1
|
Jaber D, Younes N, Khalil E, Albsoul-Younes A, Zawiah M, Al-Bakri AG. Studying Microbial Ecology of Diabetic Foot Infections: Significance of PCR Analysis for Prudent Antimicrobial Stewardship. INT J LOW EXTR WOUND 2025; 24:497-505. [PMID: 38373396 DOI: 10.1177/15347346241230288] [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: 02/21/2024]
Abstract
This study presents a comprehensive investigation into the microbial ecology of diabetic foot infections (DFIs), using molecular-polymerase chain reaction (PCR) analysis to accurately identify the causative agents. One hundred DFI patients were recruited and classified using the Depth Extent Phase and Associated Etiology (DEPA) score according to their severity. Results revealed polymicrobial infections in 75% of cases, predominantly featuring Staphylococcus epidermidis (83%) and Staphylococcus aureus (63%). Importantly, 20% of samples exhibited facultative anaerobes Bacteroides fragilis or Clostridium perfringens, exclusively in high DEPA score ulcers. Candida albicans coinfection was identified in 19.2% of cases, underscoring the need for mycological evaluation. Empirical antimicrobial therapy regimens were tailored to DEPA severity, yet our findings highlighted a potential gap in methicillin-resistant Staphylococcus aureus (MRSA) coverage. Despite an 88% prevalence of methicillin-resistant Staphylococci, vancomycin usage was suboptimal. This raises concerns about the underestimation of MRSA risk and the need for tailored antibiotic guidelines. Our study demonstrates the efficacy of molecular-PCR analysis in identifying diverse microbial communities in DFIs, influencing targeted antibiotic choices. The results advocate for refined antimicrobial guidelines, considering regional variations in microbial patterns and judiciously addressing multidrug-resistant strains. This research contributes crucial insights for optimizing DFIs management and helps the physicians to have a fast decision in selection the suitable antibiotic for each patient and to decrease the risk of bacterial resistance from the improper use of broad-spectrum empirical therapies.
Collapse
Affiliation(s)
- Deema Jaber
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Nidal Younes
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Enam Khalil
- School of Pharmacy, The University of Jordan, Amman, Jordan
| | | | | | | |
Collapse
|
2
|
Hui J, Moon W, Dong P, dos Anjos C, Negri L, Yan H, Wang Y, Tam J, Dai T, Anderson RR, Goverman J, Gelfand JA, Yun S. Low-Irradiance Antimicrobial Blue Light-Bathing Therapy for Wound Infection Control. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412493. [PMID: 40229974 PMCID: PMC12120825 DOI: 10.1002/advs.202412493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/14/2025] [Indexed: 04/16/2025]
Abstract
The prevalence of antibiotic resistance and tolerance in wound infection management poses a serious and growing health threat, necessitating the exploration of alternative approaches. Antimicrobial blue light therapy offers an appealing, non-pharmacological solution. However, its practical application has been hindered by the requirement for high irradiance levels (50-200 mW/cm2), which particularly raises safety concerns. Here, a light-bathing strategy is introduced that employs prolonged, continuous exposure to blue light at an irradiance range lower by more than an order of magnitude (5 mW/cm2). This method consistently applies bacteriostatic pressure, keeping wound bioburden low, all while minimizing photothermal risks. Leveraging tailor-made, wearable light-emitting patches, preclinical trials on rat models of wound infection are conducted, demonstrating its safety and efficacy for suppressing infections induced by methicillin-resistant Staphylococcus aureus (S. aureus) and multidrug-resistant Pseudomonas aeruginosa (P. aeruginosa). The results pave a new way for the application of blue light therapy in wound care.
Collapse
Affiliation(s)
- Jie Hui
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - Wonjoon Moon
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - Pu‐Ting Dong
- Department of MicrobiologyThe ADA Forsyth InstituteBostonMA02142USA
| | - Carolina dos Anjos
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - Laisa Negri
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - Hao Yan
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - Ying Wang
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - Joshua Tam
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
- Harvard‐MIT Health Sciences and TechnologyCambridgeMA02139USA
| | - Tianhong Dai
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - R. Rox Anderson
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
| | - Jeremy Goverman
- Department of SurgeryHarvard Medical SchoolBostonMA02115USA
- Wound CenterMassachusetts General HospitalBostonMA02114USA
| | - Jeffrey A. Gelfand
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Division of Infectious DiseasesMassachusetts General HospitalBostonMA02114USA
| | - Seok‐Hyun Yun
- Wellman Center for PhotomedicineMassachusetts General HospitalBostonMA02139USA
- Department of DermatologyHarvard Medical SchoolBostonMA02115USA
- Harvard‐MIT Health Sciences and TechnologyCambridgeMA02139USA
| |
Collapse
|
3
|
Li X, Dong S, Pan Q, Liu N, Zhang Y. Antibiotic conjugates: Using molecular Trojan Horses to overcome drug resistance. Biomed Pharmacother 2025; 186:118007. [PMID: 40268370 DOI: 10.1016/j.biopha.2025.118007] [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: 07/14/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 04/25/2025] Open
Abstract
Antimicrobial resistance (AMR) has become a global health crisis due to the rapid emergence of multi-drug-resistant bacteria. The paucity of novel antibiotics in the clinical pipeline has exacerbated this issue, thereby warranting the development of new antibacterial therapies. The 'Trojan Horse' strategy entails conjugating antibiotics with bioactive components that not only facilitate the entry of antibiotic molecules into bacterial cells by circumventing the membrane barriers, but also augment the effects of conventional antibiotics against recalcitrant pathogens. These Trojan Horse elements can also serve as a promising tool for repurposing drugs with hitherto unexamined antimicrobial activity, or drugs with limited clinical utility due to considerable toxic side effects. In this review, we have discussed the current state of research on antibiotic conjugates with monoclonal antibodies (mAbs), antimicrobial peptides (AMPs) and the iron-chelating siderophores. The rationale and mechanisms of different antibiotic conjugates have been summarized, and the preclinical and clinical evidence pertaining to the activity of these conjugates against drug-resistant pathogens have been reviewed. Furthermore, the challenges associated with the clinical translation of these novel antimicrobials, and the future research directions have also been discussed. While antibiotic conjugates offer an attractive alternative to conventional antimicrobials, there are several obstacles to their clinical translation. A greater understanding of the mechanisms underlying AMR, and continuing advances in genetic engineering, synthetic biology, and bioinformatics will be crucial in designing more selective, potent, and safe antibiotic conjugates for tackling multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Xi Li
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyuan Dong
- Department of Thoracic surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qi Pan
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China
| | - Ning Liu
- Department of Rehabilitation, the First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Yijie Zhang
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China.
