S Sant'Ana
- Department of Food Science and Nutrition, Faculty of Food Engineering, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
19
|
Biswas T, Ahmed M, Mondal S. Mixed species biofilm: Structure, challenge and its intricate involvement in hospital associated infections. Microb Pathog 2024; 195:106866. [PMID: 39159773 DOI: 10.1016/j.micpath.2024.106866] [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: 05/07/2024] [Revised: 08/06/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Hospital associated infections or healthcare associated infections (HAIs) are a major threat to healthcare and medical management, mostly because of their recalcitrant nature. The primary cause of these HAIs is bacterial associations, especially the interspecies interactions. In interspecies interactions, more than one species co-exists in a common platform of extracellular polymeric substances (EPS), establishing a strong interspecies crosstalk and thereby lead to the formation of mixed species biofilms. In this process, the internal microenvironment and the surrounding EPS matrix of the biofilms ensure the protection of the microorganisms and allow them to survive under antagonistic conditions. The communications between the biofilm members as well as the interactions between the bacterial cells and the matrix polymers, also aid in the rigidity of the biofilm structure and allow the microorganisms to evade both the host immune response and a wide range of anti-microbials. Therefore, to design a treatment protocol for HAIs is difficult and it has become a growing point of concern. This review therefore first aims to discuss the role of microenvironment, molecular structure, cell-cell communication, and metabolism of mixed species biofilms in manifestation of HAIs. In addition, we discuss the electrochemical properties of mixed-species biofilms and their mechanism in developing drug resistance. Then we focus on the most dreaded bacterial HAI including oral and gut multi-species infections, catheter-associated urinary tract infections, surgical site infections, and ventilator-associated pneumonia. Further, we highlight the challenges to eradication of the mixed species biofilms and the current and prospective future strategies for the treatment of mixed species-associated HAI. Together, the review presents a comprehensive understanding of mixed species biofilm-mediated infections in clinical scenario, and summarizes the current challenge and prospect of therapeutic strategies against HAI.
Collapse
Affiliation(s)
| | - Mehnaz Ahmed
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Susmita Mondal
- Department of Life Sciences, Presidency University, Kolkata, India.
| |
Collapse
|
20
|
Mazloumi Jourkouyeh E, Taslimi Eshkalak M, Faezi Ghasemi M, Zahmatkesh H, Rasti B, Zamani H. Diclofenac Sodium and Gentamicin Co-Encapsulated PLGA Nanoparticles: Targeting Extracellular Matrix Components to Combat Biofilm Formation in Pseudomonas aeruginosa PAO1. J CLUST SCI 2024; 35:2475-2488. [DOI: 10.1007/s10876-024-02675-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 01/05/2025]
|
21
|
Hejna M, Dell'Anno M, Liu Y, Rossi L, Aksmann A, Pogorzelski G, Jóźwik A. Assessment of the antibacterial and antioxidant activities of seaweed-derived extracts. Sci Rep 2024; 14:21044. [PMID: 39251803 PMCID: PMC11383966 DOI: 10.1038/s41598-024-71961-8] [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: 06/14/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
In swine farming, animals develop diseases that require the use of antibiotics. In-feed antibiotics as growth promoters have been banned due to the increasing concern of antimicrobial resistance. Seaweeds offer bioactive molecules with antibacterial and antioxidant properties. The aim was to estimate the in vitro properties of seaweed extracts: Ascophyllum nodosum (AN), Palmaria palmata (PP), Ulva lactuca (UL), and 1:1 mixes (ANPP, ANUL, PPUL). Escherichia coli strains were used to test for growth inhibitory activity, and chemical-based assays were performed for antioxidant properties. The treatments were 2 (with/without Escherichia coli) × 2 (F4 + and F18 +) × 5 doses (0, 1.44, 2.87, 5.75, 11.50, and 23.0 mg/mL). Bacteria were supplemented with seaweed extracts, and growth was monitored. The antioxidant activity was assessed with 6 doses (0, 1, 50, 100, 200, 500, and 600 mg/mL) × 6 compounds using two chemical assays. Data were evaluated through SAS. The results showed that AN and UL significantly inhibited (p < 0.05) the growth of F4 + and F18 +. PP and mixes did not display an inhibition of the bacteria growth. AN, PP, UL extracts, and mixes exhibited antioxidant activities, with AN showing the strongest dose-response. Thus, AN and UL seaweed extracts reveal promising antibacterial and antioxidant effects and may be candidates for in-feed additives.
