1
|
Pereira ABV, Terroso MS, Gonçalves H, Catita J, Santos D, Baptista RMF, Monteiro FJ, Lopes CM, Lúcio M, Ferraz MP. Hybrid nano-scaffolds loaded with resveratrol and Omega-3 fatty Acids: An innovative antimicrobial strategy against biofilm. Int J Pharm 2025; 680:125784. [PMID: 40446869 DOI: 10.1016/j.ijpharm.2025.125784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/26/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025]
Abstract
It is widely accepted that biofilms associated with body tissues are responsible for many antibiotic treatment failures and contribute to various chronic infections. Therefore, it is critical to create novel approaches to wound treatment. The development of nanocarriers to combat biofilms formation has been an area of much research in recent years. The main aim of this work was to develop hybrid nano-scaffolds composed by nanofibers and liposomes, i.e. large unilamellar vesicles (LUVs), loaded with resveratrol (RSV) and/or omega-3 fatty acids (ω3) and to evaluate their capacity to prevent biofilm formation. The studies carried out included the preparation and characterization of LUVs, evaluation of antibiofilm activity for Staphylococcus aureus and Escherichia coli, and cytotoxicity evaluation using fibroblasts. RSV and ω3 loaded LUVs were effective in inhibiting biofilms formation, resulting in an increased antibiofilm effect (almost 100 %) compared to free RSV and ω3 (65 % - 96.4 %). The biocompatibility of the LUVs was also confirmed (cell viability above 70 %), and a positive effect on cell proliferation (cell viability above 70 %) was observed after 7 days. The porous structure and random orientation of the produced nanofibers impregnated with LUVs enable the exchange of exudates, gases, and nutrients, while resembling the extracellular matrix of the skin, encouraging cell adhesion and proliferation. The nanofibers are characterized by adequate mechanical properties (elongation at break of 208 ± 9 % and Young's modulus of 50.3 ± 0.3 MPa) and a high surface area to volume ratio, thus increasing the release profile of RSV (94.8 ± 2.6 % of RSV within 30 min), which is advantageous for anti-biofilm activity. The impregnation of nanofibers with LUVs loaded with RSV and/or ω3 may represent a promising approach for improving wound healing therapies, allowing the control of biofilm formation and even promoting skin regeneration.
Collapse
Affiliation(s)
- Ana Beatriz V Pereira
- FEUP/DEM - Departamento de Engenharia Mecânica, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Mariana S Terroso
- FEUP/DEM - Departamento de Engenharia Mecânica, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | | | - José Catita
- Paralab, SA, Valbom, Portugal; FP-I3ID - Instituto de Investigação, Inovação e Desenvolvimento; FP-BHS - Biomedical and Health Sciences Research Unit, Faculdade Ciências da Saúde, Universidade Fernando Pessoa, Porto, Portugal; RISE - Health, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Daniela Santos
- CF-UM-UP - Centro de Física das Universidades do Minho e Porto; LAPMET - Laboratório para Materiais e Tecnologias Emergentes, Universidade do Minho, Braga, Portugal
| | - Rosa M F Baptista
- CF-UM-UP - Centro de Física das Universidades do Minho e Porto; LAPMET - Laboratório para Materiais e Tecnologias Emergentes, Universidade do Minho, Braga, Portugal
| | - Fernando J Monteiro
- FEUP/DEM - Departamento de Engenharia Mecânica, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Carla M Lopes
- FP-I3ID - Instituto de Investigação, Inovação e Desenvolvimento; FP-BHS - Biomedical and Health Sciences Research Unit, Faculdade Ciências da Saúde, Universidade Fernando Pessoa, Porto, Portugal; RISE - Health, Faculdade de Medicina da Universidade do Porto, Porto, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy; UCIBIO-Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marlene Lúcio
- CF-UM-UP - Centro de Física das Universidades do Minho e Porto; LAPMET - Laboratório para Materiais e Tecnologias Emergentes, Universidade do Minho, Braga, Portugal; CBMA - Centro de Biologia Molecular e Ambiental, Universidade do Minho, Braga, Portugal.
| | - Maria P Ferraz
- FEUP/DEM - Departamento de Engenharia Mecânica, Faculdade de Engenharia da Universidade do Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
2
|
Vivekanandan K, Kumar PV, Jaysree R, Rajeshwari T. Exploring molecular mechanisms of drug resistance in bacteria and progressions in CRISPR/Cas9-based genome expurgation solutions. Glob Med Genet 2025; 12:100042. [PMID: 40051841 PMCID: PMC11883354 DOI: 10.1016/j.gmg.2025.100042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Antibiotic resistance in bacteria is a critical global health challenge, driven by molecular mechanisms such as genetic mutations, efflux pumps, enzymatic degradation of antibiotics, target site modifications, and biofilm formation. Horizontal gene transfer (HGT) further accelerates the spread of resistance genes across bacterial populations. These mechanisms contribute to the emergence of multidrug-resistant (MDR) strains, rendering conventional antibiotics ineffective. Recent advancements in CRISPR/Cas9-based genome editing offer innovative solutions to combat drug resistance. CRISPR/Cas9 enables precise targeting of resistance genes, facilitating their deletion or inactivation, and provides a potential method to eliminate resistance-carrying plasmids. Furthermore, phage-delivered CRISPR systems show promise in selectively killing resistant bacteria while leaving susceptible strains unaffected. Despite challenges such as efficient delivery, off-target effects, and potential bacterial resistance to CRISPR itself, ongoing research and technological innovations hold promise for using CRISPR-based antimicrobials to reverse bacterial drug resistance and develop more effective therapies. These abstract highlights the molecular mechanisms underlying bacterial drug resistance and explores how CRISPR/Cas9 technology could revolutionize treatment strategies against resistant pathogens.
Collapse
Affiliation(s)
- K.E. Vivekanandan
- Department of Microbiology, PSG College of Arts and Science, Civil Aerodrome Post, Avinashi Road, Coimbatore, Tamil Nadu 641014, India
| | - P. Vinoth Kumar
- Department of Microbiology, Shri Nehru Maha Vidyalaya, Shri Gambhirmal Bafna Nagar, Malumachampatti, Coimbatore 641050, India
| | - R.C. Jaysree
- Department of Biotechnology, Nehru Arts and Science College, Thirumalayampalayam, Coimbatore 641105, India
| | - T. Rajeshwari
- Department of Biotechnology, Dhanalakshmi Srinivasan College of Arts and Science for Women, Thuraiyur, Perambalur, Tamilnadu 621212, India
| |
Collapse
|
3
|
Song J, Yuan S, Liu S, Wang Y, Yang B, Ji L, He L, Liu S. Novel yellow light-responsive SnO 2/SnS 2 piezo-photocatalysts with excellent performances for tooth whitening and biofilm eradication. J Mater Chem B 2025. [PMID: 40423942 DOI: 10.1039/d5tb00407a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Oral problems caused by pathogens and tooth discoloration have posed great threats to public health in recent years. Timely killing of cariogenic bacteria and removing surface pigments are the key points to treat yellow teeth to restore healthy whitening. Piezo-photocatalysis has been proved to be an effective strategy for treating yellow teeth. However, few effective and safe nanomaterials have been developed to address this issue in the oral field. Herein, yellow light-responsive SnO2/SnS2 heterostructures are proposed for piezo-photocatalytic biofilm eradication and tooth whitening for the first time. Initially, XRD and HRTEM results experimentally verified the formation of SnO2/SnS2 heterostructures. Further, UV-vis DRS spectra indicated that the absorbance in the visible region was effectively improved after the formation of SnO2/SnS2 heterostructures. Subsequently, yellow light with excellent biocompatibility was combined with ultrasonic treatment to explore the piezo-photocatalytic performances of SnO2/SnS2 for tooth whitening and biofilm removal. Results demonstrated that the SnO2/SnS2 heterostructure prepared with a TAA : SnO2 molar ratio of 1 : 1 for 3 h exhibited the best piezo-photocatalytic performance. The degradation efficiency for the food colorant indigo carmine reached 94.12%, which was much higher than that of single SnS2 (48.31%), single SnO2 (near zero) and treating with only irradiation (63.03%) and only ultrasonic (79.41%). Simultaneously, the heterostructures exhibited excellent piezo-photocatalytic tooth whitening effect on stained teeth. Moreover, the SnO2/SnS2 heterojunctions exhibited excellent piezo-photocatalytic performances in bacteria killing and biofilm removal, and the antibacterial efficiencies reached 77.3% and 56.5% for planktonic S. mutans and biofilms, respectively. In addition, synergistic treating process of SnO2/SnS2 heterostructures resulted in excellent biocompatibility, including much less cytotoxicity and negligible enamel damage. In-depth mechanism investigation indicated that the improved piezo-photocatalytic performances were due to the increased carrier separation efficiency and ROS productivity of SnO2/SnS2 heterostructures, demonstrating the great potential of SnO2/SnS2 heterostructures for future dental care field.
Collapse
Affiliation(s)
- Jiahe Song
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China.
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
| | - Shiping Yuan
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China.
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
| | - Shujuan Liu
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China.
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
| | - Yunsong Wang
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China.
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
| | - Bocan Yang
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China.
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
| | - Linwan Ji
- School of Materials Science and Engineering, Harbin Institute of Technology, Harbin 150001, P. R. China.
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
| | - Liangcan He
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, P. R. China
| | - Shaoqin Liu
- Key Laboratory of Micro-Systems and Micro-Structures Manufacturing, Ministry of Education, Harbin Institute of Technology, Harbin 150001, P. R. China.
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150001, P. R. China
| |
Collapse
|
4
|
Banerjee R. Tiny but Mighty: Small RNAs-The Micromanagers of Bacterial Survival, Virulence, and Host-Pathogen Interactions. Noncoding RNA 2025; 11:36. [PMID: 40407594 PMCID: PMC12101431 DOI: 10.3390/ncrna11030036] [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: 03/03/2025] [Revised: 04/07/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Bacterial pathogens have evolved diverse strategies to infect hosts, evade immune responses, and establish successful infections. While the role of transcription factors in bacterial virulence is well documented, emerging evidence highlights the significant contribution of small regulatory RNAs (sRNAs) in bacterial pathogenesis. These sRNAs function as posttranscriptional regulators that fine-tune gene expression, enabling bacteria to adapt rapidly to challenging environments. This review explores the multifaceted roles of bacterial sRNAs in host-pathogen interactions. Firstly, it examines how sRNAs regulate pathogenicity by modulating the expression of key virulence factors, including fimbriae, toxins, and secretion systems, followed by discussing the role of sRNAs in bacterial stress response mechanisms that counteract host immune defenses, such as oxidative and envelope stress. Additionally, this review investigates the involvement of sRNAs in antibiotic resistance by regulating efflux pumps, biofilm formation, and membrane modifications, which contribute to multi-drug resistance phenotypes. Lastly, this review highlights how sRNAs contribute to intra- and interspecies communication through quorum sensing, thereby coordinating bacterial behavior in response to environmental cues. Understanding these regulatory networks governed by sRNAs is essential for the development of innovative antimicrobial strategies. This review highlights the growing significance of sRNAs in bacterial pathogenicity and explores their potential as therapeutic targets for the treatment of bacterial infections.
Collapse
Affiliation(s)
- Rajdeep Banerjee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
5
|
Osunla A, Oloye F, Kayode A, Femi-Oloye O, Okiti A, Servos M, Giesy J. The Slow Pandemic: Emergence of Antimicrobial Resistance in the Postadvent of SARS-CoV-2 Pandemic. Glob Health Epidemiol Genom 2025; 2025:3172234. [PMID: 40271530 PMCID: PMC12017956 DOI: 10.1155/ghe3/3172234] [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: 06/03/2024] [Accepted: 11/29/2024] [Indexed: 04/25/2025] Open
Abstract
Background: The unprecedented outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has dramatically changed the global approach to public health, emphasizing the importance of measures to control and prevent infections. In response to the COVID-19 crisis, stringent hygiene practices and surface disinfection have become the norm, with an unprecedented surge in the use of disinfectants and antiseptics (DAs). Main Text: While these measures have been crucial in curbing the spread of the virus, an emerging concern has taken center stage: the potential impact of the prolonged and widespread use of antimicrobial compounds in these products on the development of antibiotic resistance. Antimicrobial resistance (AMR) has long been recognized as one of the most pressing global health threats. Quaternary ammonium compounds (QAC) such as benzalkonium chloride, benzethonium chloride, and cetylpyridinium chloride, which are extensively used in DAs formulations, have gained less attention in the context of AMR. Conclusion: A high abundance of QACs was detected in wastewater, and certain bacteria such as Pseudomonas aeruginosa, Acinetobacter baumannii, and Enterococcus species developed resistance to these compounds over time. We analyzed the available evidence from the scientific literature, examining the presence and concentrations of QACs in different water sources, and their resistance mechanisms. This review aimed to shed light on the multifaceted challenges that arise from the dual battle against the COVID-19 pandemic and the ongoing global fight against AMR.
Collapse
Affiliation(s)
- Ayodeji Osunla
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Microbiology, Adekunle Ajasin University, Akungba-Akoko, Ondo, Nigeria
| | - Femi Oloye
- Division of Physical and Computational Sciences, University of Pittsburgh, Bradford, Pennsylvania, USA
| | - Adeoye Kayode
- Department of Biochemistry, Genetics, and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Oluwabunmi Femi-Oloye
- Division of Physical and Computational Sciences, University of Pittsburgh, Bradford, Pennsylvania, USA
| | - Ayomide Okiti
- Department of Microbiology, Adekunle Ajasin University, Akungba-Akoko, Ondo, Nigeria
| | - Mark Servos
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - John Giesy
- Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Veterinary Biomedical Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Integrative Biology and Centre for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
6
|
Kao Godinez AK, Villicaña C, Basilio Heredia J, Valdez-Torres JB, Muy-Rangel M, León-Félix J. Facing Foodborne Pathogen Biofilms with Green Antimicrobial Agents: One Health Approach. Molecules 2025; 30:1682. [PMID: 40333625 PMCID: PMC12029461 DOI: 10.3390/molecules30081682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/31/2025] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Food safety is a significant global and local concern due to the threat of foodborne pathogens to public health and food security. Bacterial biofilms are communities of bacteria adhered to surfaces and represent a persistent contamination source in food environments. Their resistance to conventional antimicrobials exacerbates the challenge of eradication, driving the search for alternative strategies to control biofilms. Unconventional or "green" antimicrobial agents have emerged as promising solutions due to their sustainability and effectiveness. These agents include bacteriophages, phage-derived enzymes, plant extracts, and combinations of natural antimicrobials, which offer novel mechanisms for targeting biofilms. This approach aligns with the "One Health" concept, which underscores the interconnectedness of human, animal, and environmental health and advocates for integrated strategies to address public health challenges. Employing unconventional antimicrobial agents to manage bacterial biofilms can enhance food safety, protect public health, and reduce environmental impacts by decreasing reliance on conventional antimicrobials and mitigating antimicrobial resistance. This review explores the use of unconventional antimicrobials to combat foodborne pathogen biofilms, highlighting their mechanisms of action, antibiofilm activities, and the challenges associated with their application in food safety. By addressing these issues from a "One Health" perspective, we aim to demonstrate how such strategies can promote sustainable food safety, improve public health outcomes, and support environmental health, ultimately fostering a more integrated approach to combating foodborne pathogen biofilms.