| |
Collapse
|
4
|
Xu J, Li T, Li F, Qiang H, Wei X, Zhan R, Chen Y. The applications and mechanisms of Rosmarinus officinalis L. in the management of different wounds and UV-irradiated skin. Front Pharmacol 2025; 15:1461790. [PMID: 39840083 PMCID: PMC11747526 DOI: 10.3389/fphar.2024.1461790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
Chronic wounds, especially non-healing wounds, significantly affect patients' quality of life and raise the costs of therapy. Wound healing is a complicated process involving interdependent stages, which may be impaired and delayed by infections with multi-drug resistant pathogens. Current medical strategies for wound healing, especially the treatment of non-healing wounds, exert limited therapeutic effects, thus become a dramatic challenge for modern medicine. There has been growing interest in exploring complementary approaches to enhance the wound healing process, and complementary therapy using herbs and their related products has gained increasing attention. Apart from skin wounds, dermal pathological changes caused by UV irradiation, may also benefit from such complementary therapy. The antimicrobial, anti-inflammatory, antioxidant, analgesic and collagen-promoting properties of extract from Rosmarinus officinalis L. (rosemary) have all been considered to contribute to the beneficial effects on different stages and multiple aspects of skin recovery after various wounds or UV irradiation. This review aims to summarize the applications and their underlying mechanisms of rosemary as part of the complementary therapy for injured and UV-irradiated skin based on the currently available evidence. The medicinal properties of rosemary and its application in wound dressing are first discussed, followed by summarization of its application in different types of wounds. A conclusion is reached and future directions are discussed. As research in this area continue to evolve, rosemary-derived products may become an integral part of holistic wound care strategies, offering a complementary approach to conventional treatments.
Collapse
Affiliation(s)
- Jianwen Xu
- Department of Nursing, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Ting Li
- Department of Nursing, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Fei Li
- Department of Neurology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Hong Qiang
- Department of Nursing, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Xiaoxiao Wei
- Department of Nursing, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Ruiwen Zhan
- Special Committee of Scientific Research, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Yun Chen
- Wound Clinic, Department of General Surgery, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
5
|
Eng YA, Thian JY, Ignacio J. Patients with hard-to-heal wounds: a review and synthesis of their experiences and perceptions of maggot debridement. J Wound Care 2025; 34:20-30. [PMID: 39797753 DOI: 10.12968/jowc.2022.0223] [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: 01/13/2025]
Abstract
OBJECTIVE There is little use of maggot debridement therapy (MDT) worldwide, albeit there is much literature supporting its benefits and effectiveness for hard-to-heal (chronic) wounds. Hard-to-heal wounds are becoming ever more prevalent and MDT can play a pivotal role in wound care management. This underuse can be associated with patients' perceptions and experiences of MDT. The aim of this study was to synthesise available evidence exploring the perceptions and experiences of patients with hard-to-heal wounds regarding MDT. METHOD This was a systematic review of qualitative evidence using thematic synthesis. Published papers and grey literature from inception until December 2021 were identified from PubMed, Embase, Cochrane, CINAHL, PsycINFO, ProQuest Social Science Premium Collection, Academic Search Complete, Web of Science, WorldWideScience.org, MedNar and Scopus. The screening process, assessment of methodological quality, and data extraction were conducted by two independent reviewers. Papers included had qualitative designs, such as phenomenology and descriptive-exploratory. Mixed methods papers and case reports were included for their qualitative components. Eligible papers were appraised using checklists from the Joanna Briggs Institute for qualitative research and case reports. Data were extracted using a standardised form. Findings were synthesised using Thomas and Harden's thematic synthesis approach. RESULTS A total of six papers exploring patients' (n=67) perceptions and experiences regarding MDT were included. Some 52 findings were grouped into 11 categories, generating three synthesised findings: the complexity of living with a hard-to-heal wound and seeking help; the effects of interpersonal relationships; and the relationship between the individual and the larvae. CONCLUSION Findings of the review showed that health professionals are key drivers in encouraging patients' acceptance of MDT and enhancing their treatment experiences. Together with a patient-centric approach, patients' concerns can be effectively addressed. Public and patient education can be beneficial to address misconceptions regarding the therapy.
Collapse
Affiliation(s)
- Yu An Eng
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore
| | - Jie Yun Thian
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore
| | - Jeanette Ignacio
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Clinical Research Centre, Singapore
| |
Collapse
|
6
|
Hurlow J, Wolcott RD, Bowler PG. Clinical management of chronic wound infections: The battle against biofilm. Wound Repair Regen 2025; 33:e13241. [PMID: 39600232 DOI: 10.1111/wrr.13241] [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: 07/25/2024] [Revised: 10/14/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Bacteria constitute the most abundant life form on earth, of which the majority exist in a protective biofilm state. Since the 1980s, we have learned much about the role of biofilm in human chronic infections, with associated global healthcare costs recently estimated at ~$386 billion. Chronic wound infection is a prominent biofilm-induced condition that is characterised by persistent inflammation and associated host tissue destruction, and clinical signs that are distinct from signs of acute wound infection. Biofilm also enables greater tolerance to antimicrobial agents in chronic wound infections compared with acute wound infections. Given the difficulty in eliminating wound biofilm, a multi-targeted strategy (namely biofilm-based wound care) involving debridement and antimicrobial therapies were introduced and have been practiced since the early 2000s. More recently, acknowledgement of the speed at which biofilm can develop and hence quickly interfere with wound healing has highlighted the need for an early anti-biofilm strategy to combat biofilm before it takes control and prevents wound healing. This strategy, referred to as wound hygiene, involves multiple tools in combination (debridement, cleansing, and antimicrobial dressings) to maximise success in biofilm removal and encourage wound healing. This review is intended to highlight the issues and challenges associated with biofilm-induced chronic infections, and specifically address the challenges in chronic wound management, and tools required to combat biofilm and encourage wound healing.
Collapse
Affiliation(s)
- Jennifer Hurlow
- ProHeal Wound Clinic, Baptist Memorial Hospital, Memphis, Tennessee, USA
| | | | | |
Collapse
|
7
|
Roberts JM, Milo S, Metcalf DG. Harnessing the Power of Our Immune System: The Antimicrobial and Antibiofilm Properties of Nitric Oxide. Microorganisms 2024; 12:2543. [PMID: 39770746 PMCID: PMC11677572 DOI: 10.3390/microorganisms12122543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Nitric oxide (NO) is a free radical of the human innate immune response to invading pathogens. NO, produced by nitric oxide synthases (NOSs), is used by the immune system to kill microorganisms encapsulated within phagosomes via protein and DNA disruption. Owing to its ability to disperse biofilm-bound microorganisms, penetrate the biofilm matrix, and act as a signal molecule, NO may also be effective as an antibiofilm agent. NO can be considered an underappreciated antimicrobial that could be levied against infected, at-risk, and hard-to-heal wounds due to the inherent lack of bacterial resistance, and tolerance by human tissues. NO produced within a wound dressing may be an effective method of disrupting biofilms and killing microorganisms in hard-to-heal wounds such as diabetic foot ulcers, venous leg ulcers, and pressure injuries. We have conducted a narrative review of the evidence underlying the key antimicrobial and antibiofilm mechanisms of action of NO for it to serve as an exogenously-produced antimicrobial agent in dressings used in the treatment of hard-to-heal wounds.