Collapse
Affiliation(s)
- Monika Hejna
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Postępu 36A, 05-552, Jastrzębiec, Poland.
| | - Matteo Dell'Anno
- Department of Veterinary Medicine and Animal Sciences-DIVAS, Università degli Studi di Milano, Dell'Università 6, 26900, Lodi, Italy
| | - Yanhong Liu
- Department of Animal Science, University of California, 2251 Meyer Hall, One Shields Ave, Davis, CA, 95616, USA
| | - Luciana Rossi
- Department of Veterinary Medicine and Animal Sciences-DIVAS, Università degli Studi di Milano, Dell'Università 6, 26900, Lodi, Italy
| | - Anna Aksmann
- Department of Plant Experimental Biology and Biotechnology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdańsk, Poland
| | - Grzegorz Pogorzelski
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Postępu 36A, 05-552, Jastrzębiec, Poland
| | - Artur Jóźwik
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Postępu 36A, 05-552, Jastrzębiec, Poland
| |
Collapse
|
22
|
Conway J, Delanois RE, Mont MA, Stavrakis A, McPherson E, Stolarski E, Incavo S, Oakes D, Salvagno R, Adams JS, Kisch-Hancock A, Tenorio E, Leighton A, Ryser S, Kauvar LM, Bernthal NM. Phase 1 study of the pharmacokinetics and clinical proof-of-concept activity of a biofilm-disrupting human monoclonal antibody in patients with chronic prosthetic joint infection of the knee or hip. Antimicrob Agents Chemother 2024; 68:e0065524. [PMID: 39012102 PMCID: PMC11304715 DOI: 10.1128/aac.00655-24] [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: 05/02/2024] [Accepted: 06/24/2024] [Indexed: 07/17/2024] Open
Abstract
We report the results of a first-in-human phase 1 clinical study to evaluate TRL1068, a native human monoclonal antibody that disrupts bacterial biofilms with broad-spectrum activity against both Gram-positive and Gram-negative species. The study population consisted of patients with chronic periprosthetic joint infections (PJIs) of the knee or hip, including both monomicrobial and polymicrobial infections, that are highly resistant to antibiotics due to biofilm formation. TRL1068 was administered via a single pre-surgical intravenous infusion in three sequentially ascending dose groups (6, 15, and 30 mg/kg). Concomitant perioperative antibiotics were pathogen-targeted as prescribed by the treating physician. In this double-blinded study, 4 patients were randomized to receive placebo and 11 patients to receive TRL1068 on day 1, as well as targeted antibiotics for 7 days prior to the scheduled removal of the infected implant and placement of an antibiotic-eluting spacer as the first stage of the standard of care two-stage exchange arthroplasty. No adverse events attributable to TRL1068 were reported. TRL1068 serum half-life was 15-18 days. At day 8, the concentration in synovial fluid was approximately 60% of the blood level and thus at least 15-fold above the threshold for biofilm-disrupting activity in vitro. Explanted prostheses were sonicated to release adherent bacteria for culture, with elimination of the implant bacteria observed in 3 of the 11 patients who received TRL1068, which compares favorably to prior PJI treatments. None of the patients who received TRL1068 had a relapse of the original infection by the end of the study (day 169). CLINICAL TRIALS This study is registered with ClinicalTrials.gov as NCT04763759.
Collapse
Affiliation(s)
- Janet Conway
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, Maryland
| | - Ronald E. Delanois
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, Maryland
| | - Michael A. Mont
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, LifeBridge Health, Baltimore, Maryland
| | | | - Edward McPherson
- University of California Los Angeles, Los Angeles, California, USA
| | | | | | - Daniel Oakes
- University of Southern California, Los Angeles, California, USA
| | | | - John S. Adams
- University of California Los Angeles, Los Angeles, California, USA
| | | | - Edgar Tenorio
- Trellis Bioscience, Inc., Redwood City, California, USA
| | | | - Stefan Ryser
- Trellis Bioscience, Inc., Redwood City, California, USA
| | | | | |
Collapse
|
23
|
Thakkar N, Gajera G, Mehta D, Kothari V. Silversol ® (a Colloidal Nanosilver Formulation) Inhibits Growth of Antibiotic-Resistant Staphylococcus aureus by Disrupting Its Physiology in Multiple Ways. Pharmaceutics 2024; 16:726. [PMID: 38931848 PMCID: PMC11206351 DOI: 10.3390/pharmaceutics16060726] [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: 05/02/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Antibiotic-resistant strains of Staphylococcus aureus are being viewed as a serious threat by various public health agencies. Identifying novel targets in this important pathogen is crucial to the development of new effective antibacterial formulations. We investigated the antibacterial effect of a colloidal nanosilver formulation, Silversol®, against an antibiotic-resistant strain of S. aureus using appropriate in vitro assays. Moreover, we deciphered the molecular mechanisms underlying this formulation's anti-S. aureus activity using whole transcriptome analysis. Lower concentrations of the test formulation exerted a bacteriostatic effect against this pathogen, and higher concentrations exerted a bactericidal effect. Silversol® at sub-lethal concentration was found to disturb multiple physiological traits of S. aureus such as growth, antibiotic susceptibility, membrane permeability, efflux, protein synthesis and export, biofilm and exopolysaccharide production, etc. Transcriptome data revealed that the genes coding for transcriptional regulators, efflux machinery, transferases, β-lactam resistance, oxidoreductases, metal homeostasis, virulence factors, and arginine biosynthesis are expressed differently under the influence of the test formulation. Genes (argG and argH) involved in arginine biosynthesis emerged among the major targets of Silversol®'s antibacterial activity against S. aureus.