Collapse
Affiliation(s)
- Ana Karina Kao Godinez
- Centro de Investigación y Desarrollo, A.C., Culiacán 80110, Sinaloa, Mexico; (A.K.K.G.); (J.B.H.); (J.B.V.-T.); (M.M.-R.)
| | - Claudia Villicaña
- SECIHTI-Centro de Investigación en Alimentación y Desarrollo, A.C., Culiacán 80110, Sinaloa, Mexico;
| | - José Basilio Heredia
- Centro de Investigación y Desarrollo, A.C., Culiacán 80110, Sinaloa, Mexico; (A.K.K.G.); (J.B.H.); (J.B.V.-T.); (M.M.-R.)
| | - José Benigno Valdez-Torres
- Centro de Investigación y Desarrollo, A.C., Culiacán 80110, Sinaloa, Mexico; (A.K.K.G.); (J.B.H.); (J.B.V.-T.); (M.M.-R.)
| | - Maria Muy-Rangel
- Centro de Investigación y Desarrollo, A.C., Culiacán 80110, Sinaloa, Mexico; (A.K.K.G.); (J.B.H.); (J.B.V.-T.); (M.M.-R.)
| | - Josefina León-Félix
- Centro de Investigación y Desarrollo, A.C., Culiacán 80110, Sinaloa, Mexico; (A.K.K.G.); (J.B.H.); (J.B.V.-T.); (M.M.-R.)
| |
Collapse
|
7
|
Pitton M, Valente LG, Oberhaensli S, Gözel B, Jakob SM, Sendi P, Fürholz M, Cameron DR, Que YA. Targeting Chronic Biofilm Infections With Patient-derived Phages: An In Vitro and Ex Vivo Proof-of-concept Study in Patients With Left Ventricular Assist Devices. Open Forum Infect Dis 2025; 12:ofaf158. [PMID: 40182131 PMCID: PMC11966103 DOI: 10.1093/ofid/ofaf158] [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: 11/20/2024] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Background Phage therapy is being reconsidered as a valuable approach to combat antimicrobial resistance. We recently established a personalized phage therapy pipeline in healthy volunteers, where therapeutic phages were isolated from individuals' skin microbiota. In this study, we aim to validate this pipeline in end-stage heart failure patients supported by left ventricular assist devices (LVADs), focusing on phages targeting Staphylococcus epidermidis, a common pathogen responsible for LVAD infections. Methods Over a 2.5-year period, 45 LVAD patients were consistently sampled at their driveline exit sites and foreheads. S epidermidis strains from patients' foreheads were used to amplify patient-specific phages. Newly isolated phages were characterized and tested against S epidermidis isolates (n = 42) from the patient cohort. The virulent phage vB_SepS_BE22, isolated from a patient with a driveline infection, was further tested for its bactericidal activity against S epidermidis biofilms ex vivo with rifampicin on driveline biofilms. Results S epidermidis was detected in 32 patients, 3 of whom had driveline infections. Phages were isolated from 8 patients, 6 of which were unique and exhibited narrow host ranges, infecting 19%-52% of S epidermidis strains. vB_SepS_BE22, isolated from patient ID25's microbiota, was the only phage that specifically killed S epidermidis clones linked to a patient's infection. vB_SepS_BE22 also reduced bacterial loads in exponential and stationary phase cultures, as well as in biofilms on drivelines when combined with rifampicin. Conclusions This study validated a personalized phage therapy approach, where phages from a patient's own microbiota can be used in chronic infection settings as therapeutic agents.
Collapse
Affiliation(s)
- Melissa Pitton
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Luca G Valente
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Simone Oberhaensli
- Interfaculty Bioinformatics Unit, University of Bern, Bern, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Bülent Gözel
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Stephan M Jakob
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Parham Sendi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Monika Fürholz
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David R Cameron
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Yok-Ai Que
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
8
|
Fong PM, Tang VYM, Xu L, Yam BHC, Pradeep HP, Feng Y, Tao L, Kao RYT, Yang D. Synthetic Cation Transporters Eradicate Drug-Resistant Staphylococcus aureus, Persisters, and Biofilms. JACS AU 2025; 5:1328-1339. [PMID: 40151269 PMCID: PMC11938004 DOI: 10.1021/jacsau.4c01198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 03/29/2025]
Abstract
New drugs are urgently required to address the ongoing health crisis caused by methicillin-resistant Staphylococcus aureus (MRSA) infections. Added to the challenge is the difficult-to-treat persister cells and biofilm which are tolerant to the antibiotics. Here we report a new approach to these problems, describing the design and synthesis of aminoxy-acid-based dipeptides that facilitate cation transport across cell membranes to disrupt bacterial ion homeostasis. Remarkably, these synthetic cation transporters display significant antibacterial activity against MRSA, while maintaining high selectivity over mammalian cells. They also effectively eliminate bacterial persisters and reduce established biofilms. Additionally, they inhibit biofilm formation and suppress bacterial virulent protein secretion, even at subinhibitory concentrations. Their associated antibiotic effects support their in vivo efficacy in murine skin and bloodstream MRSA infection models with no observable toxicity to the host. Mode-of-action analysis indicates that these cation transporters induce cytoplasmic acidification, hyperpolarization, and calcium influx, accelerating autolysis. Given their potent activity against bacterial persisters and biofilms, synthetic cation transporters are an emergent and promising class of compounds in the fight against MRSA infections.
Collapse
Affiliation(s)
- Pak-Ming Fong
- Morningside
Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong 999077, P. R. China
| | - Victor Yat-Man Tang
- Department
of Microbiology and Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong 999077, P. R. China
| | - Lu Xu
- School
of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Bill Hin-Cheung Yam
- Department
of Microbiology and Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong 999077, P. R. China
| | - Halebeedu Prakash Pradeep
- Department
of Microbiology and Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong 999077, P. R. China
| | - Yuhui Feng
- School
of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Liang Tao
- School
of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake
Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| | - Richard Yi-Tsun Kao
- Department
of Microbiology and Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong 999077, P. R. China
| | - Dan Yang
- Morningside
Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong 999077, P. R. China
- School
of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Westlake
Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China
| |
Collapse
|
9
|
Martin A, Doyle N, O'Mahony TF. Sodium dichloroisocyanurate: a promising candidate for the disinfection of resilient drain biofilm. Infect Prev Pract 2025; 7:100446. [PMID: 40008271 PMCID: PMC11850130 DOI: 10.1016/j.infpip.2025.100446] [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: 06/21/2024] [Accepted: 01/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background Biofilms are complex multicellular communities of microorganisms embedded within a protective matrix which confers resistance to various antimicrobials, including biocides. Biofilms can cause a range of human diseases and are responsible for 1.7 million hospital-acquired infections in the US annually, providing an economic burden of $11.5 billion in treatment costs. Biofilm contained within drain and plumbing systems may contain pathogenic viruses and bacteria which pose a significant risk to patient safety within healthcare environments. Aim The aim of this study was to determine if three hospital-grade disinfectants (sodium dichloroisocyanurate, peracetic acid and sodium hypochlorite) were capable of killing microorganisms within biofilm, and thus, determining their potential as candidates for drain biofilm disinfection. Methods Pseudomonas aeruginosa biofilms were cultivated using the CDC biofilm reactor, a standardised method for determining disinfectant efficacy against biofilm within the United States of America. Each disinfectant was tested using a one-minute contact time, using the highest concentration available on the product label. Findings The sodium dichloroisocyanurate product was successful in killing biofilm microorganisms, resulting in a log reduction of ≥ 8.70. Peracetic acid reduced biofilm by 3.82 log10 units, followed by sodium hypochlorite, which produced a reduction of 3.78 log10 units. Conclusions The use of a highly effective disinfectant with proven biofilm efficacy can help ensure patient safety and reduce infection levels. Drains and plumbing systems provide a reservoir for potential pathogens and biofilm; thus, drain disinfection is critical in reducing the instance of hospital-acquired infections. Sodium dichloroisocyanurate may provide a reliable solution for drain applications and subsequently, patient wellbeing and safety.
Collapse
Affiliation(s)
- Abbie Martin
- Microbiology & Validation Technician, R&D Department, Kersia Healthcare, Wexford, Ireland
| | - Natasha Doyle
- Principal R&D Scientist, R&D Department, Kersia Healthcare, Wexford, Ireland
| | - Tom F. O'Mahony
- R&D Manager Healthcare, R&D Department, Kersia Healthcare, Wexford, Ireland
| |
Collapse
|
10
|
Kralj S, Da Silva C, Nemec S, Caf M, Fourquaux I, Rols MP, Golzio M, Mertelj A, Kolosnjaj-Tabi J. Dynamically Assembling Magnetic Nanochains as New Generation of Swarm-Type Magneto-Mechanical Nanorobots Affecting Biofilm Integrity. Adv Healthc Mater 2025; 14:e2403736. [PMID: 39757480 DOI: 10.1002/adhm.202403736] [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: 09/28/2024] [Revised: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Bacterial resistance is gaining ground and novel, unconventional strategies are required to improve antibiotic treatments. As a synthetic analog of planktonic bacilli, the natural bacterial swimmers that can penetrate bacterial biofilms, ultra-short propelling magnetic nanochains are presented as bioinspired magnetic nanorobots, enhancing the antibiotic treatment in biofilm-forming Staphylococcus epidermidis. Propelling nanochains, activated by a low intensity (<20 mT) and low frequency (<10 Hz) rotating magnetic field (RMF), prompt the otherwise resistant biofilm-forming bacteria to become sensitive to methicillin, resulting in the killing of 99.99% of bacteria. While magnetic force-driven spherical magnetic nanoparticles were previously reported as unidirectional biofilm channel diggers, propelling nanochains emerge as second-generation magnetic nanorobots, which, due to their magnetic core, shape anisotropy, and negative zeta potential, combine magnetic responsiveness, torque-driven movement, and attractive electrostatic interactions to attach to bacterial aggregates and multi-directionally protrude throughout the biofilm, indulging mechanical forces. These synergistic effects, in combination with an antibiotic drug, destroy the bacterial extracellular matrix and eradicate the formed biofilm, as confirmed with several complementary techniques.
Collapse
Affiliation(s)
- Slavko Kralj
- Jožef Stefan Institute, Jamova cesta 39, Ljubljana, 1000, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana, 1000, Slovenia
| | - Charlotte Da Silva
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), 205 Route de Narbonne, Toulouse, 31400, France
| | - Sebastjan Nemec
- Jožef Stefan Institute, Jamova cesta 39, Ljubljana, 1000, Slovenia
| | - Maja Caf
- Jožef Stefan Institute, Jamova cesta 39, Ljubljana, 1000, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana, 1000, Slovenia
| | - Isabelle Fourquaux
- Centre de microscopie electronique appliquée à la biologie, 133 Route de Narbonne, Toulouse, 31400, France
| | - Marie-Pierre Rols
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), 205 Route de Narbonne, Toulouse, 31400, France
| | - Muriel Golzio
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), 205 Route de Narbonne, Toulouse, 31400, France
| | - Alenka Mertelj
- Jožef Stefan Institute, Jamova cesta 39, Ljubljana, 1000, Slovenia
| | - Jelena Kolosnjaj-Tabi
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), 205 Route de Narbonne, Toulouse, 31400, France
| |
Collapse
|
11
|
Wilkinson RC, Thomas NE, Bhatti A, Burton MR, Joyce N, Jenkins RE. Phage-Encoded Antimicrobial Peptide gp28 Demonstrates LL-37-Like Antimicrobial Activity Against Multidrug-Resistant Pseudomonas aeruginosa. PHAGE (NEW ROCHELLE, N.Y.) 2025; 6:12-19. [PMID: 40351406 PMCID: PMC12063199 DOI: 10.1089/phage.2024.0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Background Pseudomonas aeruginosa (P. aeruginosa) is a gram-negative bacterial pathogen commonly associated with nosocomial infections. Treatment of P. aeruginosa infections is notoriously difficult due to biofilm formation and antibiotic resistance. Antimicrobial peptides (AMPs) are thought to be promising new antimicrobials. Gp28, a phage-derived AMP, is a novel class of characterized phage AMPs with activity against Escherichia coli in a manner similar to the human peptide LL-37. LL-37 exhibits strong antimicrobial activity against P. aeruginosa as well as biofilm disruption and synergy with certain antibiotics posing the question whether gp28 could act similarly. Methods Antibacterial activity of gp28 against P. aeruginosa was established using growth inhibition assays, with minimum inhibitory concentration calculated. Biofilm disruption was assessed using crystal violet staining and scanning electron microscopy. Combined treatment of gp28 with tobramycin against P. aeruginosa was measured using a modified time-kill assay at sublethal concentrations. Results Gp28 inhibits P. aeruginosa planktonic growth, with a minimum inhibitory concentration of 109 μg mL-1 and disrupts established biofilms. We demonstrate that gp28 increases the susceptibility of P. aeruginosa to tobramycin. Conclusions Gp28 demonstrates potential for development as a putative therapeutic agent against a clinically resistant P. aeruginosa strain either alone or in combination with the frontline antibiotic tobramycin.
Collapse
Affiliation(s)
- Rachael C. Wilkinson
- Swansea University Medical School, Healthcare Technology Centre, Swansea University, Swansea, United Kingdom
| | - Nerissa E. Thomas
- Swansea University Medical School, Healthcare Technology Centre, Swansea University, Swansea, United Kingdom
| | - Amita Bhatti
- Swansea University Medical School, Healthcare Technology Centre, Swansea University, Swansea, United Kingdom
| | - Matthew R. Burton
- SPECIFIC-IKC, Department of Materials Science and Engineering, Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom
| | - Naomi Joyce
- Swansea University Medical School, Healthcare Technology Centre, Swansea University, Swansea, United Kingdom
| | - Rowena E. Jenkins
- Swansea University Medical School, Healthcare Technology Centre, Swansea University, Swansea, United Kingdom
| |
Collapse
|
12
|
Gondil V, Ashcraft M, Ghalei S, Kumar A, Wilson SN, Devine R, Handa H, Brisbois EJ. Anti-Infective Bacteriophage Immobilized Nitric Oxide-Releasing Surface for Prevention of Thrombosis and Device-Associated Infections. ACS APPLIED BIO MATERIALS 2025; 8:1362-1376. [PMID: 39895136 PMCID: PMC11836933 DOI: 10.1021/acsabm.4c01638] [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: 11/07/2024] [Revised: 12/31/2024] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
The treatment of critically ill patients has made great strides in the past few decades due to the rapid development of indwelling medical devices. Despite immense advancements in the design of these devices, indwelling medical device-associated infections and thrombosis are two major clinical problems that may lead to device failure and compromise clinical outcomes. Antibiotics are the current treatment choice for these infections; however, the global emergence of antibiotic-resistance and their biofilm formation abilities complicate the management of such infections. Moreover, systemic administration of anticoagulants has been used to counter medical device-induced thrombosis, but a range of serious adverse effects associated with all types of available anticoagulants entails exploring alternative options to counter device-associated thrombosis. In this study, bacteriophages (phages) were covalently immobilized on polydimethylsiloxane (PDMS) surface containing the nitric oxide (NO) donor S-nitroso-N-acetylpenicillamine (SNAP) via SNAP impregnation method. This dual strategy combines the targeted antibacterial activity of phages against bacterial pathogens with the antibacterial-antithrombotic activity of NO released from the polymeric surface. The PDMS, SNAP-PDMS, phage-immobilized PDMS (PDMS-Phage), and phage-immobilized SNAP-PDMS (SNAP-PDMS-Phage) surfaces were characterized for their surface topology, elemental composition, contact angle, SNAP loading, NO release and phage distribution. SNAP-PDMS and SNAP-PDMS-Phage surfaces showed similar and consistent NO release profiles over 24 h of incubation. Immobilization of whole phages on PDMS and SNAP-PDMS was achieved with densities of 2.4 ± 0.54 and 2.1 ± 0.33 phages μm-2, respectively. Immobilized phages were found to retain their activity, and SNAP-PDMS-Phage surfaces showed a significant reduction in planktonic (99.99 ± 0.08%) as well as adhered (99.80 ± 0.05%) Escherichia coli as compared to controls in log killing assays. The SNAP-PDMS-Phage surfaces also exhibited significantly reduced platelet adhesion by 64.65 ± 2.95% as compared to control PDMS surfaces. All fabricated surfaces were found to be nonhemolytic and do not exhibit any significant cytotoxic effects toward mammalian fibroblast cells. This study is the first of its kind to demonstrate the combinatorial pertinence of phages and NO to prevent antibiotic-resistant/sensitive bacterial infections and thrombosis associated with indwelling medical devices.