Collapse
Affiliation(s)
| | | | - Daniel Gary Metcalf
- Advanced Wound Care Research & Development, Convatec, Deeside Industrial Park, Deeside CH5 2NU, UK; (J.M.R.); (S.M.)
| |
Collapse
|
8
|
Atif M, Babuççu G, Riool M, Zaat S, Jonas U. Antimicrobial Peptide SAAP-148-Functionalized Hydrogels from Photocrosslinkable Polymers with Broad Antibacterial Activity. Macromol Rapid Commun 2024; 45:e2400785. [PMID: 39530205 DOI: 10.1002/marc.202400785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Antimicrobial peptides (AMPs) are promising alternatives to traditional antibiotics for treating skin wound infections. Nonetheless, their short half-life in biological environments restricts clinical applicability. Covalent immobilization of AMPs onto suitable substrates offers a comprehensive solution, creating contact-killing surfaces with long-term functionality. Here, a copolymer of poly[(hydroxy ethyl acrylamide)-co-(4-benzophenone acrylamide)-co-(pentafluorophenyl acrylate)-co-(ECOSURF EH-3 acrylate)], in short poly(HEAAm-co-BPAAm-co-PFPA-co-EH3A), is synthesized by free radical polymerization. Subsequent modification of active ester groups with the amine groups of SAAP-148, results in a copolymer, that is non-cytotoxic to human lung fibroblasts. UV photocrosslinking of the benzophenone units yields a polymer network that forms a hydrogel after swelling with aqueous medium. Both the SAAP-148-modified polymer in solution and the photocrosslinked hydrogels show good antimicrobial activity against strains of Escherichia coli, Staphylococcus aureus, Pseudomonas aeruginosa, and Acinetobacter baumannii, including multidrug-resistant strains, frequently found in wound infections. The covalent attachment of SAAP-148 prevents leaching, ensuring sustained antimicrobial activity for at least 48 h in diluted human blood plasma and 14 days in PBS. This prolonged retention of antimicrobial activity in human blood plasma significantly enhances its clinical potential. Overall, this study shows the potential of the AMP-functionalized photocrosslinkable polymer as antimicrobial wound dressings, providing an effective alternative to antibiotics.
Collapse
Affiliation(s)
- Muhammad Atif
- Macromolecular Chemistry, Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Strasse 2, 57076, Siegen, Germany
| | - Gizem Babuççu
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Immunology and Infectious Diseases, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Martijn Riool
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Immunology and Infectious Diseases, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, Am Biopark 9, 93053, Regensburg, Germany
| | - Sebastian Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, Amsterdam institute for Immunology and Infectious Diseases, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Ulrich Jonas
- Macromolecular Chemistry, Department of Chemistry and Biology, University of Siegen, Adolf-Reichwein-Strasse 2, 57076, Siegen, Germany
| |
Collapse
|
9
|
Dilhari A, Campbell PM, Munasinghe A, Brown H, Kaluarachchi TDJ, Gunasekara C, Pathirage S, Fernando N, Weerasekara D, Humphreys GJ, McBain AJ, Weerasekera M. Biofilms and microbiome profiles in chronic wounds: links to antibiotic use and wound severity in a Sri Lankan cohort. J Appl Microbiol 2024; 135:lxae262. [PMID: 39420474 DOI: 10.1093/jambio/lxae262] [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: 06/23/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024]
Abstract
AIMS We have characterized the microbiome of infected chronic diabetic wounds (CDWs), exploring associations with antibiotic use and wound severity in a Sri Lankan cohort. METHODS AND RESULTS Fifty CDW patients were enrolled, 38 of whom received antibiotics. Tissue biopsies were analysed by microbiome profiling, and wounds were graded using the University of Texas Wound Grading System. Biofilm presence was assessed in 20 wounds. The microbiome was largely dominated by Enterobacteriaceae, Pseudomonadaceae, Streptococcaceae, and Corynebacteriaceae. Proteobacteria levels were significantly higher in antibiotic-treated wounds (P = .019), with increased Pseudomonas abundance. Wounds were categorized as grade 1 (10), grade 2 (29), and grade 3 (11). Alpha diversity varied by wound grade (P = .015), with grade 2 wounds showing the highest diversity and grade 3 the lowest. All 20 tested wounds were biofilm-positive, and community composition varied more in antibiotic-treated wounds (P = .004). CONCLUSIONS CDW microbiomes were dominated by Enterobacteriaceae and Pseudomonadaceae, with elevated Proteobacteria in antibiotic-treated wounds. Alpha diversity correlated with wound severity, peaking in grade 2 wounds. The high prevalence of biofilms in wounds underscores the need for management of CDWs that address microbial complexity.
Collapse
Affiliation(s)
- Ayomi Dilhari
- Department of Basic Sciences, Faculty of Allied Health Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, 10250, Sri Lanka
- Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, 10250, Sri Lanka
| | - Paul M Campbell
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, United Kingdom
| | | | - Hannah Brown
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Dublin, D04W6F6, Ireland
| | - Thilini D J Kaluarachchi
- Department of Parasitology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, 10250, Sri Lanka
| | - Chinthika Gunasekara
- Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, 10250, Sri Lanka
| | - Sujatha Pathirage
- Department of Bacteriology, Medical Research Institute, Colombo 08, 00800, Sri Lanka
| | - Neluka Fernando
- Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, 10250, Sri Lanka
| | - Deepaka Weerasekara
- Department of Surgery, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, 10250, Sri Lanka
| | - Gavin J Humphreys
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL, United Kingdom
| | - Manjula Weerasekera
- Department of Microbiology, Faculty of Medical Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, 10250, Sri Lanka
| |
Collapse
|
10
|
Olutoye OO, Eriksson E, Menchaca AD, Kirsner RS, Tanaka R, Schultz G, Weir D, Wagner TL, Fabia RB, Naik-Mathuria B, Liu PY, Ead JK, Adebayo T, Armstrong DG, McMullin N, Samora JB, Akingba AG. Management of Acute Wounds-Expert Panel Consensus Statement. Adv Wound Care (New Rochelle) 2024; 13:553-583. [PMID: 38618741 DOI: 10.1089/wound.2023.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Significance: The Wound Healing Foundation recognized the need for consensus-based unbiased recommendations for the treatment of wounds. As a first step, a consensus on the treatment of chronic wounds was developed and published in 2022. The current publication on acute wounds represents the second step in this process. Acute wounds may result from any number of conditions, including burns, military and combat operations, and trauma to specific areas of the body. The management of acute wounds requires timely and evidence-driven intervention to achieve optimal clinical outcomes. This consensus statement provides the clinician with the necessary foundational approaches to the causes, diagnosis, and therapeutic management of acute wounds. Presented in a structured format, this is a useful guide for clinicians and learners in all patient care settings. Recent Advances: Recent advances in the management of acute wounds have centered on stabilization and treatment in the military and combat environment. Specifically, advancements in hemostasis, resuscitation, and the mitigation of infection risk through timely initiation of antibiotics and avoidance of high-pressure irrigation in contaminated soft tissue injury. Critical Issues: Critical issues include infection control, pain management, and the unique considerations for the management of acute wounds in pediatric patients. Future Directions: Future directions include new approaches to preventing the progression and conversion of burns through the use of specific gel formulations. Additionally, the use of three-dimensional bioprinting and photo-modulation for reconstruction is a promising area for continued discovery.