Collapse
Affiliation(s)
- Nidhi Thakkar
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| | - Gemini Gajera
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| | - Dilip Mehta
- Viridis BioPharma Pvt. Ltd., Mumbai 400043, India;
| | - Vijay Kothari
- Institute of Science, Nirma University, Ahmedabad 382481, India; (N.T.); (G.G.)
| |
Collapse
|
24
|
Shen Y, Pan M, Gao H, Zhang Y, Wang R, Li J, Mao Z. New azole derivatives linked to indole/indoline moieties combined with FLC against drug-resistant Candida albicans. RSC Med Chem 2024; 15:1236-1246. [PMID: 38665837 PMCID: PMC11042159 DOI: 10.1039/d4md00021h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/26/2024] [Indexed: 04/28/2024] Open
Abstract
Candida albicans is the most common fungal pathogen associated with human opportunistic infections. Invasive infections caused by C. albicans are becoming increasingly serious. However, with the rising incidence of fungal infection, many fungi are resistant to commonly used drugs. Therefore, there is an urgent need for exploring new anti-fungal drugs that fungi are not resistant to. A series of novel azole derivatives linked to indole/indoline moieties were prepared, and in vitro antifungal activity evaluated. All compounds combined with FLC showed excellent activity against drug-resistant C. albicans with low toxicity. A preliminary mechanistic study indicated that S1 combined with FLC could inhibit the formation of C. albicans biofilms as well as destroy the integrity of cell-membrane structure and mitochondrial function. S1 could be considered a new fungal agent for further study.
Collapse
Affiliation(s)
- Yunhong Shen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 PR China
| | - Min Pan
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 PR China
| | - Hui Gao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 PR China
| | - Yi Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 PR China
| | - Ruirui Wang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 PR China
| | - Jun Li
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 PR China
| | - Zewei Mao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 PR China
| |
Collapse
|
25
|
Carvalho-Silva JM, Teixeira ABV, Valente MLDC, Shimano MVW, Dos Reis AC. Antimicrobial activity of essential oils against biofilms formed in dental acrylic resin: a systematic review of in vitro studies. BIOFOULING 2024; 40:114-129. [PMID: 38538551 DOI: 10.1080/08927014.2024.2332709] [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: 05/06/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024]
Abstract
This study aimed to answer the question formulated according to the PICO strategy: 'Which essential oils show antimicrobial activity against biofilms formed on dental acrylic resin?' composed by population (dental acrylic resin), intervention (application of essential oils), comparison (denture cleansers, antifungal drugs, chlorhexidine, and oral mouthwashes), and outcome (antibiofilm activity). In vitro experimental studies evaluating the activity of EOs on biofilm formed on acrylic resin were included. PRISMA guidelines were followed, and the search was performed in the PubMed, Science Direct, Embase, and Lilacs databases and in the gray literature using Google Scholar and ProQuest in December 2023. A manual search of the reference lists of the included primary studies was performed. Of the 1467 articles identified, 37 were selected for full-text reading and 12 were included. Twelve EOs were evaluated, of which 11 showed activity against Candida spp., 3 against Staphylococcus aureus, and 1 against Pseudomonas aeruginosa. The EOs of Cymbopogon citratus, Cinnamomum zeylanicum, and Cymbopogon nardus showed higher action than chlorhexidine, C. nardus higher than Listerine, C. citratus higher than nystatin, and Melaleuca alternifolia higher than fluconazole and nystatin. However, chlorhexidine was more effective than Lippia sidoides and Salvia officinalis, sodium hypochlorite was more effective than L. sidoides, nystatin was more effective than Zingiber officinale, Amphotericin B more effective than Eucalyptus globulus and M. alternifolia. In conclusion, the EOs of C. zeylanicum, C. citratus, C. nardus, and M. alternifolia showed antimicrobial activity to reduce biofilm on dental acrylic resin.
Collapse
Affiliation(s)
- João Marcos Carvalho-Silva
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Ana Beatriz Vilela Teixeira
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Mariana Lima da Costa Valente
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Marcos Vinicius Wada Shimano
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Andréa Cândido Dos Reis
- Department of Dental Materials and Prosthesis, Ribeirão Preto School of Dentistry, University of São Paulo (USP), Ribeirão Preto, Brazil
| |
Collapse
|