Collapse
Affiliation(s)
- Vijay
Singh Gondil
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Morgan Ashcraft
- Pharmaceutical
and Biomedical Sciences Department, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Sama Ghalei
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Anil Kumar
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Sarah N. Wilson
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Ryan Devine
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Hitesh Handa
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
- Pharmaceutical
and Biomedical Sciences Department, College of Pharmacy, University of Georgia, Athens, Georgia 30602, United States
| | - Elizabeth J. Brisbois
- School
of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
13
|
Song L, Schwinn LS, Barthel J, Ketter V, Lechler P, Linne U, Rastan AJ, Vogt S, Ruchholtz S, Paletta JRJ, Günther M. Implant-Derived S. aureus Isolates Drive Strain-Specific Invasion Dynamics and Bioenergetic Alterations in Osteoblasts. Antibiotics (Basel) 2025; 14:119. [PMID: 40001363 PMCID: PMC11852183 DOI: 10.3390/antibiotics14020119] [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: 12/09/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Implants are integral to modern orthopedic surgery. The outcomes are good, but infections remain a serious issue. Staphylococcus aureus (S. aureus), along with Staphylococcus epidermidis, are predominant pathogens responsible for implant-associated infections, as conventional antibiotic treatments often fail due to biofilm formation or the pathogens' ability to invade cells and to persist intracellularly. Objectives: This study therefore focused on interactions of S. aureus isolates from infected implants with MG63 and SaOS2 osteoblasts by investigating the adhesion, invasion, and the impact on the bioenergetics of osteoblasts. Methods and Results: We found that the ability of S. aureus to adhere to osteoblasts depends on the isolate and was not associated with a single gene or expression pattern of characteristic adhesion proteins, and further, was not correlated with invasion. However, analysis of invasion capabilities identified better invasion conditions for S. aureus isolates with the SaOS2 osteoblastic cells. Interestingly, metabolic activity of osteoblasts remained unaffected by S. aureus infection, indicating cell survival. In contrast, respiration assays revealed an altered mitochondrial bioenergetic turnover in infected cells. While basal as well as maximal respiration in MG63 osteoblasts were not influenced statistically by S. aureus infections, we found increased non-mitochondrial respiration and enhanced glycolytic activity in the osteoblasts, which was again, more pronounced in the SaOS2 osteoblastic cells. Conclusions: Our findings highlight the complexity of S. aureus-host interactions, where both the pathogen and the host cell contribute to intracellular persistence and survival, representing a major factor for therapeutic failures.
Collapse
Affiliation(s)
- Lei Song
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Lea-Sophie Schwinn
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Juliane Barthel
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Vanessa Ketter
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Philipp Lechler
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Uwe Linne
- Faculty of Chemistry, Philipps-University Marburg, 35032 Marburg, Germany
| | - Ardawan J. Rastan
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Sebastian Vogt
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Steffen Ruchholtz
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Jürgen R. J. Paletta
- Center of Orthopedics and Trauma Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| | - Madeline Günther
- Department of Cardiac and Thoracic Vascular Surgery, Philipps-University Marburg, Universitätsklinikum Gießen and Marburg GmbH, 35043 Marburg, Germany
| |
Collapse
|
14
|
Leggi L, Terzi S, Sartori M, Salamanna F, Boriani L, Asunis E, Griffoni C, Giavaresi G, Gasbarrini A. First Clinical Evidence About the Use of a New Silver-Coated Titanium Alloy Instrumentation to Counteract Surgical Site Infection at the Spine Level. J Funct Biomater 2025; 16:30. [PMID: 39852586 PMCID: PMC11766446 DOI: 10.3390/jfb16010030] [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: 12/11/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Background: Surgical site infections (SSIs) following spinal instrumentation surgery are among the most concerning complications. This study is aimed at assessing the effectiveness of a new treatment approach for SSIs that includes a single-stage approach with the removal of the previous hardware, accurate debridement, and single-stage instrumentation using a silver fixation system (SFS) made of titanium alloy coated with silver (Norm Medical, Ankara, Turkey) by means of a retrospective observational study. Materials and Methods: The demographic data, type of surgery, comorbidities, pathogens, and treatment details of consecutive patients with an SSI who received the SFS between 2018 and 2021 were extracted from their medical records and analyzed. The patients treated with the SFS for primary pyogenic infections were excluded. The patients were re-evaluated at multiple endpoints in order to assess the rate of reinfection and the local and general complications. Results: Fifty-six patients were treated with the SFS and thirty-four patients met the inclusion criteria. Out of those 34 patients, the rate of infection recurrence or insurgence after the implantation of the SFS was 11.8%, with infection detected in 4 out of 34 cases and mechanical problems detected in 2 of the 34 cases (5.9%). The overall success rate in controlling infection recurrence or emergence was 88.2% (30 out of 34 cases). The overall survival rate of the SFS was 87%, 78%, and 71% at one, two, and three years, respectively. Conclusions: The surgical strategy with the SFS demonstrated promising outcomes in preventing infection recurrence or insurgence, with a low incidence of mechanical complications. However, further structured and comprehensive studies are essential for validating these initial findings.
Collapse
Affiliation(s)
- Lucrezia Leggi
- Spine Surgery, IRCCS—Istituto Ortopedico Rizzoli, Via G.C. Pupilli 1, 40136 Bologna, Italy; (L.L.); (S.T.); (L.B.); (E.A.); (C.G.); (A.G.)
| | - Silvia Terzi
- Spine Surgery, IRCCS—Istituto Ortopedico Rizzoli, Via G.C. Pupilli 1, 40136 Bologna, Italy; (L.L.); (S.T.); (L.B.); (E.A.); (C.G.); (A.G.)
| | - Maria Sartori
- Surgical Sciences and Technologies, IRCCS—Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (G.G.)
| | - Francesca Salamanna
- Surgical Sciences and Technologies, IRCCS—Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (G.G.)
| | - Luca Boriani
- Spine Surgery, IRCCS—Istituto Ortopedico Rizzoli, Via G.C. Pupilli 1, 40136 Bologna, Italy; (L.L.); (S.T.); (L.B.); (E.A.); (C.G.); (A.G.)
| | - Emanuela Asunis
- Spine Surgery, IRCCS—Istituto Ortopedico Rizzoli, Via G.C. Pupilli 1, 40136 Bologna, Italy; (L.L.); (S.T.); (L.B.); (E.A.); (C.G.); (A.G.)
| | - Cristiana Griffoni
- Spine Surgery, IRCCS—Istituto Ortopedico Rizzoli, Via G.C. Pupilli 1, 40136 Bologna, Italy; (L.L.); (S.T.); (L.B.); (E.A.); (C.G.); (A.G.)
| | - Gianluca Giavaresi
- Surgical Sciences and Technologies, IRCCS—Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (F.S.); (G.G.)
| | - Alessandro Gasbarrini
- Spine Surgery, IRCCS—Istituto Ortopedico Rizzoli, Via G.C. Pupilli 1, 40136 Bologna, Italy; (L.L.); (S.T.); (L.B.); (E.A.); (C.G.); (A.G.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Massarenti 9, 40128 Bologna, Italy
| |
Collapse
|
15
|
Alzahrani KJ. Tribulus terrestris Fruit Extract: Bioactive Compounds, ADMET Analysis, and Molecular Docking with Penicillin-Binding Protein 2a Transpeptidase of Methicillin-Resistant Staphylococcus epidermidis. Curr Issues Mol Biol 2025; 47:52. [PMID: 39852168 PMCID: PMC11764108 DOI: 10.3390/cimb47010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Tribulus terrestris is a rich source of bioactive molecules and thrives in Mediterranean and desert climate regions worldwide. In this study, Tribulus terrestris methanolic HPLC fractions were evaluated for bioactive compounds and PBP2a transpeptidase inhibitors against methicillin-resistant Staphylococcus epidermidis (MRSE). Among the collected HPLC fractions, F02 of the methanol extract demonstrated potential activity against MRSE01 (15 ± 0.13 mm), MRSE02 (13 ± 0.21 mm), and MRSE03 (16 ± 0.14 mm) isolates. GC-MS analysis of the F02 fraction identified seventeen compounds. Among seventeen compounds, eight have favorable pharmacokinetics and medicinal chemistry; however, on the basis of in silico high water solubility, high GI absorption, blood-brain barrier non-permeability, lack of toxicity, and potential drug-likeness, 1-ethylsulfanylmethyl-2,8,9-trioxa-5-aza-1-sila-bicyclo[3.3.3]undecane and phthalimide, N-(1-hydroxy-2-propyl), were processed for molecular docking. 1-ethylsulfanylmethyl-2,8,9-trioxa-5-aza-1-sila-bicyclo[3.3.3]undecane formed three hydrogen bonds with Ser-452, Thr-584, and Asn-454 residues of the PBP2a transpeptidase. Similarly, phthalimide, N-(1-hydroxy-2-propyl)-formed four hydrogen bonds with Ser-396, Asn-454, Lys-399, and Ser-452 residues of PBP2a transpeptidase. These two compounds are proposed as novel putative PBP2a transpeptidase inhibitors. Further characterization of compounds extracted from Tribulus terrestris may aid in identifying novel PBP2a inhibitory agents for managing MRSE infections.
Collapse
Affiliation(s)
- Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| |
Collapse
|
16
|
Ma X, Poma A. Clinical translation and envisioned impact of nanotech for infection control: Economy, government policy and public awareness. NANOTECHNOLOGY TOOLS FOR INFECTION CONTROL 2025:299-392. [DOI: 10.1016/b978-0-12-823994-0.00004-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
17
|
Patra S, Saha S, Singh R, Tomar N, Gulati P. Biofilm battleground: Unveiling the hidden challenges, current approaches and future perspectives in combating biofilm associated bacterial infections. Microb Pathog 2025; 198:107155. [PMID: 39586337 DOI: 10.1016/j.micpath.2024.107155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
A biofilm is a complex aggregation of microorganisms, either of the same or different species, that adhere to a surface and are encased in an extracellular polymeric substances (EPS) matrix. Quorum sensing (QS) and biofilm formation are closely linked, as QS genes regulate the development, maturation, and breakdown of biofilms. Inhibiting QS can be utilized as an effective approach to combat the impacts of biofilm infection. The impact of biofilms includes chronic infections, industrial biofouling, infrastructure corrosion, and environmental contamination as well. Therefore, a deep understanding of biofilms is crucial for enhancing public health, advancing industrial processes, safeguarding the environment, and deepening our knowledge of microbial life as well. This review aims to offer a comprehensive examination of challenges posed by bacterial biofilms, contemporary approaches and strategies for effectively eliminating biofilms, including the inhibition of quorum sensing pathways, while also focusing on emerging technologies and techniques for biofilm treatment. In addition, future research is projected to target the challenges associated with the bacterial biofilms, striving to develop new approaches and improve existing strategies for their effective control and eradication.
Collapse
Affiliation(s)
- Sandeep Patra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sumana Saha
- Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Randhir Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Nandini Tomar
- Department of Biotechnology, South Asian University, New Delhi, India
| | - Pallavi Gulati
- Ram Lal Anand College, University of Delhi, New Delhi, India.
| |
Collapse
|
18
|
Satarzadeh N, Saraee A, Hatif Mahdi Z, Sadeghi Dousari A, Armanpour M, Taati Moghadam M. Mechanisms in colistin-resistant superbugs transmissible from veterinary, livestock and animal food products to humans. IRANIAN JOURNAL OF VETERINARY RESEARCH 2025; 25:298-311. [PMID: 40386099 PMCID: PMC12085205 DOI: 10.22099/ijvr.2024.50497.7453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/18/2024] [Indexed: 05/20/2025]
Abstract
In the era of antibiotic resistance, where multidrug-resistant (MDR), extensively drug resistant (XDR), and pan-drug resistant (PDR) Gram-negative infections are prevalent, it is crucial to identify the primary sources of antibiotic resistance, understand resistant mechanisms, and develop strategies to combat these mechanisms. The emergence of resistance to last-resort antibiotics like colistin has sparked a war between humanity and resistant bacteria, leaving humanity struggling to find effective countermeasures. Although colistin is used as a highly toxic antibiotic in infections that are not treated with routine antibiotics, its widespread use in animal breeding and veterinary medicine has contributed to the spread of colistin-resistant bacteria, plasmid-borne colistin resistance genes (mcr), and antibiotic residues in livestock and animal-derived foods. These sources can potentially transmit colistin resistance to humans through various routes. Therefore, managing the use of colistin in livestock and animal foods, implementing strict monitoring, and establishing guidelines for its proper use are essential to prevent the escalation of colistin resistance. This review article discusses the latest mechanisms of colistin antibiotic resistance, particularly biofilm production as a public health threat, the livestock and animal food sources of this resistance, and the routes of transmission to humans.
Collapse
Affiliation(s)
- N. Satarzadeh
- Ph.D. in Pharmaceutical Biotechnology, Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - A. Saraee
- Graduated from College of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Z. Hatif Mahdi
- Department of Pathological Analysis, College of Applied Medical Sciences, University of Karbala, Karbala, Iraq
| | - A. Sadeghi Dousari
- Ph.D. in Bacteriology, Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - M. Armanpour
- Department of Pharmacy, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - M. Taati Moghadam
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
19
|
Elsebaei MM, Ezzat HG, Helal AM, El-Shershaby MH, Abdulrahman MS, Alsedawy M, Aljohani AKB, Almaghrabi M, Alsulaimany M, Almohaywi B, Alghamdi R, Miski SF, Musa A, Ahmed HEA. Rational design and synthesis of novel phenyltriazole derivatives targeting MRSA cell wall biosynthesis. RSC Adv 2024; 14:39977-39994. [PMID: 39713184 PMCID: PMC11659749 DOI: 10.1039/d4ra07367c] [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: 10/14/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024] Open
Abstract
Antimicrobial resistance in methicillin-resistant Staphylococcus aureus (MRSA) is a major global health challenge. This study reports the design and synthesis of novel phenyltriazole derivatives as potential anti-MRSA agents. The new scaffold replaces the thiazole core with a 1,2,3-triazole ring, enhancing antimicrobial efficacy and physicochemical properties. A series of derivatives were synthesized and evaluated, with four compounds (20, 23, 29 and 30) showing significant activity against MRSA (MIC ≤ 4 μg mL-1). Compound 29 emerged as the most promising candidate, showing rapid bactericidal activity and superior performance over vancomycin in time-kill assays. It exhibited selective toxicity against bacterial cells, minimal cytotoxicity in human cell lines and low hemolytic activity. Mechanistic studies showed that compound 29 targets the bacterial cell wall by binding to penicillin-binding protein 2a (PBP2a), disrupting cell wall integrity. Additionally, it showed strong anti-biofilm activity and reduced MRSA biofilms by up to 40%. Preliminary pharmacokinetic profiles suggested a favorable profile, including a prolonged plasma half-life and good oral bioavailability. These results suggest that compound 29 is a promising lead for further development in the fight against MRSA.