Collapse
Affiliation(s)
- Oluyinka O Olutoye
- Department of Surgery, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Elof Eriksson
- Harvard Medical School, Cambridge, Massachusetts, USA
| | - Alicia D Menchaca
- Department of General Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert S Kirsner
- University of Miami Hospital and Clinics Wound Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rica Tanaka
- Juntendo University Graduate School of Medicine, Division of Regenerative Therapy, Department of Plastic & Reconstructive Surgery, Juntendo University Hospital Podiatry Center, Tokyo, Japan
| | - Greg Schultz
- University of Florida, Gainesville, Florida, USA
| | - Dot Weir
- Saratoga Hospital Center for Wound Healing and Hyperbaric Medicine, Saratoga Springs, New York, USA
| | - Tracey L Wagner
- Department of Pediatrics, Section of Emergency Medicine, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Renata B Fabia
- Department of Surgery, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | - Paul Y Liu
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - J Karim Ead
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Temitope Adebayo
- Temple University School of Podiatric Medicine, Philadelphia, Pennsylvania, USA
| | - David G Armstrong
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Neil McMullin
- Plastic Surgery Consultant to the Surgeon General of the Army, Evans Army Community Hospital, Fort Carson, Colorado, USA
| | - Julie Balch Samora
- Department of Orthopedics, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - A George Akingba
- Department of Vascular Surgery, VA Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
11
|
Cassa MA, Gentile P, Girón-Hernández J, Ciardelli G, Carmagnola I. Smart self-defensive coatings with bacteria-triggered antimicrobial response for medical devices. Biomater Sci 2024; 12:5433-5449. [PMID: 39320148 DOI: 10.1039/d4bm00936c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Bacterial colonization and biofilm formation on medical devices represent one of the most urgent and critical challenges in modern healthcare. These issues not only pose serious threats to patient health by increasing the risk of infections but also exert a considerable economic burden on national healthcare systems due to prolonged hospital stays and additional treatments. To address this challenge, there is a need for smart, customized biomaterials for medical device fabrication, particularly through the development of surface modification strategies that prevent bacterial adhesion and the growth of mature biofilms. This review explores three bioinspired approaches through which antibacterial and antiadhesive coatings can be engineered to exhibit smart, stimuli-responsive features. This responsiveness is greatly valuable as it provides the coatings with a controlled, on-demand antibacterial response that is activated only in the presence of bacteria, functioning as self-defensive coatings. Such coatings can be designed to release antibacterial agents or change their surface properties/conformation in response to specific stimuli, like changes in pH, temperature, or the presence of bacterial enzymes. This targeted approach minimizes the risk of developing antibiotic resistance and reduces the need for continuous, high-dose antibacterial treatments, thereby preserving the natural microbiome and further reducing healthcare costs. The final part of the review reports a critical analysis highlighting the potential improvements and future evolutions regarding antimicrobial self-defensive coatings and their validation.
Collapse
Affiliation(s)
- Maria Antonia Cassa
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Torino 10129, Italy.
- Politecnico di Torino, Polito BIOmed Lab, Torino 10129, Italy
| | - Piergiorgio Gentile
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Joel Girón-Hernández
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Gianluca Ciardelli
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Torino 10129, Italy.
- Politecnico di Torino, Polito BIOmed Lab, Torino 10129, Italy
- National Research Council, Institute for Chemical and Physical Processes (CNR-IPCF), Pisa 56124, Italy
| | - Irene Carmagnola
- Politecnico di Torino, Department of Mechanical and Aerospace Engineering, Torino 10129, Italy.
- Politecnico di Torino, Polito BIOmed Lab, Torino 10129, Italy
| |
Collapse
|
12
|
Abdel-Fatah SS, Mohammad NH, Elshimy R, Mosallam FM. Impeding microbial biofilm formation and Pseudomonas aeruginosa virulence genes using biologically synthesized silver Carthamus nanoparticles. Microb Cell Fact 2024; 23:240. [PMID: 39238019 PMCID: PMC11378559 DOI: 10.1186/s12934-024-02508-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/09/2024] [Indexed: 09/07/2024] Open
Abstract
Long-term antibiotic treatment results in the increasing resistance of bacteria to antimicrobials drugs, so it is necessary to search for effective alternatives to prevent and treat pathogens that cause diseases. This study is aimed for biological synthesis of silver Carthamus nanoparticles (Ag-Carth-NPs) to combat microbial biofilm formation and Pseudomonas aeruginosa virulence genes. Ag-Carth-NPs are synthesized using Carthamus tenuis aqueous extract as environmentally friendly method has no harmful effect on environment. General factorial design is used to optimize Ag-Carth-NPs synthesis using three variables in three levels are Carthamus extract concentration, silver nitrate concentration and gamma radiation doses. Analysis of response data indicates gamma radiation has a significant effect on Ag-Carth-NPs production. Ag-Carth-NPs have sharp peak at λ max 425 nm, small and spherical particles with size 20.0 ± 1.22 nm, high stability up to 240 day with zeta potential around - 43 ± 0.12 mV, face centered cubic crystalline structure and FT-IR spectroscopy shows peak around 620 cm-1 that corresponding to AgNPs that stabilized by C. tenuis extract functional moiety. The antibacterial activity of Ag-Carth-NPs against pathogenic bacteria and fungi was determined using well diffusion method. The MIC values of Ag-Carth-NPs were (6.25, 6.25, 3.126, 25, 12.5, 12.5, 25 and 12.5 µg/ml), MBC values were (12.5, 12.5, 6.25, 50, 25, 25, 50 and 25 µg/ml) and biofilm inhibition% were (62.12, 68.25, 90.12, 69.51, 70.61, 71.12, 75.51 and 77.71%) against Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, Bacillus subtilis, Staphylococcus aureus, Staphylococcus epidermidis, Candida tropicalis and Candida albicans respectively. Ag-Carth-NPs has bactericidal efficacy and significantly reduced the swarming, swimming motility, pyocyanin and protease production of P. aeruginosa. Furthermore, P. aeruginosa ToxA gene expression was significantly down regulated by 81.5%, while exoU reduced by 78.1%, where lasR gene expression reduction was 68%, while the reduction in exoU was 66% and 60.1% decrease in lasB gene expression after treatment with Ag-Carth-NPs. This activity is attributed to effect of Ag-Carth-NPs on cell membrane integrity, down regulation of virulence gene expression, and induction of general and oxidative stress in P. aeruginosa. Ag-Carth-NPs have no significant cytotoxic effects on normal human cell (Hfb4) but have IC50 at 5.6µg/mL against of HepG-2 cells. Limitations of the study include studies with low risks of silver nanoparticles for in vitro antimicrobial effects and its toxicity.
Collapse
Affiliation(s)
- Sobhy S Abdel-Fatah
- Drug Radiation Research Department, Drug Microbiology Lab, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Nasser H Mohammad
- Radiation Microbiology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Rana Elshimy
- Microbiology and Immunology, Egyptian Drug Authority, Cairo, Egypt
- Microbiology and immunology, Faculty of Pharmacy, AL-Aharm Canadian University (ACU), Giza, Egypt
| | - Farag M Mosallam
- Drug Radiation Research Department, Drug Microbiology Lab, Biotechnology Division, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| |
Collapse
|
13
|
Meredith K, Coleborn MM, Forbes LE, Metcalf DG. Assessment of the Antibiofilm Performance of Silver-Containing Wound Dressings: A Dual-Species Biofilm Model. Cureus 2024; 16:e70086. [PMID: 39449882 PMCID: PMC11500486 DOI: 10.7759/cureus.70086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Background It is commonly accepted that microorganisms found within hard-to-heal wounds are present in biofilm form. Biofilms are often polymicrobial in nature, which increases their virulence and tolerance to antimicrobial agents. The aim of this study was to compare the antibiofilm activity of silver-containing antimicrobial wound dressings in a dual-species simulated wound biofilm model. Materials and methods Four silver-containing wound dressings were evaluated in vitro: Aquacel® Ag+ Extra™ dressing, KerraContact® Ag dressing, Durafiber* Ag dressing, and UrgoClean Ag dressing. Each dressing was applied to a simulated wound assembly containing biofilm-gauze inoculated with Klebsiella pneumoniae and methicillin-resistant Staphylococcus aureus (MRSA). Each biofilm-inoculated gauze was incubated at 35±3ºC for 6, 24, 48 and 72 hours. Enumeration of surviving biofilm bacteria at each time point was performed in triplicate for each test dressing and its equivalent control. Results Aquacel® Ag+ Extra™ dressing was observed to reduce the biofilm population within 24 hours with a >4 log10 kill observed for K. pneumoniae and >6 log10 for MRSA from an initial biofilm challenge of 4.16×109 CFU/mL. This kill rate was sustained for the duration of the challenge period, with Aquacel® Ag+ Extra™ dressing reducing the biofilm population to non-detectable levels (<30 Colony Forming Units (CFU) per test) by 72 hours for K. pneumoniae and by 48 hours for MRSA. KerraContact® Ag dressing demonstrated an initial reduction at 6 hours of ~2 log10 in both K. pneumoniae and MRSA. Durafiber* Ag dressing exhibited a slight, gradual reduction in biofilm population over the course of the test period, reducing each challenge organism by ~2.5 log10 by 72 hours. UrgoClean Ag was shown to have little to no impact on the dual-species biofilm with levels remaining similar or greater than that recovered prior to dressing application. The no-dressing biofilm-colonised gauze control demonstrated that the biofilm bacteria remained viable throughout the test period and species population proportionality was maintained. Conclusion Using a dual-species simulated wound biofilm model comprising the pathogens K. pneumoniae and MRSA, Aquacel® Ag+ Extra™ dressing demonstrated significantly greater antibiofilm activity than the other silver-containing dressings. The enhanced antibiofilm activity of Aquacel® Ag+ Extra™ dressing in this study may be attributed to the additional antibiofilm agents, ethylenediaminetetraacetic acid and benzethonium chloride, contained within the dressing.
Collapse
|
14
|
Yu J, Han W, Xu Y, Shen L, Zhao H, Zhang J, Xiao Y, Guo Y, Yu F. Biofilm-producing ability of methicillin-resistant Staphylococcus aureus clinically isolated in China. BMC Microbiol 2024; 24:241. [PMID: 38961344 PMCID: PMC11223284 DOI: 10.1186/s12866-024-03380-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Staphylococcus aureus, a commensal bacterium, colonizes the skin and mucous membranes of approximately 30% of the human population. Apart from conventional resistance mechanisms, one of the pathogenic features of S. aureus is its ability to survive in a biofilm state on both biotic and abiotic surfaces. Due to this characteristic, S. aureus is a major cause of human infections, with Methicillin-Resistant Staphylococcus aureus (MRSA) being a significant contributor to both community-acquired and hospital-acquired infections. RESULTS Analyzing non-repetitive clinical isolates of MRSA collected from seven provinces and cities in China between 2014 and 2020, it was observed that 53.2% of the MRSA isolates exhibited varying degrees of ability to produce biofilm. The biofilm positivity rate was notably high in MRSA isolates from Guangdong, Jiangxi, and Hubei. The predominant MRSA strains collected in this study were of sequence types ST59, ST5, and ST239, with the biofilm-producing capability mainly distributed among moderate and weak biofilm producers within these ST types. Notably, certain sequence types, such as ST88, exhibited a high prevalence of strong biofilm-producing strains. The study found that SCCmec IV was the predominant type among biofilm-positive MRSA, followed by SCCmec II. Comparing strains with weak and strong biofilm production capabilities, the positive rates of the sdrD and sdrE were higher in strong biofilm producers. The genetic determinants ebp, icaA, icaB, icaC, icaD, icaR, and sdrE were associated with strong biofilm production in MRSA. Additionally, biofilm-negative MRSA isolates showed higher sensitivity rates to cefalotin (94.8%), daptomycin (94.5%), mupirocin (86.5%), teicoplanin (94.5%), fusidic acid (81.0%), and dalbavancin (94.5%) compared to biofilm-positive MRSA isolates. The biofilm positivity rate was consistently above 50% in all collected specimen types. CONCLUSIONS MRSA strains with biofilm production capability warrant increased vigilance.
Collapse
Affiliation(s)
- Jingyi Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weihua Han
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanlei Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huilin Zhao
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Zhang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanghua Xiao
- School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Yinjuan Guo
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
15
|
Kamath P, Paul S, Valdes J, Gil J, Solis M, Higa A, Davis SC. In vitro analysis of interactions between Pseudomonas aeruginosa and Candida albicans treated with silver sulfadiazine in wound infections. JAC Antimicrob Resist 2024; 6:dlae075. [PMID: 38741896 PMCID: PMC11089415 DOI: 10.1093/jacamr/dlae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
Background Microorganisms tend to rely on close relationships with other species to survive. Consequently, biofilms formed by interactions of different species have been shown to delay the wound healing process. Studies suggest these mixed-population infections contribute to the development of drug resistance and inhibition of host immune response. Silver sulfadiazine (SSD) has been shown to effectively decrease the risk of infection in an open wound. Typically, these are bacterial wound infections; however, the role of fungal species needs further attention. Objectives The purpose of this in vitro study was to determine the effect of SSD on interactions between Pseudomonas aeruginosa 09-009 (PA1) or P. aeruginosa 09-010 (PA2) and Candida albicans ATTC 64550 (CA). Methods A mixture of 4 mL of tryptic soy broth (TSB) and 100 µL of CA and/or PA1 or PA2 (∼106 log cfu/mL) inoculums were deposited into either wells or vials. The wells or vials were then sonicated (50 W for 10 s) to separate microorganisms attached to the walls. After incubation, cell counts were performed at 24 and 48 h for each microorganism using specific media. Results Our results show that without SSD treatment, P. aeruginosa exhibits an inhibitory effect on C. albicans. Treatment with SSD demonstrated significant reduction of P. aeruginosa; however, C. albicans persisted. This experiment demonstrates that SSD was effective in reducing the bioburden of both P. aeruginosa strains after 24 and 48 h; however, it was not as effective in reducing C. albicans. Conclusions The data suggest that for polymicrobial mixed infections containing Pseudomonas spp. and C. albicans, treatment with SSD may be beneficial but does not provide adequate microorganism eradication. As such, added treatments that provide coverage for Candida infection are necessary. Additional in vivo studies are needed to obtain a better understanding of the complex interactions between these organisms.