Collapse
Affiliation(s)
- Mohamed M Elsebaei
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Hany G Ezzat
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Ahmed M Helal
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Mohamed H El-Shershaby
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Mohammed S Abdulrahman
- Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Moaz Alsedawy
- Microbiology and Immunology Department, Faculty of Pharmacy, Al-Azhar University Nasr City 11884 Cairo Egypt
| | - Ahmed K B Aljohani
- Pharmacognosy and Pharmaceutical Chemistry Department, Pharmacy College, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| | - Mohammed Almaghrabi
- Pharmacognosy and Pharmaceutical Chemistry Department, Pharmacy College, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| | - Marwa Alsulaimany
- Pharmacognosy and Pharmaceutical Chemistry Department, Pharmacy College, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| | - Basmah Almohaywi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University Abha 61421 Saudi Arabia
| | - Read Alghamdi
- Pharmacognosy and Pharmaceutical Chemistry Department, Pharmacy College, Taibah University Al-Madinah Al-Munawarah 41477 Saudi Arabia
| | - Samar F Miski
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University Medina 42353 Saudi Arabia
| | - Arafa Musa
- Department of Pharmacognosy, College of Pharmacy, Jouf University Sakaka Aljouf 72341 Saudi Arabia
| | - Hany E A Ahmed
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Al-Azhar University Nasr City 11884 Cairo Egypt
| |
Collapse
|
20
|
Omwenga EO, Awuor SO. The Bacterial Biofilms: Formation, Impacts, and Possible Management Targets in the Healthcare System. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:1542576. [PMID: 39717533 PMCID: PMC11666319 DOI: 10.1155/cjid/1542576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/04/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024]
Abstract
Introduction: The persistent increase in multidrug-resistant pathogens has catalyzed the creation of novel strategies to address antivirulence and anti-infective elements. Such methodologies aim to diminish the selective pressure exerted on bacterial populations, decreasing the likelihood of resistance emergence. This review explores the role of biofilm formation as a significant virulence factor and its impact on the development of antimicrobial resistance (AMR). Case Presentation: The ability of bacteria to form a superstructure-biofilm-has made resistance cases in the microbial world a big concern to public health and other sectors as it is a crucial virulence factor that causes difficulties in the management of infections, hence enhancing chronic infection occurrence. Biofilm formation dates to about 3.4 billion years when prokaryotes were discovered to be forming them and since then due to evolution and growth in science, they are more understood. Management and Outcome: The unique microenvironments within bacterial biofilms diminish antibiotic effectiveness and help bacteria evade the host immune system. Biofilm production is a widespread capability among diverse bacterial species. Biofilm formation is enhanced by quorum sensing (QS), reduction of nutrients, or harsh environments for the bacteria. Conclusion: The rise of severe, treatment-resistant biofilm infections poses major challenges in medicine and agriculture, yet much about how these biofilms form remains unknown.
Collapse
Affiliation(s)
- Eric Omori Omwenga
- Department of Medical Microbiology & Parasitology, School of Health Sciences, Kisii University, Kisii, Kenya
| | - Silas Onyango Awuor
- Department of Applied Health Sciences, School of Health Sciences, Kisii University, Kisii, Kenya
- Department of Medical Microbiology, Jaramogi Oginga Odinga Teaching and Referral Hospital, Kisumu, Kenya
| |
Collapse
|
21
|
Tanasă F, Nechifor M, Teacă CA. Essential Oils as Alternative Green Broad-Spectrum Biocides. PLANTS (BASEL, SWITZERLAND) 2024; 13:3442. [PMID: 39683235 DOI: 10.3390/plants13233442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Natural compounds from plants represent suitable options to replace synthetic biocides when employed against microorganisms in various applications. Essential oils (EOs) have attracted increased interest due to their biocompatible and rather innocuous nature, and complex biological activity (fungicide, biocide and anti-inflammatory, antioxidant, immunomodulatory action, etc.). EOs are complex mixtures of derived metabolites with high volatility obtained from various vegetal parts and employed to a great extent in different healthcare (natural cures, nutrition, phyto- and aromatherapy, spices) and cosmetics applications (perfumery, personal and beauty care), as well as in cleaning products, agriculture and pest control, food conservation and active packaging, or even for restauration and preservation of cultural artifacts. EOs can act in synergy with other compounds, organic and synthetic as well, when employed in different complex formulations. This review will illustrate the employment of EOs in different applications based on some of the most recent reports in a systematic and comprehensive, though not exhaustive, manner. Some critical assessments will also be included, as well as some perspectives in this regard.
Collapse
Affiliation(s)
- Fulga Tanasă
- Polyaddition and Photochemistry Department, "Petru Poni" Institute of Macromolecular Chemistry, 41A Gr. Ghica-Voda Alley, 700487 Iasi, Romania
| | - Marioara Nechifor
- Polyaddition and Photochemistry Department, "Petru Poni" Institute of Macromolecular Chemistry, 41A Gr. Ghica-Voda Alley, 700487 Iasi, Romania
| | - Carmen-Alice Teacă
- Center of Advanced Research in Bionanoconjugates and Biopolymers, "Petru Poni" Institute of Macromolecular Chemistry, 41A Gr. Ghica-Voda Alley, 700487 Iasi, Romania
| |
Collapse
|
22
|
Squyres GR, Newman DK. Biofilms as more than the sum of their parts: lessons from developmental biology. Curr Opin Microbiol 2024; 82:102537. [PMID: 39241276 PMCID: PMC12124288 DOI: 10.1016/j.mib.2024.102537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/09/2024] [Accepted: 08/11/2024] [Indexed: 09/09/2024]
Abstract
Although our understanding of both bacterial cell physiology and the complex behaviors exhibited by bacterial biofilms is expanding rapidly, we cannot yet sum the behaviors of individual cells to understand or predict biofilm behavior. This is both because cell physiology in biofilms is different from planktonic growth and because cell behavior in biofilms is spatiotemporally patterned. We use developmental biology as a guide to examine this phenotypic patterning, discussing candidate cues that may encode spatiotemporal information and possible roles for phenotypic patterning in biofilms. We consider other questions that arise from the comparison between biofilm and eukaryotic development, including what defines normal biofilm development and the nature of biofilm cell types and fates. We conclude by discussing what biofilm development can tell us about developmental processes, emphasizing the additional challenges faced by bacteria in biofilm development compared with their eukaryotic counterparts.
Collapse
Affiliation(s)
- Georgia R Squyres
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA 91125, USA
| | - Dianne K Newman
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA 91125, USA; Division of Geological and Planetary Sciences, Caltech, Pasadena, CA 91125, USA.
| |
Collapse
|
23
|
Lu R, Xia S, Deng G, Li W, Huang Z, Ling H. Constructing a nomogram for the recurrence of tibial osteomyelitis after debridement surgery based on platelet to lymphocyte ratio at admission: a dual-center retrospective study from China. BMC Musculoskelet Disord 2024; 25:967. [PMID: 39592998 PMCID: PMC11600715 DOI: 10.1186/s12891-024-08106-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/22/2024] [Indexed: 11/28/2024] Open
Abstract
AIM To construction of a nomogram to predict the prognosis of patients with tibial osteomyelitis after debridement surgery. METHOD This study used binary logistic regression analysis to identify clinical independent predictive factors, and then used R language to construct a nomogram. RESULT The results showed that WBC (3.36[1.77-6.37])、LYM (0.29[0.15-0.54])、BAS (2.19[1.22-3.91])、FIB (0.27[0.14-0.54])、ALB (0.16[0.09-0.3])、TBIL (0.41[0.21-0.83])、D-Dimer (1.77[1.03-3.03])、CRP (0.43[0.2-0.91])、ESR (3.08[1.6-5.79]) and PLR (0.47[0.24-0.9]) were independent predictive factors. Good prediction performance with modest errors was shown by the nomogram in both the training and validation groups. CONCLUSION WBC、LYM、BAS、FIB、ALB、TBIL、D-Dimer、CRP、ESR and PLR are independent predictors of the prognosis of patients with tibial osteomyelitis after debridement surgery. The constructed nomogram can help clinicians assess and treat patients early, benefiting more patients.
Collapse
Affiliation(s)
- Rongbin Lu
- Dept. Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Shaohuai Xia
- Dept. Neurotumor, Beijing Xiaotangshan Hospital, Beijing, 102211, China
| | - Gaoyong Deng
- Dept. Spine Surgery, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, China
| | - Wencai Li
- Dept. Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - Zhao Huang
- Dept. Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 ShuangYong Road, Nanning, Guangxi, 530022, China
| | - He Ling
- Dept. Orthopedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, NO. 6 ShuangYong Road, Nanning, Guangxi, 530022, China.
| |
Collapse
|
24
|
Oh JW, Shin MK, Park HR, Kim S, Lee B, Yoo JS, Chi WJ, Sung JS. PA-Win2: In Silico-Based Discovery of a Novel Peptide with Dual Antibacterial and Anti-Biofilm Activity. Antibiotics (Basel) 2024; 13:1113. [PMID: 39766503 PMCID: PMC11672609 DOI: 10.3390/antibiotics13121113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The emergence and prevalence of antibiotic-resistant bacteria (ARBs) have become a serious global threat, as the morbidity and mortality associated with ARB infections are continuously rising. The activation of quorum sensing (QS) genes can promote biofilm formation, which contributes to the acquisition of drug resistance and increases virulence. Therefore, there is an urgent need to develop new antimicrobial agents to control ARB and prevent further development. Antimicrobial peptides (AMPs) are naturally occurring defense molecules in organisms known to suppress pathogens through a broad range of antimicrobial mechanisms. Methods: In this study, we utilized a previously developed deep-learning model to identify AMP candidates from the venom gland transcriptome of the spider Pardosa astrigera, followed by experimental validation. Results: PA-Win2 was among the top-scoring predicted peptides and was selected based on physiochemical features. Subsequent experimental validation demonstrated that PA-Win2 inhibits the growth of Bacillus subtilis, Escherichia coli, Staphylococcus aureus, Staphylococcus epidermidis, Pseudomonas aeruginosa, and multidrug-resistant P. aeruginosa (MRPA) strain CCARM 2095. The peptide exhibited strong bactericidal activity against P. aeruginosa, and MRPA CCARM 2095 through the depolarization of bacterial cytoplasmic membranes and alteration of gene expression associated with bacterial survival. In addition, PA-Win2 effectively inhibited biofilm formation and degraded pre-formed biofilms of P. aeruginosa. The gene expression study showed that the peptide treatment led to the downregulation of QS genes in the Las, Pqs, and Rhl systems. Conclusions: These findings suggest PA-Win2 as a promising drug candidate against ARB and demonstrate the potential of in silico methods in discovering functional peptides from biological data.
Collapse
Affiliation(s)
- Jin Wook Oh
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Min Kyoung Shin
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Hye-Ran Park
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Sejun Kim
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| | - Byungjo Lee
- Research Institute, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Jung Sun Yoo
- Wildlife Quarantine Center, National Institute of Wildlife Disease Control and Prevention, Incheon 22382, Republic of Korea;
| | - Won-Jae Chi
- Species Diversity Research Division, National Institute of Biological Resources, Incheon 22689, Republic of Korea;
| | - Jung-Suk Sung
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (J.W.O.); (M.K.S.); (H.-R.P.); (S.K.)
| |
Collapse
|
25
|
Faghani-Eskandarkolaei P, Heli H, Akbari N, Koohi-Hosseinabadi O, Sari Aslani F, Sattarahmady N. Antibacterial and anti-biofilm activities of gold-curcumin nanohybrids and its polydopamine form upon photo-sonotherapy of Staphylococcus aureus infected implants: In vitro and animal model studies. Int J Biol Macromol 2024; 282:137430. [PMID: 39528199 DOI: 10.1016/j.ijbiomac.2024.137430] [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: 06/10/2024] [Revised: 10/27/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Implant-related infections are among the major post-surgery problems, and treatment of these infections is challenging due to the formation of biofilms by microorganisms such as Staphylococcus aureus. Herein, a novel gold-curcumin nanohybrid (GCNH) was synthesized for the first time and characterized. GCNH had a band gap energy of 2.41 eV, a zeta potential of -15 mV, and comprised uniform spherical particles with a mean diameter of 8 ± 2 nm. The biological macromolecule of polydopamine was then coated on GCNH to prepare a gold-curcumin-polydopamine nanohybrid (GCDNH). The nanohybrids were employed as novel dual photo-sonosensitizers for bacterial eradication by near-infrared (NIR) light and ultrasound (US) irradiations. GCNH and GCDNH represented photothermal conversion efficiencies of 26 and 32 %, respectively, and GCDNH represented a hemolysis rate of 2.3 % under both near-infrared (NIR) light and ultrasound (US) irradiations. NIR light and US irradiations (photo-sonotherapy) of Staphylococcus aureus using GCDNH depicted anti-bacterial and anti-biofilm efficiencies of 98 and 99 %, respectively, in synergistic manners, which are higher or as high as other sensitizers reported previously. The mechanism of photo-sonotherapy was related to generation of high levels of reactive oxygen species (ROS), and protein and nucleic acid leakages. In an in vivo infection model, NIR light and US irradiations annihilated Staphylococcus aureus on GCDNH-covered implants with high efficiency, without causing damage to normal tissues.
Collapse
Affiliation(s)
- P Faghani-Eskandarkolaei
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - H Heli
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - N Akbari
- Department of Microbiology, Faculty of Science, Arak Branch, Islamic Azad University, Arak, Iran
| | - O Koohi-Hosseinabadi
- Central Research Laboratory, Shiraz University of Medical Sciences, Shiraz, Iran
| | - F Sari Aslani
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - N Sattarahmady
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
26
|
Brahma U, Singothu S, Suresh A, Vemula D, Munagalasetty S, Sharma P, Bhandari V. MMV 1804559 is a potential antistaphylococcal and antibiofilm agent targeting the clfA gene of Staphylococcus aureus. J Appl Microbiol 2024; 135:lxae276. [PMID: 39474886 DOI: 10.1093/jambio/lxae276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024]
Abstract
AIMS Staphylococcus aureus, a high-priority pathogen proclaimed to cause infections ranging from mild to life-threatening, presents significant challenges in treatment. New therapies can be developed quicker using open drug discovery platforms offering a distinct approach to expedite the development of innovative antibacterial and anti-biofilm therapeutics. This study set out to address these issues by finding new uses for current medications to find compounds that are effective against S. aureus. METHODS AND RESULTS In this study, we screened the global priority health box, launched by Medicines for Malaria Ventures containing 240 compounds, for their effectiveness against S. aureus. MMV1795508, MMV1542799, MMV027331, MMV1593278, and MMV1804559 showed potential antibacterial activity at 10 µM concentration. These compounds underwent further evaluation for their ability to clear intracellular bacteria, disrupt biofilm formation, and eradicate existing biofilms. MMV1804559 demonstrated strong efficacy across all tested parameters, achieving 94% inhibition of intracellular bacteria, 79.19% disruption of biofilm cells, and 66.18% inhibition of biofilm formation. Scanning electron microscopy revealed notable membrane perforations and blebbing in MMV1804559-treated cells, indicating its impact on bacterial membranes. Gene expression analysis of cells treated with MMV1804559 showed downregulation of clfA and clfB genes, critical for biofilm formation. Additionally, docking studies confirmed the binding affinity of MMV1804559 with clfA, supported by favorable docking scores, MM/GBSA binding energy, and increased hydrogen bond interactions in the binding pocket, suggesting clfA as a target for MMV1804559. CONCLUSIONS MMV1804559 could serve as a potential therapy for S. aureus by targeting biofilm development and cell adhesion processes.