Collapse
Affiliation(s)
- Preetha Kamath
- Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, RMSB Room 2089 1600 NW 10 Avenue, Miami, FL 33136, USA
| | - Suchismita Paul
- Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, RMSB Room 2089 1600 NW 10 Avenue, Miami, FL 33136, USA
| | - Jose Valdes
- Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, RMSB Room 2089 1600 NW 10 Avenue, Miami, FL 33136, USA
| | - Joel Gil
- Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, RMSB Room 2089 1600 NW 10 Avenue, Miami, FL 33136, USA
| | - Michael Solis
- Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, RMSB Room 2089 1600 NW 10 Avenue, Miami, FL 33136, USA
| | - Alex Higa
- Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, RMSB Room 2089 1600 NW 10 Avenue, Miami, FL 33136, USA
| | - Stephen C Davis
- Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, RMSB Room 2089 1600 NW 10 Avenue, Miami, FL 33136, USA
| |
Collapse
|
16
|
Mayer DO, Tettelbach WH, Ciprandi G, Downie F, Hampton J, Hodgson H, Lazaro-Martinez JL, Probst A, Schultz G, Stürmer EK, Parnham A, Frescos N, Stang D, Holloway S, Percival SL. Best practice for wound debridement. J Wound Care 2024; 33:S1-S32. [PMID: 38829182 DOI: 10.12968/jowc.2024.33.sup6b.s1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Affiliation(s)
- Dieter O Mayer
- General and Vascular Surgeon, Institute for Advanced Wound Care and Education, Hausen am Albis, Switzerland
| | - William H Tettelbach
- Chief Medical Officer, RestorixHealth, Metairie, LA; Adjunct Assistant Professor, Duke University School of Medicine, Durham, NC, US
| | - Guido Ciprandi
- Plastic and Paediatric Surgeon, Bambino Gesu' Children's Hospital, Research Institute, Rome, Italy
| | - Fiona Downie
- Senior Lecturer Advanced Practice, Faculty of Health, Education, Medicine and Social Care, Anglia Ruskin University, UK
| | - Jane Hampton
- Consultant Nurse, Aarhus Kommune, Middle Jutland, Denmark
| | - Heather Hodgson
- Lead Nurse, Tissue Viability, Acute and Partnerships, NHS Greater Glasgow and Clyde, UK
| | | | - Astrid Probst
- ANP Woundmanagement, Kreiskliniken Reutlingen gGmbH, Germany
| | - Greg Schultz
- Professor of Obstetrics and Gynecology, Director, Institute for Wound Research, University of Florida, US
| | - Ewa Klara Stürmer
- Surgical Head of the Comprehensive Wound Centre UKE, Head of Translational Wound Research, Department of Vascular Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Alison Parnham
- Teaching Associate, Clinical Nurse specialist, Tissue Viability, University of Nottingham, UK
| | | | - Duncan Stang
- Podiatrist and Diabetes Foot Coordinator for Scotland, UK
| | - Samantha Holloway
- Reader and Programme Director, Masters in Wound Healing and Tissue Repair, Centre for Medical Education, School of Medicine, Cardiff University, UK
| | - Steve L Percival
- CEO and Director, Biofilm Centre, 5D Health Protection Group and Professor (Hon), Faculty of Biology, Medicine and Health, University of Manchester, UK
| |
Collapse
|
17
|
Garzón HS, Loaiza-Oliva M, Martínez-Pabón MC, Puerta-Suárez J, Téllez Corral MA, Bueno-Silva B, Suárez DR, Díaz-Báez D, Suárez LJ. Antibiofilm and Immune-Modulatory Activity of Cannabidiol and Cannabigerol in Oral Environments-In Vitro Study. Antibiotics (Basel) 2024; 13:342. [PMID: 38667018 PMCID: PMC11047394 DOI: 10.3390/antibiotics13040342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/29/2024] Open
Abstract
OBJECTIVE To evaluate the in vitro antimicrobial and antibiofilm properties and the immune modulatory activity of cannabidiol (CBD) and cannabigerol (CBG) on oral bacteria and periodontal ligament fibroblasts (PLF). METHODS Cytotoxicity was assessed by propidium iodide flow cytometry on fibroblasts derived from the periodontal ligament. The minimum inhibitory concentration (MIC) of CBD and CBG for S. mutans and C. albicans and the metabolic activity of a subgingival 33-species biofilm under CBD and CBG treatments were determined. The Quantification of cytokines was performed using the LEGENDplex kit (BioLegend, Ref 740930, San Diego, CA, USA). RESULTS CBD-treated cell viability was greater than 95%, and for CBG, it was higher than 88%. MIC for S. mutans with CBD was 20 µM, and 10 µM for CBG. For C. albicans, no inhibitory effect was observed. Multispecies biofilm metabolic activity was reduced by 50.38% with CBD at 125 µg/mL (p = 0.03) and 39.9% with CBG at 62 µg/mL (p = 0.023). CBD exposure at 500 µg/mL reduced the metabolic activity of the formed biofilm by 15.41%, but CBG did not have an effect. CBG at 10 µM caused considerable production of anti-inflammatory mediators such as TGF-β and IL-4 at 12 h. CBD at 10 µM to 20 µM produced the highest amount of IFN-γ. CONCLUSION Both CBG and CBD inhibit S. mutans; they also moderately lower the metabolic activity of multispecies biofilms that form; however, CBD had an effect on biofilms that had already developed. This, together with the production of anti-inflammatory mediators and the maintenance of the viability of mammalian cells from the oral cavity, make these substances promising for clinical use and should be taken into account for future studies.
Collapse
Affiliation(s)
- Hernan Santiago Garzón
- Programa de Doctorado en Ingeniería, Facultad de Ingeniería, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (H.S.G.); (D.R.S.)
| | - Manuela Loaiza-Oliva
- Laboratory of Oral Microbiology, Faculty of Dentistry, University of Antioquia, Medellín 050010, Colombia; (M.L.-O.); (M.C.M.-P.); (J.P.-S.)
| | - María Cecilia Martínez-Pabón
- Laboratory of Oral Microbiology, Faculty of Dentistry, University of Antioquia, Medellín 050010, Colombia; (M.L.-O.); (M.C.M.-P.); (J.P.-S.)
| | - Jenniffer Puerta-Suárez
- Laboratory of Oral Microbiology, Faculty of Dentistry, University of Antioquia, Medellín 050010, Colombia; (M.L.-O.); (M.C.M.-P.); (J.P.-S.)
- Grupo Reproducción, Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Antioquia, Medellín 050012, Colombia
| | - Mayra Alexandra Téllez Corral
- Centro de Investigaciones Odontológicas, Facultad de Odontología, Pontificia Universidad Javeriana, Bogotá 110231, Colombia;
| | - Bruno Bueno-Silva
- Department of Periodontology, Dental Research Division, Guarulhos University, Guarulhos 07023-070, Brazil;
- Departamento de Biociências, Faculdade de Odontologia de Piracicaba, Universidade de Campinas (UNICAMP), Piracicaba 13414-903, Brazil
| | - Daniel R. Suárez
- Programa de Doctorado en Ingeniería, Facultad de Ingeniería, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (H.S.G.); (D.R.S.)
| | - David Díaz-Báez
- Unit of Basic Oral Investigation-UIBO, Facultad de Odontología, Universidad El Bosque, Bogotá 11001, Colombia;
| | - Lina J. Suárez
- Centro de Investigaciones Odontológicas, Departamento del Sistema Periodontal, Facultad de Odontología, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
- Departamento de Ciencias Básicas y Medicina Oral, Facultad de Odontología, Universidad Nacional de Colombia, Bogotá 111321, Colombia
| |
Collapse
|
18
|
Brożyna M, Dudek B, Kozłowska W, Malec K, Paleczny J, Detyna J, Fabianowska-Majewska K, Junka A. The chronic wound milieu changes essential oils' antibiofilm activity-an in vitro and larval model study. Sci Rep 2024; 14:2218. [PMID: 38278929 PMCID: PMC10817982 DOI: 10.1038/s41598-024-52424-6] [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: 09/26/2023] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Essential Oils (EOs) are currently being researched as potential antibiofilm agents to combat infections related to chronic wound biofilms. As documented in the literature, EOs' in vitro antibacterial properties are often assessed using standard microbiological media and conditions that do not accurately reflect the actual environment of a chronic wound. To address this issue, In vitro Wound Milieu (IVWM) medium, which closely resembles the environment of a chronic wound, was applied for culturing S. aureus biofilms (n = 12) in this research. Biofilms cultivated in the standard Tryptic Soy Broth (TSB) medium served as a control for the experiment. Key biofilm features were analyzed and compared. Subsequently, staphylococci were exposed to the activity of thyme or rosemary EOs (T-EO and R-EO, respectively). As proof of concept, the cytotoxicity of T-EO and its antimicrobial in vivo activity were assessed using a G. mellonella larvae model. Key features of biofilm-forming cells were lower in the IVWM than in the TSB medium: biomass (up to 8 times), metabolic activity (up to 9 times), cell number (up to 100 times), and the live/dead cells ratio. Conversely, biofilm thickness was higher (up to 25%) in IVWM. These differences translated into varied responses of the biofilms to EOs exposure. The application of T-EO led to a greater reduction (up to 2 times) in 67% of biofilm-forming strains in IVWM compared to the TSB medium. Conversely, exposure to R-EO resulted in a higher reduction (up to 2.6 times) of 83% of biofilm-forming strains in TSB than in IVWM. The application of T-EO was not only non-toxic to G. mellonella larvae but also increased the survival of larvae infected with staphylococci (from 48 to 85%). Our findings suggest that EOs not only show promise as agents for treating biofilm-related wound infections but also that providing conditions reflecting the specific niche of the human body is of paramount importance in influencing the results obtained. However, before clinical application, challenges related to the methods of assessing their activity, microbial intra-species variability, and different levels of activity of various EOs should be analyzed and standardized.
Collapse
Affiliation(s)
- Malwina Brożyna
- Platform for Unique Models Application, Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, Wroclaw, Poland.
| | - Bartłomiej Dudek
- Platform for Unique Models Application, Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, Wroclaw, Poland
| | - Weronika Kozłowska
- Division of Pharmaceutical Biotechnology, Department of Pharmaceutical Biology and Biotechnology, Wroclaw Medical University, Wroclaw, Poland
| | - Katarzyna Malec
- Department of Drug Form Technology, Wroclaw Medical University, Wroclaw, Poland
| | - Justyna Paleczny
- Platform for Unique Models Application, Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, Wroclaw, Poland
| | - Jerzy Detyna
- Department of Mechanics, Materials and Biomedical Engineering, Wroclaw University of Science and Technology, Wroclaw, Poland
| | | | - Adam Junka
- Platform for Unique Models Application, Department of Pharmaceutical Microbiology and Parasitology, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
19
|
Cheng L, Du WL, Zhang Y, Chen Z, Shen YM. Application of Staged Negative Pressure Wound Therapy and Flap Surgery for Infection Control After Patellar Internal Fixation Surgery. J Craniofac Surg 2024; 35:e74-e78. [PMID: 37982783 DOI: 10.1097/scs.0000000000009854] [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: 06/16/2022] [Accepted: 10/07/2023] [Indexed: 11/21/2023] Open
Abstract
The anatomic position of the patella is superficial, making it vulnerable to injuries. Treatment of patella infection after internal fixation surgery remains a big challenge due to minimal soft tissue coverage and vital tissue exposure. Forty-two patients aged 10 to 59 years were admitted to the institution's burn unit between January 2010 and December 2019. Each presented with infection after patellar fracture surgery. Twenty-seven infections were superficial, whereas 15 were deep with pyogenous arthritis of the knee. Negative pressure wound therapy (NPWT) was applied after radical debridement to remove necrotic subcutaneous tissues and internal fixation devices. In addition, cases with septic arthritis were irrigated continuously with normal saline. After 5 to 10 days of NPWT treatment and irrigation, wound infection was well controlled. Afterward, 42 wounds were resurfaced with pedicled flaps, the 42 patients received 17 reverse-flow anterolateral thigh (ALT) perforator flap, 12 medial sural artery perforator flaps, 7 gastrocnemius musculocutaneous flaps, as well as 6 saphenous artery flaps. Thirty-seven flaps survived uneventfully. However, 3 flaps developed venous congestion in the distal end. Two flaps developed tip necrosis. All patients were followed up between 3 and 48 months. Infection beneath the flap occurred in 3 patients and healed after an additional debridement surgery. The staged NPWT and flap surgery strategy focus on thorough debridement and immediate internal fixation devices removal, effective fracture fixation, efficient NPWT application, targeted administration of antibiotics, and adequate soft tissue coverage. This study established that the procedure was effective in infection control after patellar internal fixation surgery.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Beijing, P.R. China
| | | | | | | | | |
Collapse
|
20
|
Suresh G, Srivastava S. A concise review on genes involved in biofilm-related disease and differential gene expression in medical-related biofilms. MICROBIAL BIOFILMS 2024:215-235. [DOI: 10.1016/b978-0-443-19252-4.00012-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
21
|
Shin MK, Hwang IW, Jang BY, Bu KB, Han DH, Lee SH, Oh JW, Yoo JS, Sung JS. The Identification of a Novel Spider Toxin Peptide, Lycotoxin-Pa2a, with Antibacterial and Anti-Inflammatory Activities. Antibiotics (Basel) 2023; 12:1708. [PMID: 38136742 PMCID: PMC10740532 DOI: 10.3390/antibiotics12121708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
With the increasing challenge of controlling infectious diseases due to the emergence of antibiotic-resistant strains, the importance of discovering new antimicrobial agents is rapidly increasing. Animal venoms contain a variety of functional peptides, making them a promising platform for pharmaceutical development. In this study, a novel toxin peptide with antibacterial and anti-inflammatory activities was discovered from the spider venom gland transcriptome by implementing computational approaches. Lycotoxin-Pa2a (Lytx-Pa2a) showed homology to known-spider toxin, where functional prediction indicated the potential of both antibacterial and anti-inflammatory peptides without hemolytic activity. The colony-forming assay and minimum inhibitory concentration test showed that Lytx-Pa2a exhibited comparable or stronger antibacterial activity against pathogenic strains than melittin. Following mechanistic studies revealed that Lytx-Pa2a disrupts both cytoplasmic and outer membranes of bacteria while simultaneously inducing the accumulation of reactive oxygen species. The peptide exerted no significant toxicity when treated to human primary cells, murine macrophages, and bovine red blood cells. Moreover, Lytx-Pa2a alleviated lipopolysaccharide-induced inflammation in mouse macrophages by suppressing the expression of inflammatory mediators. These findings not only suggested that Lytx-Pa2a with dual activity can be utilized as a new antimicrobial agent for infectious diseases but also demonstrated the implementation of in silico methods for discovering a novel functional peptide, which may enhance the future utilization of biological resources.