Collapse
Affiliation(s)
- Umarani Brahma
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Siva Singothu
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Akash Suresh
- National Institute of Animal Biotechnology (NIAB)-DBT, Gowlidoddi, Hyderabad-500049, Telangana, India
- Manipal Academy of Higher Education, Manipal 576104, India
| | - Divya Vemula
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Sharon Munagalasetty
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| | - Paresh Sharma
- National Institute of Animal Biotechnology (NIAB)-DBT, Gowlidoddi, Hyderabad-500049, Telangana, India
| | - Vasundhra Bhandari
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Balanagar, Kukatpally Industrial Area, Hyderabad- 500037, Telangana, India
| |
Collapse
|
27
|
Dou L, Wang X, Bai Y, Li Q, Luo L, Yu W, Wang Z, Wen K, Shen J. Mussel-like polydopamine-assisted aggregation-induced emission nanodot for enhanced broad-spectrum antimicrobial activity: In vitro and in vivo validation. Int J Biol Macromol 2024; 282:136762. [PMID: 39486741 DOI: 10.1016/j.ijbiomac.2024.136762] [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: 06/11/2024] [Revised: 09/22/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Emerging luminogens with aggregation-induced emission properties, namely AIEgens, demonstrated excellent anti-bacteria activity potential. However, their application still limited by the low antibacterial activity caused by the poor binding with bacteria. Polydopamine (PDA), an important biological macromolecule, possesses superior adhesion ability toward various material surface, including bacteria. In this study, the novel mussel-like PDA-assisted AIE Nanodot was proposed, achieving with robust bacterial binding ability and enhanced broad-spectrum antibacterial activity. Binding ability inherited from the PDA enables the constructed PDA-assisted AIE Nanodot to adhere efficiently to the bacterial membrane surface. Meanwhile, the AIE properties endowed them with monitoring capability, allowing for tracking their interaction with bacteria through facile fluorescence imaging in real time. More importantly, excellent killing of both Gram-positive and Gram-negative bacteria were successfully achieved in vitro antibacterial tests with excellent biocompatibility. Furthermore, in the treatment of Methicillin-resistant S. aureus (MRSA)-infected mouse-wound model, the mice exhibited accelerated wound healing with low bacterial load. This novel integrated strategy introduced a simple but effectively design to enhance the binding and antibacterial ability of AIEgens and would diversify the existing pool of antibacterial agents.
Collapse
Affiliation(s)
- Leina Dou
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China; College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shanxi, China
| | - Xiaonan Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Yuchen Bai
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Qing Li
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Liang Luo
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Wenbo Yu
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Zhanhui Wang
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China
| | - Kai Wen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| | - Jianzhong Shen
- National Key Laboratory of Veterinary Public Health Security, College of Veterinary Medicine, China Agricultural University, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing Laboratory for Food Quality and Safety, Beijing 100193, China.
| |
Collapse
|
28
|
Sivori F, Cavallo I, Truglio M, Pelagalli L, Mariani V, Fabrizio G, Abril E, Santino I, Fradiani PA, Solmone M, Pimpinelli F, Toma L, Arcioni R, De Blasi RA, Di Domenico EG. Biofilm-mediated antibiotic tolerance in Staphylococcus aureus from spinal cord stimulation device-related infections. Microbiol Spectr 2024; 12:e0168324. [PMID: 39470274 PMCID: PMC11619394 DOI: 10.1128/spectrum.01683-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Staphylococcus aureus is a predominant cause of infections in individuals with spinal cord stimulation (SCS) devices. Biofilm formation complicates these infections, commonly requiring both surgical and antibiotic treatments. This study explored the biofilm matrix composition and antimicrobial susceptibility of planktonic and biofilm-growing S. aureus isolates from individuals with SCS-related infections. Whole-genome sequencing (WGS) examined genotypes, virulome, resistome, and the pan-genome structure. The study also analyzed biofilm matrix composition, early surface adhesion, hemolytic activity, and antibiotic-susceptibility testing. WGS revealed genetic diversity among isolates. One isolate, though oxacillin susceptible, contained the mecA gene. The median number of virulence factor genes per isolate was 58. All isolates harbored the biofilm-related icaA/D genes. When assessing phenotypic characteristics, all strains demonstrated the ability to form biofilms in vitro. The antimicrobial susceptibility profile indicated that oxacillin, rifampin, and teicoplanin showed the highest efficacy against S. aureus biofilm. Conversely, high biofilm tolerance was observed for vancomycin, trimethoprim/sulfamethoxazole, and levofloxacin. These findings suggest that S. aureus isolates are highly virulent and produce robust biofilms. In cases of suspected biofilm infections caused by S. aureus, vancomycin should not be the primary choice due to its low activity against biofilm. Instead, oxacillin, rifampin, and teicoplanin appear to be more effective options to manage SCS infections.IMPORTANCESCS devices are increasingly used to manage chronic pain, but infections associated with these devices, particularly those caused by Staphylococcus aureus, present significant clinical challenges. These infections are often complicated by biofilm formation, which protects bacteria from immune responses and antibiotic treatments, making them difficult to eradicate. Understanding the genetic diversity, virulence, and biofilm characteristics of S. aureus isolates from SCS infections is critical to improving treatment strategies. Our study highlights the need to reconsider commonly used antibiotics like vancomycin, which shows reduced activity against biofilm-growing cells. Identifying more effective alternatives, such as oxacillin, rifampin, and teicoplanin, provides valuable insight for clinicians when managing biofilm-related S. aureus infections in patients with SCS implants. This research contributes to the growing evidence that biofilm formation is crucial in treating device-related infections, emphasizing the importance of tailoring antimicrobial strategies to the biofilm phenotype.
Collapse
Affiliation(s)
- Francesca Sivori
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Ilaria Cavallo
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Mauro Truglio
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Lorella Pelagalli
- Sultan Qaboos Comprehensive Cancer Care and Research Centre (SQCCCR), Mascate, Oman
| | - Valerio Mariani
- Dipartimento di Scienze Medico-Chirurgiche e Medicina Traslazionale, Sapienza University, Rome, Italy
| | - Giorgia Fabrizio
- Department of Biology and Biotechnology "C. Darwin", Sapienza University, Rome, Italy
| | - Elva Abril
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Iolanda Santino
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University, Microbiology Unit, Sant'Andrea Hospital, Rome, Italy
| | | | | | - Fulvia Pimpinelli
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Luigi Toma
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute IRCCS, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| | - Roberto Arcioni
- Sultan Qaboos Comprehensive Cancer Care and Research Centre (SQCCCR), Mascate, Oman
| | - Roberto Alberto De Blasi
- Dipartimento di Scienze Medico-Chirurgiche e Medicina Traslazionale, Sapienza University, Rome, Italy
| | - Enea Gino Di Domenico
- Microbiology and Virology Unit, San Gallicano Dermatological Institute, IRCCS, Istituti Fisioterapici Ospitalieri (IFO), Rome, Italy
| |
Collapse
|
29
|
Yun S, Min J, Han S, Sim HS, Kim SK, Lee JB, Yoon JW, Yeom J, Park W. Experimental evolution under different nutritional conditions changes the genomic architecture and virulence of Acinetobacter baumannii. Commun Biol 2024; 7:1274. [PMID: 39369115 PMCID: PMC11455985 DOI: 10.1038/s42003-024-06978-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
This study uncovers the molecular processes governing the adaptive evolution of multidrug-resistant (MDR) pathogens without antibiotic pressure. Genomic analysis of MDR Acinetobacter baumannii cells cultured for 8000 generations under starvation conditions (EAB1) or nutrient-rich conditions (EAB2) revealed significant genomic changes, primarily by insertion sequence (IS)-mediated insertions and deletions. Only two Acinetobacter-specific prophage-related deletions and translocations were observed in the EAB1 strain. Both evolved strains exhibited higher virulence in mouse infection studies, each with different modes of action. The EAB1 strain displayed a heightened ability to cross the epithelial barrier of human lung tissue, evade the immune system, and spread to lung tissues, ultimately resulting in cellular mortality. In contrast, the EAB2 strain strongly attached to epithelial cells, leading to increased synthesis of proinflammatory cytokines and chemokines. The genomic alterations and increased virulence observed in evolved strains during short-term evolution underscore the need for caution when handling these pathogens, as these risks persist even without antibiotic exposure.
Collapse
Affiliation(s)
- Sohyeon Yun
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea
| | - Jihyeon Min
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea
| | - Sunyong Han
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea
| | - Ho Seok Sim
- Department of Microbiology and Immunology, Department of Biomedical Science, and Cancer Research Institute, College of Medicine, Seoul National University, Jongno-gu, Seoul, Republic of Korea
| | - Se Kye Kim
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jun Bong Lee
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jang Won Yoon
- College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, Gangwon, Republic of Korea
| | - Jinki Yeom
- Department of Microbiology and Immunology, Department of Biomedical Science, and Cancer Research Institute, College of Medicine, Seoul National University, Jongno-gu, Seoul, Republic of Korea.
| | - Woojun Park
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Science and Ecological Engineering, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Dhadwal S, Handa S, Chatterjee M, Banat IM. Sophorolipid: An Effective Biomolecule for Targeting Microbial Biofilms. Curr Microbiol 2024; 81:388. [PMID: 39367190 DOI: 10.1007/s00284-024-03892-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Biofilms are microbial aggregates encased in a matrix that is attached to biological or nonbiological surfaces and constitute serious problems in food, medical, and marine industries and can have major negative effects on both health and the economy. Biofilm's complex microbial community provides a resistant environment that is difficult to eradicate and is extremely resilient to antibiotics and sanitizers. There are various conventional techniques for combating biofilms, including, chemical removal, physical or mechanical removal, use of antibiotics and disinfectants to destroy biofilm producing organisms. In contrast to free living planktonic cells, biofilms are very resistant to these methods. Hence, new strategies that differ from traditional approaches are urgently required. Microbial world offers a wide range of effective "green" compounds such as biosurfactants. They outperform synthetic surfactants in terms of biodegradability, superior stabilization, and reduced toxicity concerns. They also have better antiadhesive and anti-biofilm capabilities which can be used to treat biofilm-related problems. Sophorolipids (SLs) are a major type of biosurfactants that have gained immense interest in the healthcare industries because of their antiadhesive and anti-biofilm properties. Sophorolipids may therefore prove to be attractive substances that can be used in biomedical applications as adjuvant to other antibiotics against some infections through growth inhibition and/or biofilm disruption.
Collapse
Affiliation(s)
- Sunidhi Dhadwal
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India
| | - Shristi Handa
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India
| | - Mary Chatterjee
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh, 160014, India.
| | - Ibrahim M Banat
- Faculty of Life & Health Sciences, University of Ulster, Coleraine, BT52 1SA, UK.
| |
Collapse
|
31
|
Chang J, Kerr D, Zheng M, Seyler T. Chondrocyte Invasion May Be a Mechanism for Persistent Staphylococcus Aureus Infection In Vitro. Clin Orthop Relat Res 2024; 482:1839-1847. [PMID: 38662927 PMCID: PMC11419450 DOI: 10.1097/corr.0000000000003074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Recurrent bone and joint infection with Staphylococcus aureus is common. S. aureus can invade and persist in osteoblasts and fibroblasts, but little is known about this mechanism in chondrocytes. If S. aureus were able to invade and persist within chondrocytes, this could be a difficult compartment to treat. QUESTION/PURPOSE Can S. aureus infiltrate and persist intracellularly within chondrocytes in vitro? METHODS Cell lines were cultured in vitro and infected with S. aureus. Human chondrocytes (C20A4) were compared with positive controls of human osteoblasts (MG63) and mouse fibroblasts (NIH3T3), which have previously demonstrated S. aureus invasion and persistence (human fibroblasts were not available to us). Six replicates per cell type were followed for 6 days after infection. Cells were treated daily with antibiotic media for extracellular killing. To determine whether S. aureus can infiltrate chondrocytes, fluorescence microscopy was performed to qualitatively assess the presence of intracellular bacteria, and intracellular colony-forming units (CFU) were enumerated 2 hours after infection. To determine whether S. aureus can persist within chondrocytes, intracellular CFUs were enumerated from infected host cells each day postinfection. RESULTS S. aureus invaded human chondrocytes (C20A4) at a level (2.8 x 10 5 ± 5.5 x 10 4 CFUs/mL) greater than positive controls of human osteoblasts (MG63) (9.5 x 10 2 ± 2.5 x 10 2 CFUs/mL; p = 0.01) and mouse fibroblasts (NIH3T3) (9.1 x 10 4 ± 2.5 x 10 4 CFUs/mL; p = 0.02). S. aureus also persisted within human chondrocytes (C20A4) for 6 days at a level (1.4 x 10 3 ± 5.3 x 10 2 CFUs/mL) greater than that of human osteoblasts (MG63) (4.3 x 10 2 ± 3.5 x 10 1 CFUs/mL; p = 0.02) and mouse fibroblasts (NIH3T3) (0 CFUs/mL; p < 0.01). S. aureus was undetectable within mouse fibroblasts (NIH3T3) after 4 days. There were 0 CFUs yielded from cell media, confirming extracellular antibiotic treatment was effective. CONCLUSION S. aureus readily invaded human chondrocytes (C20A4) in vitro and persisted viably for 6 days after infection, evading extracellular antibiotics. Chondrocytes demonstrated a greater level of intracellular invasion and persistence by S. aureus than positive control human osteoblast (MG63) and mouse fibroblast (NIH3T3) cell lines. CLINICAL RELEVANCE Chondrocyte invasion and persistence may contribute to recurrent bone and joint infections. Additional research should assess longer periods of persistence and whether this mechanism is present in vivo.
Collapse
Affiliation(s)
- Jerry Chang
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - David Kerr
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Megan Zheng
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Thorsten Seyler
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| |
Collapse
|
32
|
Alduaij OK, Hussein RK, Abu Alrub S, Zidan SAH. Antimicrobial activities of Diltiazem Hydrochloride: drug repurposing approach. PeerJ 2024; 12:e17809. [PMID: 39329140 PMCID: PMC11426324 DOI: 10.7717/peerj.17809] [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: 02/27/2024] [Accepted: 07/03/2024] [Indexed: 09/28/2024] Open
Abstract
Background The growing concern of antibiotic-resistant microbial strains worldwide has prompted the need for alternative methods to combat microbial resistance. Biofilm formation poses a significant challenge to antibiotic efficiency due to the difficulty of penetrating antibiotics through the sticky microbial aggregates. Drug repurposing is an innovative technique that aims to expand the use of non-antibiotic medications to address this issue. The primary objective of this study was to evaluate the antimicrobial properties of Diltiazem HCl, a 1,5-benzothiazepine Ca2 + channel blocker commonly used as an antihypertensive agent, against four pathogenic bacteria and three pathogenic yeasts, as well as its antiviral activity against the Coxsackie B4 virus (CoxB4). Methods To assess the antifungal and antibacterial activities of Diltiazem HCl, the well diffusion method was employed, while crystal violet staining was used to determine the anti-biofilm activity. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) colorimetric assay was utilized to evaluate the antiviral activity of Diltiazem HCl against the CoxB4 virus. Results This study revealed that Diltiazem HCl exhibited noticeable antimicrobial properties against Gram-positive bacteria, demonstrating the highest inhibition of Staphylococcus epidermidis, followed by Staphylococcus aureus. It effectively reduced the formation of biofilms by 95.1% and 90.7% for S. epidermidis, and S. aureus, respectively. Additionally, the antiviral activity of Diltiazem HCl was found to be potent against the CoxB4 virus, with an IC50 of 35.8 ± 0.54 μg mL-1 compared to the reference antiviral Acyclovir (IC50 42.71 ± 0.43 μg mL-1). Conclusion This study suggests that Diltiazem HCl, in addition to its antihypertensive effect, may also be a potential treatment option for infections caused by Gram-positive bacteria and the CoxB4 viruses, providing an additional off-target effect for Diltiazem HCl.
Collapse
Affiliation(s)
- Omar K. Alduaij
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Rageh K. Hussein
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sharif Abu Alrub
- Department of Physics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sabry A. H. Zidan
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Assiut-Branch, Assiut, Egypt
| |
Collapse
|
33
|
Bucher MJ, Czyż DM. Phage against the Machine: The SIE-ence of Superinfection Exclusion. Viruses 2024; 16:1348. [PMID: 39339825 PMCID: PMC11436027 DOI: 10.3390/v16091348] [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: 05/28/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Prophages can alter their bacterial hosts to prevent other phages from infecting the same cell, a mechanism known as superinfection exclusion (SIE). Such alterations are facilitated by phage interactions with critical bacterial components involved in motility, adhesion, biofilm production, conjugation, antimicrobial resistance, and immune evasion. Therefore, the impact of SIE extends beyond the immediate defense against superinfection, influencing the overall fitness and virulence of the bacteria. Evaluating the interactions between phages and their bacterial targets is critical for leading phage therapy candidates like Pseudomonas aeruginosa, a Gram-negative bacterium responsible for persistent and antibiotic-resistant opportunistic infections. However, comprehensive literature on the mechanisms underlying SIE remains scarce. Here, we provide a compilation of well-characterized and potential mechanisms employed by Pseudomonas phages to establish SIE. We hypothesize that the fitness costs imposed by SIE affect bacterial virulence, highlighting the potential role of this mechanism in the management of bacterial infections.
Collapse
Affiliation(s)
- Michael J Bucher
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Daniel M Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
34
|
Motta EVS, de Jong TK, Gage A, Edwards JA, Moran NA. Glyphosate effects on growth and biofilm formation in bee gut symbionts and diverse associated bacteria. Appl Environ Microbiol 2024; 90:e0051524. [PMID: 39012136 PMCID: PMC11337805 DOI: 10.1128/aem.00515-24] [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: 03/20/2024] [Accepted: 06/22/2024] [Indexed: 07/17/2024] Open
Abstract
Biofilm formation is a common adaptation enabling bacteria to thrive in various environments and withstand external pressures. In the context of host-microbe interactions, biofilms play vital roles in establishing microbiomes associated with animals and plants and are used by opportunistic microbes to facilitate survival within hosts. Investigating biofilm dynamics, composition, and responses to environmental stressors is crucial for understanding microbial community assembly and biofilm regulation in health and disease. In this study, we explore in vivo colonization and in vitro biofilm formation abilities of core members of the honey bee (Apis mellifera) gut microbiota. Additionally, we assess the impact of glyphosate, a widely used herbicide with antimicrobial properties, and a glyphosate-based herbicide formulation on growth and biofilm formation in bee gut symbionts as well as in other biofilm-forming bacteria associated with diverse animals and plants. Our results demonstrate that several strains of core bee gut bacterial species can colonize the bee gut, which probably depends on their ability to form biofilms. Furthermore, glyphosate exposure elicits variable effects on bacterial growth and biofilm formation. In some instances, the effects correlate with the bacteria's ability to encode a susceptible or tolerant version of the enzyme inhibited by glyphosate in the shikimate pathway. However, in other instances, no such correlation is observed. Testing the herbicide formulation further complicates comparisons, as results often diverge from glyphosate exposure alone, suggesting that co-formulants influence bacterial growth and biofilm formation. These findings highlight the nuanced impacts of environmental stressors on microbial biofilms, with both ecological and host health-related implications. IMPORTANCE Biofilms are essential for microbial communities to establish and thrive in diverse environments. In the honey bee gut, the core microbiota member Snodgrassella alvi forms biofilms, potentially aiding the establishment of other members and promoting interactions with the host. In this study, we show that specific strains of other core members, including Bifidobacterium, Bombilactobacillus, Gilliamella, and Lactobacillus, also form biofilms in vitro. We then examine the impact of glyphosate, a widely used herbicide that can disrupt the bee microbiota, on bacterial growth and biofilm formation. Our findings demonstrate the diverse effects of glyphosate on biofilm formation, ranging from inhibition to enhancement, reflecting observations in other beneficial or pathogenic bacteria associated with animals and plants. Thus, glyphosate exposure may influence bacterial growth and biofilm formation, potentially shaping microbial establishment on host surfaces and impacting health outcomes.
Collapse
Affiliation(s)
- Erick V. S. Motta
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
- Department of Entomology, Texas A&M University, College Station, Texas, USA
| | - Tyler K. de Jong
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Alejandra Gage
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Joseph A. Edwards
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, Texas, USA
| | - Nancy A. Moran
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
35
|
Zacher AT, Mirza K, Thieme L, Nietzsche S, Senft C, Schwarz F. Biofilm formation of Staphylococcus aureus on various implants used for surgical treatment of destructive spondylodiscitis. Sci Rep 2024; 14:19364. [PMID: 39169088 PMCID: PMC11339328 DOI: 10.1038/s41598-024-70244-6] [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: 12/02/2023] [Accepted: 08/14/2024] [Indexed: 08/23/2024] Open
Abstract
The incidence of spondylodiscitis has witnessed a significant increase in recent decades. Surgical intervention becomes necessary in case of bone destruction to remove infected tissue and restore spinal stability, often involving the implantation of a cage. Despite appropriate treatment, relapses occur in up to 20 percent of cases, resulting in substantial economic and social burdens. The formation of biofilm has been identified as a major contributor to relapse development. Currently, there is no consensus among German-speaking spinal surgeons or in the existing literature regarding the preferred choice of material to minimize relapse rates. Thus, the objective of this study is to investigate whether certain materials used in spinal implants exhibit varying degrees of susceptibility to bacterial attachment, thereby providing valuable insights for improving treatment outcomes.Eight cages of each PEEK, titanium-coated PEEK (Ti-PEEK), titanium (Ti), polyetherketoneketone (PEKK), tantalum (Ta) and antibiotic-loaded bone cement were incubated with 20% human plasma for 24 h. Subsequently, four implants were incubated with S. aureus for 24 h or 48 h each. The biofilm was then removed by sonication and the attained solution plated for Colony Forming Units (CFU) counting. Scanning electron microscopy was used to confirm bacterial attachment. The CFUs have been compared directly and in relation to the cages surface area. The surface area of the implants was PEEK 557 mm2, Ti-PEEK 472 mm2, Ti 985 mm2, PEKK 594 mm2, Ta 706 mm2, bone cement 123 mm2. The mean CFU count per implant and per mm2 surface area after 24 h and after 48 h was calculated. Bone cement was found to have significantly more CFUs per mm2 surface area than the other materials tested. When comparing the CFU count per implant, bone cement was statistically significantly more prone to biofilm formation than PEEK after 48 h. There was no statistical significance between the other materials when comparing both CFU count per mm2 surface area and CFU count per implant. The electron microscopic analysis showed the attachment of the bacteria, as well as production of extracellular polymeric substances (EPS) as a sign for beginning biofilm formation. Antibiotic-loaded bone cement has shown statistically significantly more bacterial attachment than the other examined materials. No difference was found between the other materials regarding bacterial attachment after 24 h and 48 h. Proposed hypotheses for further studies include testing whether differences become apparent after longer incubation or with different pathogens involved in the pathogenesis of pyogenic spondylodiscitis.
Collapse
Affiliation(s)
- Amrei T Zacher
- Department of Neurosurgery, Jena University Hospital, Friedrich-Schiller-University of Jena, Am Klinikum 1, 07747, Jena, Germany.
| | - Kamran Mirza
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Friedrich-Schiller-University of Jena, Erlanger Allee 103, 07747, Jena, Germany
- Leibnitz Center for Photonics in Infection Research, Jena University Hospital, Friedrich- Schiller-University of Jena, Erlanger Allee 103, 07747, Jena, Germany
| | - Lara Thieme
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Friedrich-Schiller-University of Jena, Erlanger Allee 103, 07747, Jena, Germany
- Leibnitz Center for Photonics in Infection Research, Jena University Hospital, Friedrich- Schiller-University of Jena, Erlanger Allee 103, 07747, Jena, Germany
| | - Sandor Nietzsche
- Centre for Electron Microscopy, Jena University Hospital, Friedrich-Schiller-University of Jena, Ziegelmühlenweg 1, 07743, Jena, Germany
| | - Christian Senft
- Department of Neurosurgery, Jena University Hospital, Friedrich-Schiller-University of Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Falko Schwarz
- Department of Neurosurgery, Jena University Hospital, Friedrich-Schiller-University of Jena, Am Klinikum 1, 07747, Jena, Germany
| |
Collapse
|
36
|
Xiang Q, Stryhanyuk H, Schmidt M, Kümmel S, Richnow HH, Zhu YG, Cui L, Musat N. Stable isotopes and nanoSIMS single-cell imaging reveals soil plastisphere colonizers able to assimilate sulfamethoxazole. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 355:124197. [PMID: 38782163 DOI: 10.1016/j.envpol.2024.124197] [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: 08/28/2023] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
The presence and accumulation of both, plastics and antibiotics in soils may lead to the colonization, selection, and propagation of soil bacteria with certain metabolic traits, e.g., antibiotic resistance, in the plastisphere. However, the impact of plastic-antibiotic tandem on the soil ecosystem functioning, particularly on microbial function and metabolism remains currently unexplored. Herein, we investigated the competence of soil bacteria to colonize plastics and degrade 13C-labeled sulfamethoxazole (SMX). Using single-cell imaging, isotope tracers, soil respiration and SMX mineralization bulk measurements we show that microbial colonization of polyethylene (PE) and polystyrene (PS) surfaces takes place within the first 30 days of incubation. Morphologically diverse microorganisms were colonizing both plastic types, with a slight preference for PE substrate. CARD-FISH bacterial cell counts on PE and PS surfaces formed under SMX amendment ranged from 5.36 × 103 to 2.06 × 104, and 2.06 × 103 to 3.43 × 103 hybridized cells mm-2, respectively. Nano-scale Secondary Ion Mass Spectrometry measurements show that 13C enrichment was highest at 130 days with values up to 1.29 atom%, similar to those of the 13CO2 pool (up to 1.26 atom%, or 22.55 ‰). Independent Mann-Whitney U test showed a significant difference between the control plastisphere samples incubated without SMX and those in 13C-SMX incubations (P < 0.001). Our results provide direct evidence demonstrating, at single-cell level, the capacity of bacterial colonizers of plastics to assimilate 13C-SMX from contaminated soils. These findings expand our knowledge on the role of soil-seeded plastisphere microbiota in the ecological functioning of soils impacted by anthropogenic stressors.
Collapse
Affiliation(s)
- Qian Xiang
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China; Department of Isotope Biochemistry, Currently Merged As Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research-UFZ, 04318, Leipzig, Germany
| | - Hryhoriy Stryhanyuk
- Department of Isotope Biochemistry, Currently Merged As Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research-UFZ, 04318, Leipzig, Germany
| | - Matthias Schmidt
- Department of Isotope Biochemistry, Currently Merged As Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research-UFZ, 04318, Leipzig, Germany
| | - Steffen Kümmel
- Department of Isotope Biochemistry, Currently Merged As Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research-UFZ, 04318, Leipzig, Germany
| | - Hans H Richnow
- Department of Isotope Biochemistry, Currently Merged As Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research-UFZ, 04318, Leipzig, Germany
| | - Yong-Guan Zhu
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China; State Key Lab of Urban and Regional Ecology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Li Cui
- Key Lab of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, 361021, China
| | - Niculina Musat
- Department of Isotope Biochemistry, Currently Merged As Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research-UFZ, 04318, Leipzig, Germany; Department of Biology, Section for Microbiology, Aarhus University, 8000, Aarhus C, Denmark.
| |
Collapse
|
37
|
Uthra C, Nagaraj K, Wadaan MA, Karuppiah C, Maity P, Baabbad A, Kaliyaperumal R, Venkatachalapathy R, Shah F, Kumar P. Zinc and Copper Oxide Nanoparticles: Pioneering Antibacterial and Antibiofilm Strategies for Environmental Restoration against Antibiotic-Resistant Bacteria. MATERIALS (BASEL, SWITZERLAND) 2024; 17:3444. [PMID: 39063741 PMCID: PMC11278220 DOI: 10.3390/ma17143444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024]
Abstract
This study explores the challenge of antimicrobial resistance by investigating the utilization of zinc oxide (ZnO) and copper oxide (Cu2O) nanoparticles (NPs) to combat antibiotic-resistant bacteria in wastewater treatment plants (WWTPs). The synthesized metal oxide NPs underwent thorough characterization through various analytical techniques, confirming their nanoparticulate nature. Electronic absorption and X-ray diffraction (XRD) analyses revealed successful reduction processes and crystalline properties, respectively. Fourier transform infrared spectroscopy (FTIR) results indicated the stabilization of nanoparticles in solution. Scanning electron microscopy (SEM) observations revealed well-defined spherical and flower-like morphologies for the zinc and copper oxide nanoparticles, with sizes approximately ranging from 50 nm to 25 nm Notably, the synthesized nanoparticles exhibited heightened efficacy in impeding biofilm formation, with zinc oxide NPs displaying superior antibacterial activity compared to copper. These findings suggest the promising potential of these nanoparticles in controlling antibiotic-resistant organisms, even following WWTP treatment processes. This research contributes to the ongoing advancements in nanotechnology aimed at combating antibiotic resistance, offering new prospects for the development of effective wastewater treatment strategies.
Collapse
Affiliation(s)
- Chandrabose Uthra
- Department of Microbiology, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
| | - Karuppiah Nagaraj
- School of Pharmacy, National Forensic Sciences University, 6M56+XP8, Police Bhavan Rd, Sector 9, Gandhinagar 382007, Gujarat, India
| | - Mohammad Ahmad Wadaan
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11541, Saudi Arabia
| | - Chelladurai Karuppiah
- Battery Research Center of Green Energy, Ming Chi University of Technology, New Taipei City 243303, Taiwan
- Center of Molecular Medicine and Dianostics (COMManD), Saveetha Dental College and Hospitals, SIMTS, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Prasenjit Maity
- School of Environmental Technology, National Forensic Sciences University, 6M56+XP8, Police Bhavan Rd, Sector 9, Gandhinagar 382007, Gujarat, India
| | - Almohannad Baabbad
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11541, Saudi Arabia
| | - Raja Kaliyaperumal
- Department of Chemistry, St. Joseph University, Chumoukedima 797115, Nagaland, India
| | - Renuka Venkatachalapathy
- School of Environmental Technology, National Forensic Sciences University, 6M56+XP8, Police Bhavan Rd, Sector 9, Gandhinagar 382007, Gujarat, India
| | - Flora Shah
- School of Pharmacy, National Forensic Sciences University, 6M56+XP8, Police Bhavan Rd, Sector 9, Gandhinagar 382007, Gujarat, India
| | - Puneet Kumar
- School of Pharmacy, National Forensic Sciences University, 6M56+XP8, Police Bhavan Rd, Sector 9, Gandhinagar 382007, Gujarat, India
| |
Collapse
|
38
|
Caudal F, Roullier C, Rodrigues S, Dufour A, Artigaud S, Le Blay G, Bazire A, Petek S. Anti-Biofilm Extracts and Molecules from the Marine Environment. Mar Drugs 2024; 22:313. [PMID: 39057422 PMCID: PMC11278325 DOI: 10.3390/md22070313] [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: 05/28/2024] [Revised: 07/02/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Pathogenic bacteria and their biofilms are involved in many diseases and represent a major public health problem, including the development of antibiotic resistance. These biofilms are known to cause chronic infections for which conventional antibiotic treatments are often ineffective. The search for new molecules and innovative solutions to combat these pathogens and their biofilms has therefore become an urgent need. The use of molecules with anti-biofilm activity would be a potential solution to these problems. The marine world is rich in micro- and macro-organisms capable of producing secondary metabolites with original skeletons. An interest in the chemical strategies used by some of these organisms to regulate and/or protect themselves against pathogenic bacteria and their biofilms could lead to the development of bioinspired, eco-responsible solutions. Through this original review, we listed and sorted the various molecules and extracts from marine organisms that have been described in the literature as having strictly anti-biofilm activity, without bactericidal activity.
Collapse
Affiliation(s)
- Flore Caudal
- Laboratoire de Biotechnologie et Chimie Marines, Université Bretagne Sud, EMR CNRS 6076, IUEM, 56100 Lorient, France; (F.C.); (S.R.); (A.D.)
- IRD, Univ Brest, CNRS, Ifremer, LEMAR, IUEM, 29280 Plouzane, France; (S.A.); (G.L.B.)
| | - Catherine Roullier
- Institut des Substances et Organismes de la Mer, Nantes Université, ISOMER, UR 2160, 40000 Nantes, France;
| | - Sophie Rodrigues
- Laboratoire de Biotechnologie et Chimie Marines, Université Bretagne Sud, EMR CNRS 6076, IUEM, 56100 Lorient, France; (F.C.); (S.R.); (A.D.)
| | - Alain Dufour
- Laboratoire de Biotechnologie et Chimie Marines, Université Bretagne Sud, EMR CNRS 6076, IUEM, 56100 Lorient, France; (F.C.); (S.R.); (A.D.)
| | - Sébastien Artigaud
- IRD, Univ Brest, CNRS, Ifremer, LEMAR, IUEM, 29280 Plouzane, France; (S.A.); (G.L.B.)
| | - Gwenaelle Le Blay
- IRD, Univ Brest, CNRS, Ifremer, LEMAR, IUEM, 29280 Plouzane, France; (S.A.); (G.L.B.)
| | - Alexis Bazire
- Laboratoire de Biotechnologie et Chimie Marines, Université Bretagne Sud, EMR CNRS 6076, IUEM, 56100 Lorient, France; (F.C.); (S.R.); (A.D.)
| | - Sylvain Petek
- IRD, Univ Brest, CNRS, Ifremer, LEMAR, IUEM, 29280 Plouzane, France; (S.A.); (G.L.B.)
| |
Collapse
|
39
|
Salem S, Abdelsalam NA, Shata AH, Mouftah SF, Cobo-Díaz JF, Osama D, Atteya R, Elhadidy M. Unveiling the microevolution of antimicrobial resistance in selected Pseudomonas aeruginosa isolates from Egyptian healthcare settings: A genomic approach. Sci Rep 2024; 14:15500. [PMID: 38969684 PMCID: PMC11226647 DOI: 10.1038/s41598-024-65178-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: 05/09/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024] Open
Abstract
The incidence of Pseudomonas aeruginosa infections in healthcare environments, particularly in low-and middle-income countries, is on the rise. The purpose of this study was to provide comprehensive genomic insights into thirteen P. aeruginosa isolates obtained from Egyptian healthcare settings. Phenotypic analysis of the antimicrobial resistance profile and biofilm formation were performed using minimum inhibitory concentration and microtiter plate assay, respectively. Whole genome sequencing was employed to identify sequence typing, resistome, virulome, and mobile genetic elements. Our findings indicate that 92.3% of the isolates were classified as extensively drug-resistant, with 53.85% of these demonstrating strong biofilm production capabilities. The predominant clone observed in the study was ST773, followed by ST235, both of which were associated with the O11 serotype. Core genome multi-locus sequence typing comparison of these clones with global isolates suggested their potential global expansion and adaptation. A significant portion of the isolates harbored Col plasmids and various MGEs, all of which were linked to antimicrobial resistance genes. Single nucleotide polymorphisms in different genes were associated with the development of antimicrobial resistance in these isolates. In conclusion, this pilot study underscores the prevalence of extensively drug-resistant P. aeruginosa isolates and emphasizes the role of horizontal gene transfer facilitated by a diverse array of mobile genetic elements within various clones. Furthermore, specific insertion sequences and mutations were found to be associated with antibiotic resistance.
Collapse
Affiliation(s)
- 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 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
| | - Ahmed H Shata
- 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
| | - Shaimaa F Mouftah
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt
| | - José F Cobo-Díaz
- Department of Food Hygiene and Technology, Institute of Food Science and Technology, Universidad de León, León, Spain
| | - Dina Osama
- Department of Microbiology and Immunology, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA), Cairo, Egypt
| | - Reham Atteya
- Center for Genomics, Helmy Institute for Medical Sciences, Zewail City of Science and Technology, Giza, 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
|
40
|
Sorrentino PJ, MacArthur SL. Use of intranasal povidone-iodine packing in the management of infectious rhinosinusitis in three cats. JFMS Open Rep 2024; 10:20551169241275303. [PMID: 39502682 PMCID: PMC11536372 DOI: 10.1177/20551169241275303] [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: 11/08/2024] Open
Abstract
Case series summary Described are three cats diagnosed with rhinosinusitis secondary to Mycobacterium bouchedurhonense, Aspergillus species and Alternaria species, respectively. Medical records were retrospectively reviewed to identify cats with decreased nasal airflow and mucopurulent discharge that failed to improve on antibiotic therapy of 3 months or longer duration. Surgical debridement was followed by nasal packing using 5% povidone-iodine saturated umbilical tape, which was replaced at 24 h postoperatively. At 48 h postoperatively, the rhinotomy site was closed. Systemic therapy continued in the postoperative period. All cases were minimally responsive to previous medical management. History, signalment, clinical signs, diagnostic findings, treatment, and short- and long-term outcomes were retrieved. All cats were middle-aged with outdoor access and had clinical signs that commenced during the summer months. CT revealed turbinate destruction and soft tissue densities within the nasal passages. The otic apparatuses and calvaria were intact in all cats before surgery. A repeat CT examination revealed an improvement of the proliferative tissue identified in preoperative imaging in all cases. All cats achieved successful interruption of nasal discharge and restoration of nasal airflow with follow-up times of up to 16 months postoperatively. Relevance and novel information To the authors' knowledge, this is the first report of the use of intranasal povidone-iodine packing in cats for the management of infectious rhinosinusitis. Surgical debridement and intranasal packing in addition to systemic therapy were successful in restoring nasal airflow and resolving nasal discharge in all cats with long-term follow-up.
Collapse
|
41
|
Qi J, Zhang P, Zhang T, Zhang R, Zhang Q, Wang J, Zong M, Gong Y, Liu X, Wu X, Li B. Metal-doped carbon dots for biomedical applications: From design to implementation. Heliyon 2024; 10:e32133. [PMID: 38868052 PMCID: PMC11168406 DOI: 10.1016/j.heliyon.2024.e32133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/24/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024] Open
Abstract
Carbon dots (CDs), as a new kind of fluorescent nanomaterials, show great potential for application in several fields due to their unique nano-size effect, easy surface functionalization, controllable photoluminescence, and excellent biocompatibility. Conventional preparation methods for CDs typically involve top-down and bottom-up approaches. Doping is a major step forward in CDs design methodology. Chemical doping includes both non-metal and metal doping, in which non-metal doping is an effective strategy for modulating the fluorescence properties of CDs and improving photocatalytic performance in several areas. In recent years, Metal-doped CDs have aroused the interest of academics as a promising nano-doping technique. This approach has led to improvements in the physicochemical and optical properties of CDs by altering their electron density distribution and bandgap capacity. Additionally, the issues of metal toxicity and utilization have been addressed to a large extent. In this review, we categorize metals into two major groups: transition group metals and rare-earth group metals, and an overview of recent advances in biomedical applications of these two categories, respectively. Meanwhile, the prospects and the challenges of metal-doped CDs for biomedical applications are reviewed and concluded. The aim of this paper is to break through the existing deficiencies of metal-doped CDs and fully exploit their potential. I believe that this review will broaden the insight into the synthesis and biomedical applications of metal-doped CDs.
Collapse
Affiliation(s)
- Jin Qi
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Pengfei Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Tong Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Ran Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Qingmei Zhang
- Taiyuan University of Science and Technology, Taiyuan, 030024, Shanxi, China
| | - Jue Wang
- The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Mingrui Zong
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Yajuan Gong
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Xiaoming Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Xiuping Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001 Shanxi, China
| |
Collapse
|
42
|
Tabassum N, Khan F, Jeong GJ, Jo DM, Kim YM. Silver nanoparticles synthesized from Pseudomonas aeruginosa pyoverdine: Antibiofilm and antivirulence agents. Biofilm 2024; 7:100192. [PMID: 38544742 PMCID: PMC10966193 DOI: 10.1016/j.bioflm.2024.100192] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 06/16/2025] Open
Abstract
The increasing incidence of antimicrobial resistance exhibited by biofilm-forming microbial pathogens has been recognized as one of the major issues in the healthcare sector. In the present study, nanomaterial-based controlling the biofilm and virulence properties has been considered an alternative approach. Pyoverdine (PVD) isolated from the Pseudomonas aeruginosa was utilized as a biological corona to synthesize silver nanoparticles (AgNPs), which will be helpful in a targeted action to microbial pathogens due to the recognition of the corona of the nanoparticles by the pathogenic membrane. Synthesized PVD-AgNPs were spherical to irregular, with an average size value of 251.87 ± 21.8 nm and zeta potential with a value of -36.51 ± 0.69 mV. The MIC value of PVD-AgNPs towards P. aeruginosa, Listeria monocytogenes, Staphylococcus aureus, Streptococcus mutans, Escherichia coli, and Candida albicans in the standard and host-mimicking media were observed in decreasing order in a multi-fold, such as standard growth media > sputum > synthetic human urine > saliva. Both the initial stage and the well-established biofilms of these microbial pathogens have been effectively inhibited and eradicated by PVD-AgNPs. PVD-AgNPs increase the susceptibility of tetracycline, PVD, and amphotericin B towards established mature mono- and mixed-species biofilms of S. aureus and C. albicans. Additionally, PVD-AgNPs attenuate several virulence properties, such as inhibition of protease activity, motility, and PVD and pyocyanin production in P. aeruginosa. The inhibition of gene expression of biofilm and virulence-associated genes in P. aeruginosa validates its phenotypic effects.
Collapse
Affiliation(s)
- Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Institute of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Du-Min Jo
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| |
Collapse
|
43
|
Wu R, Kong L, Liu F. Regulation of biofilm gene expression by DNA replication in Bacillus subtilis. J Cell Mol Med 2024; 28:e18481. [PMID: 38899542 PMCID: PMC11187747 DOI: 10.1111/jcmm.18481] [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: 04/27/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Bacillus subtilis relies on biofilms for survival in harsh environments. Extracellular polymeric substance (EPS) is a crucial component of biofilms, yet the dynamics of EPS production in single cells remain elusive. To unveil the modulation of EPS synthesis, we built a minimal network model comprising the SinI-SinR-SlrR module, Spo0A, and EPS. Stochastic simulations revealed that antagonistic interplay between SinI and SinR enables EPS production in bursts. SlrR widens these bursts and increases their frequency by stabilizing SinR-SlrR complexes and depleting free SinR. DNA replication and chromosomal positioning of key genes dictate pulsatile changes in the slrR:sinR gene dosage ratio (gr) and Spo0A-P levels, each promoting EPS production in distinct phases of the cell cycle. As the cell cycle lengthens with nutrient stress, the duty cycle of gr pulsing decreases, whereas the amplitude of Spo0A-P pulses elevates. This coordinated response facilitates keeping a constant proportion of EPS-secreting cells within colonies across diverse nutrient conditions. Our results suggest that bacteria may 'encode' eps expression through strategic chromosomal organization. This work illuminates how stochastic protein interactions, gene copy number imbalance, and cell-cycle dynamics orchestrate EPS synthesis, offering a deeper understanding of biofilm formation.
Collapse
Affiliation(s)
- Renjie Wu
- National Laboratory of Solid State Microstructures, Department of Physics, Collaborative Innovation Center of Advanced Microstructures and Institute for Brain SciencesNanjing UniversityNanjingP. R. China
| | - Ling‐Xing Kong
- National Laboratory of Solid State Microstructures, Department of Physics, Collaborative Innovation Center of Advanced Microstructures and Institute for Brain SciencesNanjing UniversityNanjingP. R. China
| | - Feng Liu
- National Laboratory of Solid State Microstructures, Department of Physics, Collaborative Innovation Center of Advanced Microstructures and Institute for Brain SciencesNanjing UniversityNanjingP. R. China
| |
Collapse
|
44
|
Tabassum N, Jeong GJ, Jo DM, Khan F, Kim YM. Attenuation of biofilm and virulence factors of Pseudomonas aeruginosa by tetramethylpyrazine-gold nanoparticles. Microb Pathog 2024; 191:106658. [PMID: 38643850 DOI: 10.1016/j.micpath.2024.106658] [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: 01/16/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Pseudomonas aeruginosa is often identified as the causative agent in nosocomial infections. Their adapted resistance makes them strong towards antimicrobial treatments. They protect and empower their survival behind strong biofilm architecture that works as their armor toward antimicrobial therapy. Additionally, P. aeruginosa generates virulence factors, contributing to chronic infection and recalcitrant phenotypic characteristics. The current study utilizes the benevolence of nanotechnology to develop an alternate technique to control the spreading of P. aeruginosa by limiting its biofilm and virulence development. This study used a natural compound, tetramethylpyrazine, to generate gold nanoparticles. Tetramethylpyrazine-gold nanoparticles (Tet-AuNPs) were presented in spherical shapes, with an average size of 168 ± 52.49 nm and a zeta potential of -12.22 ± 2.06 mV. The minimum inhibition concentration (MIC) of Tet-AuNPs that proved more than 90 % effective in inhibiting P. aeruginosa was 256 μg/mL. Additionally, it also shows antibacterial activities against Staphylococcus aureus (MIC, 256 μg/mL), Streptococcus mutans (MIC, 128 μg/mL), Klebsiella pneumoniae (MIC, 128 μg/mL), Listeria monocytogenes (MIC, 256 μg/mL), and Escherichia coli (MIC, 256 μg/mL). The sub-MIC values of Tet-AuNPs significantly inhibited the early-stage biofilm formation of P. aeruginosa. Moreover, this concentration strongly affected hemolysis, protease activity, and different forms of motilities in P. aeruginosa. Additionally, Tet-AuNPs destroyed the well-established mature biofilm of P. aeruginosa. The expression of genes linked with the biofilm formation and virulence in P. aeruginosa treated with sub-MIC doses of Tet-AuNPs was shown to be significantly suppressed. Gene expression studies support biofilm- and virulence-suppressing effects of Tet-AuNPs at the phenotypic level.
Collapse
Affiliation(s)
- Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Geum-Jae Jeong
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea
| | - Du-Min Jo
- Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea; National Marine Biodiversity Institute of Korea, Seochun, Chungcheongnam-do, 33662, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Institute of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| | - Young-Mog Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan, 48513, Republic of Korea; Department of Food Science and Technology, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
45
|
Khosravi A, Chen Q, Echterhof A, Koff JL, Bollyky PL. Phage Therapy for Respiratory Infections: Opportunities and Challenges. Lung 2024; 202:223-232. [PMID: 38772946 PMCID: PMC11570333 DOI: 10.1007/s00408-024-00700-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/13/2024] [Indexed: 05/23/2024]
Abstract
We are entering the post-antibiotic era. Antimicrobial resistance (AMR) is a critical problem in chronic lung infections resulting in progressive respiratory failure and increased mortality. In the absence of emerging novel antibiotics to counter AMR infections, bacteriophages (phages), viruses that infect bacteria, have become a promising option for chronic respiratory infections. However, while personalized phage therapy is associated with improved outcomes in individual cases, clinical trials demonstrating treatment efficacy are lacking, limiting the therapeutic potential of this approach for respiratory infections. In this review, we address the current state of phage therapy for managing chronic respiratory diseases. We then discuss how phage therapy may address major microbiologic obstacles which hinder disease resolution of chronic lung infections with current antibiotic-based treatment practices. Finally, we highlight the challenges that must be addressed for successful phage therapy clinical trials. Through this discussion, we hope to expand on the potential of phages as an adjuvant therapy in chronic lung infections, as well as the microbiologic challenges that need to be addressed for phage therapy to expand beyond personalized salvage therapy.
Collapse
Affiliation(s)
- Arya Khosravi
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA.
- Division of Infectious Diseases, Department of Medicine, Stanford University, 279 Campus Drive, Beckman Center, Room B237, Stanford, CA, 94305, USA.
| | - Qingquan Chen
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| | - Arne Echterhof
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jonathan L Koff
- Section of Pulmonary, Critical Care & Sleep Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Paul L Bollyky
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
46
|
Kosmeri C, Giapros V, Serbis A, Balomenou F, Baltogianni M. Antibiofilm Strategies in Neonatal and Pediatric Infections. Antibiotics (Basel) 2024; 13:509. [PMID: 38927176 PMCID: PMC11200539 DOI: 10.3390/antibiotics13060509] [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: 04/28/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Biofilm-related infections pose significant challenges in neonatal and pediatric care, contributing to increased morbidity and mortality rates. These complex microbial communities, comprising bacteria and fungi, exhibit resilience against antibiotics and host immune responses. Bacterial species such as Enterococcus faecalis, Pseudomonas aeruginosa, Staphylococcus aureus, and Staphylococcus epidermidis commonly form biofilms on medical devices, exacerbating infection risks. Neonates and children, particularly those in intensive care units, are highly susceptible to biofilm-associated infections due to the prolonged use of invasive devices, such as central lines and endotracheal tubes. Enteral feeding tubes, crucial for neonatal nutritional support, also serve as potential sites for biofilm formation, contributing to recurrent microbial contamination. Moreover, Candida species, including Candida pelliculosa, present emerging challenges in neonatal care, with multi-drug resistant strains posing treatment complexities. Current antimicrobial therapies, while important in managing infections, often fall short in eradicating biofilms, necessitating alternative strategies. The aim of this review is to summarize current knowledge regarding antibiofilm strategies in neonates and in children. Novel approaches focusing on biofilm inhibition and dispersal show promise, including surface modifications, matrix-degrading enzymes, and quorum-sensing inhibitors. Prudent use of medical devices and exploration of innovative antibiofilm therapies are imperative in mitigating neonatal and pediatric biofilm infections.
Collapse
Affiliation(s)
- Chrysoula Kosmeri
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece; (C.K.); (A.S.)
| | - Vasileios Giapros
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| | - Anastasios Serbis
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece; (C.K.); (A.S.)
| | - Foteini Balomenou
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| | - Maria Baltogianni
- Neonatal Intensive Care Unit, School of Medicine, University of Ioannina, 45110 Ioannina, Greece; (F.B.); (M.B.)
| |
Collapse
|
47
|
Taşkın Kafa AH, Aslan R, Durna Daştan S, Çeli K C, Hasbek M, Emi Noğlu A. Molecular diversity of Klebsiella pneumoniae clinical isolates: antimicrobial resistance, virulence, and biofilm formation. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 44:361-377. [PMID: 38718417 DOI: 10.1080/15257770.2024.2344741] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 03/21/2024] [Accepted: 04/09/2024] [Indexed: 04/01/2025]
Abstract
One of the mechanisms responsible for antibiotic resistance in Klebsiella pneumoniae is the enzymes produced by the bacteria; another important mechanism is the ability to form biofilm. In this study, antibiotic resistance, genes associated with virulence, and biofilm-forming properties of K. pneumoniae strains were investigated. A total of 100 K. pneumoniae isolates were obtained from different clinical samples identified by Matrix-Assisted Laser Desorption/Ionization time-of-flight Mass Spectrometry. Antimicrobial susceptibility testing was performed with the Phoenix 100 apparatus. The biofilm forming properties of strains were determined by the microtiter plate method. For molecular analysis, genes encoding the carbapenemase enzyme (blaOXA-48, blaNDM-1, blaIMP, and blaVIM) and biofilm-related genes (treC, luxS, mrkA, and wza) were investigated by polymerase chain reaction (PCR). While 76% of clinical isolates were resistant to three or more antimicrobials, 24% were classified as non-multidrug resistant (non-MDR). When biofilm-forming capacities of clinical isolates were tested, it was determined that the resistant-isolates produced 59.2% strong biofilm, and susceptible-isolates produced 12.5% strong biofilm. According to PCR results, carbapenemase genes were determined as follows: blaOXA-48-70%, blaNDM-49%, and blaKPC-19%, blaOXA-48/blaNDM/blaKPC-12%, blaOXA-48/blaNDM-26%, and blaOXA-48/blaKPC-4%. The biofilm-associated genes in bacterial isolates were determined as follows: luxS-98%, treC-94%, mrkA-88%, and wza-15%. In addition, Hierarchical Clustering Tree and Heatmap analysis revealed an association between isolates that lacks resistance genes and isolates lacks biofilm-formation related genes that were included in MDR or non-MDR classes. As a result, biofilm should be considered in the treatment of MDR infections, and therapy should be planned accordingly. In addition, pursuing the data and genes of antibiotic resistance is significant for combating resistance.
Collapse
Affiliation(s)
- Ayşe Hümeyra Taşkın Kafa
- Department of Medical Microbiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Rukiye Aslan
- Department of Medical Microbiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
- Department of Medical Services and Techniques, Vocational School of Health Services, Sivas Cumhuriyet University, Sivas, Turkey
| | - Sevgi Durna Daştan
- Department of Biology, Faculty of Science, Sivas Cumhuriyet University, Sivas, Turkey
| | - Cem Çeli K
- Department of Medical Microbiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Mürşit Hasbek
- Department of Medical Microbiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ayşenur Emi Noğlu
- Department of Biology, Faculty of Art and Science, Recep Tayyip Erdogan University, Rize, Turkey
| |
Collapse
|
48
|
Jarman E, Burgess J, Sharma A, Hayashigatani K, Singh A, Fox P. Human-Derived collagen hydrogel as an antibiotic vehicle for topical treatment of bacterial biofilms. PLoS One 2024; 19:e0303039. [PMID: 38701045 PMCID: PMC11068178 DOI: 10.1371/journal.pone.0303039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 04/13/2024] [Indexed: 05/05/2024] Open
Abstract
The complexity of chronic wounds creates difficulty in effective treatments, leading to prolonged care and significant morbidity. Additionally, these wounds are incredibly prone to bacterial biofilm development, further complicating treatment. The current standard treatment of colonized superficial wounds, debridement with intermittent systemic antibiotics, can lead to systemic side-effects and often fails to directly target the bacterial biofilm. Furthermore, standard of care dressings do not directly provide adequate antimicrobial properties. This study aims to assess the capacity of human-derived collagen hydrogel to provide sustained antibiotic release to disrupt bacterial biofilms and decrease bacterial load while maintaining host cell viability and scaffold integrity. Human collagen harvested from flexor tendons underwent processing to yield a gellable liquid, and subsequently was combined with varying concentrations of gentamicin (50-500 mg/L) or clindamycin (10-100 mg/L). The elution kinetics of antibiotics from the hydrogel were analyzed using liquid chromatography-mass spectrometry. The gel was used to topically treat Methicillin-resistant Staphylococcus aureus (MRSA) and Clostridium perfringens in established Kirby-Bauer and Crystal Violet models to assess the efficacy of bacterial inhibition. 2D mammalian cell monolayers were topically treated, and cell death was quantified to assess cytotoxicity. Bacteria-enhanced in vitro scratch assays were treated with antibiotic-embedded hydrogel and imaged over time to assess cell death and mobility. Collagen hydrogel embedded with antibiotics (cHG+abx) demonstrated sustained antibiotic release for up to 48 hours with successful inhibition of both MRSA and C. perfringens biofilms, while remaining bioactive up to 72 hours. Administration of cHG+abx with antibiotic concentrations up to 100X minimum inhibitory concentration was found to be non-toxic and facilitated mammalian cell migration in an in vitro scratch model. Collagen hydrogel is a promising pharmaceutical delivery vehicle that allows for safe, precise bacterial targeting for effective bacterial inhibition in a pro-regenerative scaffold.
Collapse
Affiliation(s)
- Evan Jarman
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Jordan Burgess
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Ayushi Sharma
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Kate Hayashigatani
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Amar Singh
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Paige Fox
- Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Division of Plastic & Reconstructive Surgery, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| |
Collapse
|
49
|
Rojas EM, Zhang H, Velu SE, Wu H. Tetracyclic homoisoflavanoid (+)-brazilin: a natural product inhibits c-di-AMP-producing enzyme and Streptococcus mutans biofilms. Microbiol Spectr 2024; 12:e0241823. [PMID: 38591917 PMCID: PMC11064632 DOI: 10.1128/spectrum.02418-23] [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: 06/21/2023] [Accepted: 03/02/2024] [Indexed: 04/10/2024] Open
Abstract
The tenacious biofilms formed by Streptococcus mutans are resistant to conventional antibiotics and current treatments. There is a growing need for novel therapeutics that selectively inhibit S. mutans biofilms while preserving the normal oral microenvironment. Previous studies have shown that increased levels of cyclic di-AMP, an important secondary messenger synthesized by diadenylate cyclase (DAC), favored biofilm formation in S. mutans. Thus, targeting S. mutans DAC is a novel strategy to inhibit S. mutans biofilms. We screened a small NCI library of natural products using a fluorescence detection assay. (+)-Brazilin, a tetracyclic homoisoflavanoid found in the heartwood of Caesalpinia sappan, was identified as one of the 11 "hits," with the greatest reduction (>99%) in fluorescence at 100 µM. The smDAC inhibitory profiles of the 11 "hits" established by a quantitative high-performance liquid chromatography assay revealed that (+)-brazilin had the most enzymatic inhibitory activity (87% at 100 µM) and was further studied to determine its half maximal inhibitory concentration (IC50 = 25.1 ± 0.98 µM). (+)-Brazilin non-competitively inhibits smDAC's enzymatic activity (Ki = 140.0 ± 27.13 µM), as determined by a steady-state Michaelis-Menten kinetics assay. In addition, (+)-brazilin's binding profile with smDAC (Kd = 11.87 µM) was illustrated by a tyrosine intrinsic fluorescence quenching assay. Furthermore, at low micromolar concentrations, (+)-brazilin selectively inhibited the biofilm of S. mutans (IC50 = 21.0 ± 0.60 µM) and other oral bacteria. S. mutans biofilms were inhibited by a factor of 105 in colony-forming units when treated with 50 µM (+)-brazilin. In addition, a significant dose-dependent reduction in extracellular DNA and glucan levels was evident by fluorescence microscopy imaging of S. mutans biofilms exposed to different concentrations of (+)-brazilin. Furthermore, colonization of S. mutans on a representative model of enamel using suspended hydroxyapatite discs showed a >90% reduction with 50 µM (+)-brazilin. In summary, we have identified a drug-like natural product inhibitor of S. mutans biofilm that not only binds to smDAC but can also inhibit the function of smDAC. (+)-Brazilin could be a good candidate for further development as a potent therapeutic for the prevention and treatment of dental caries.IMPORTANCEThis study represents a significant advancement in our understanding of potential therapeutic options for combating cariogenic biofilms produced by Streptococcus mutans. The research delves into the use of (+)-brazilin, a natural product, as a potent inhibitor of Streptococcus mutans' diadenylate cyclase (smDAC), an enzyme crucial in the formation of biofilms. The study establishes (+)-brazilin as a non-competitive inhibitor of smDAC while providing initial insights into its binding mechanism. What makes this finding even more promising is that (+)-brazilin does not limit its inhibitory effects to S. mutans alone. Instead, it demonstrates efficacy in hindering biofilms in other oral bacteria as well. The broader spectrum of anti-biofilm activity suggests that (+)-brazilin could potentially serve as a versatile tool in a natural product-based treatment for combating a range of conditions caused by resilient biofilms.
Collapse
Affiliation(s)
- Edwin M. Rojas
- School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hua Zhang
- Division of Biomaterial & Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Sadanandan E. Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hui Wu
- Division of Biomaterial & Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
50
|
Milovanović J, Božić DD, Pavlović B, Jotić A, Brkić S, Ćirković I. Biofilm-producing Bacteria and Quality of Life after Endoscopic Sinus Surgery in Patients with Chronic Rhinosinusitis with Nasal Polyposis. Am J Rhinol Allergy 2024; 38:159-168. [PMID: 38454786 DOI: 10.1177/19458924241236233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is one of the most common health disorders in humans and has a major impact on health-related quality of life (HRQoL). Of the many factors contributing to the etiology of CRS, less is known about the correlation between CRS and bacterial biofilms and their impact on HRQoL. OBJECTIVE The aim of this prospective study was to investigate the relationship between biofilm-producing bacteria and patients' objective findings and HRQoL. METHODS Forty-eight patients with CRSwNP were enrolled in a 12-month prospective study. The Lund-Mackay (LM) CT and endoscopic Lund-Kennedy (LK) scores were obtained before endoscopic sinus surgery (ESS), and patients completed the HRQoL instruments: the 22-item Sinonasal Outcome Test (SNOT-22), the 36-item Short Questionnaire (SF-36), and the visual analog scale (VAS). A sinus culture was obtained at ESS, bacteria were isolated, and in vitro quantification of the biofilm was performed. The LK score and HRQoL were determined postoperatively at months 1, 3, 6, and 12. RESULTS The most common bacterial isolates in patients with CRSwNP were Staphylococcus aureus (28%), coagulase-negative staphylococci (52%), and Pseudomonas aeruginosa (8%). Preoperatively, the highest LM and LK scores were found in patients with strong biofilm producers. Postoperative LK scores were significantly reduced in all patients. Postoperative VAS scores were significantly reduced from month 1 to month 12 postoperatively. Patients with strong biofilm producers had significantly worse nasal blockage, secretion, headache, facial pressure and pain, and loss of smell preoperatively, compared to patients with low biofilm producers. The most significant reduction in preoperative scores SNOT-22 and SF-36 (excluding physical functioning) was seen in patients with S. aureus and P. aeruginosa. CONCLUSIONS Patients with strong biofilm producers had higher LK and LM scores preoperatively, and greater improvement in LK and HRQoL scores postoperatively. Microbiologic surveillance of all CRS patients is recommended.
Collapse
Affiliation(s)
- Jovica Milovanović
- Clinic of Otorhinolaryngology and Maxillofacial Surgery, University Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade-Faculty of Medicine, Belgrade, Serbia
| | - Dragana D Božić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, Belgrade, Serbia
| | - Bojan Pavlović
- Clinic of Otorhinolaryngology and Maxillofacial Surgery, University Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade-Faculty of Medicine, Belgrade, Serbia
| | - Ana Jotić
- Clinic of Otorhinolaryngology and Maxillofacial Surgery, University Clinical Centre of Serbia, Belgrade, Serbia
- University of Belgrade-Faculty of Medicine, Belgrade, Serbia
| | - Snežana Brkić
- Institute for Laboratory Diagnostics "Konzilijum", Belgrade, Serbia
| | - Ivana Ćirković
- University of Belgrade-Faculty of Medicine, Belgrade, Serbia
- Institute of Microbiology and Immunology, Belgrade, Serbia
| |
Collapse
|