Collapse
Affiliation(s)
- Min Kyoung Shin
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| | - In-Wook Hwang
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| | - Bo-Young Jang
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| | - Kyung-Bin Bu
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| | - Dong-Hee Han
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| | - Seung-Ho Lee
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| | - Jin Wook Oh
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| | - Jung Sun Yoo
- 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; (M.K.S.); (I.-W.H.); (B.-Y.J.); (K.-B.B.); (D.-H.H.); (S.-H.L.); (J.W.O.)
| |
Collapse
|
22
|
Shaikh SA, Patel B, Priyadarsini IK, Vavilala SL. Combating planktonic and biofilm growth of Serratia marcescens by repurposing ebselen. Int Microbiol 2023; 26:693-704. [PMID: 36507979 DOI: 10.1007/s10123-022-00301-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 10/21/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022]
Abstract
AIM OF THE STUDY The rising instances of multidrug-resistant pathogens are rapidly evolving into a global healthcare crisis. Identifying new ways of synthesis of antibiotics is both time-consuming and expensive. Repurposing existing drugs for the treatment of such antimicrobial-resistant pathogens has also been explored. METHODS AND RESULTS In the current study, ebselen was screened for antibacterial and antibiofilm activity against Serratia marcescens. Various antibacterial studies such as minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), time-kill curves, intracellular reactive oxygen species (ROS) quantification, and colony-forming unit assays were performed. The antibiofilm potential was assayed by biofilm inhibition, cell surface hydrophobicity assay, eradication, quantification of extracellular DNA (eDNA), and extracellular polymeric substance (EPS) layer and scanning electron microscopy (SEM) analysis were performed. Anti-quorum sensing assay was validated by quantifying the virulence factors production. Further molecular docking of ebselen with two quorum sensing (QS) specific proteins was also carried out. Antibacterial susceptibility tests showed potent antimicrobial activity of ebselen against S. marcescens with MIC50 of 14 μg/mL. Ebselen's ability to disturb the redox environment by inducing significant ROS generation led to bacterial death. It also showed concentration-dependent bactericidal activity as indicated by reduced bacterial growth and colony-forming unit propagation. Ebselen was also found to prevent biofilm attachment by altering the cell surface hydrophobicity while also being effective against preformed biofilms as validated by scanning electron microscopy (SEM) analysis. Additionally, ebselen showed reduced virulence factors like urease enzyme activity and prodigiosin pigment production indicating its promising anti-quorum sensing potential. Molecular docking analysis validated the strong binding of ebselen with QS-specific proteins (1Joe and PigG) with binding energies of - 6.6 and - 8.1kj/mol through hydrogen bonds and aromatic interactions. These results show that ebselen has potent antibiofilm potential that can be explored to identify treatment against bacterial infections.
Collapse
Affiliation(s)
- Shaukat Ali Shaikh
- School of Chemical Sciences, UM DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina Campus, Mumbai, India
| | - Bharti Patel
- School of Biological Sciences, UM DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina Campus, Mumbai, India
| | - Indira K Priyadarsini
- School of Chemical Sciences, UM DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina Campus, Mumbai, India.
| | - Sirisha L Vavilala
- School of Biological Sciences, UM DAE Centre for Excellence in Basic Sciences, University of Mumbai, Kalina Campus, Mumbai, India.
| |
Collapse
|
23
|
Debroy R, Ramaiah S. Consolidated knowledge-guided computational pipeline for therapeutic intervention against bacterial biofilms - a review. BIOFOULING 2023; 39:928-947. [PMID: 38108207 DOI: 10.1080/08927014.2023.2294763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Biofilm-associated bacterial infections attributed to multifactorial antimicrobial resistance have caused worldwide challenges in formulating successful treatment strategies. In search of accelerated yet cost-effective therapeutics, several researchers have opted for bioinformatics-based protocols to systemize targeted therapies against biofilm-producing strains. The present review investigated the up-to-date computational databases and servers dedicated to anti-biofilm research to design/screen novel biofilm inhibitors (antimicrobial peptides/phytocompounds/synthetic compounds) and predict their biofilm-inhibition efficacy. Scrutinizing the contemporary in silico methods, a consolidated approach has been highlighted, referred to as a knowledge-guided computational pipeline for biofilm-targeted therapy. The proposed pipeline has amalgamated prominently employed methodologies in genomics, transcriptomics, interactomics and proteomics to identify potential target proteins and their complementary anti-biofilm compounds for effective functional inhibition of biofilm-linked pathways. This review can pave the way for new portals to formulate successful therapeutic interventions against biofilm-producing pathogens.
Collapse
Affiliation(s)
- Reetika Debroy
- Medical and Biological Computing Laboratory, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
- Department of Bio-Medical Sciences, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
- Department of Bio-Sciences, School of Bio-Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| |
Collapse
|
24
|
Moiketsi BN, Makale KPP, Rantong G, Rahube TO, Makhzoum A. Potential of Selected African Medicinal Plants as Alternative Therapeutics against Multi-Drug-Resistant Bacteria. Biomedicines 2023; 11:2605. [PMID: 37892979 PMCID: PMC10604549 DOI: 10.3390/biomedicines11102605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/04/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Antimicrobial resistance is considered a "One-Health" problem, impacting humans, animals, and the environment. The problem of the rapid development and spread of bacteria resistant to multiple antibiotics is a rising global health threat affecting both rich and poor nations. Low- and middle-income countries are at highest risk, in part due to the lack of innovative research on the surveillance and discovery of novel therapeutic options. Fast and effective drug discovery is crucial towards combatting antimicrobial resistance and reducing the burden of infectious diseases. African medicinal plants have been used for millennia in folk medicine to cure many diseases and ailments. Over 10% of the Southern African vegetation is applied in traditional medicine, with over 15 species being partially or fully commercialized. These include the genera Euclea, Ficus, Aloe, Lippia. And Artemisia, amongst many others. Bioactive compounds from indigenous medicinal plants, alone or in combination with existing antimicrobials, offer promising solutions towards overcoming multi-drug resistance. Secondary metabolites have different mechanisms and modes of action against bacteria, such as the inhibition and disruption of cell wall synthesis; inhibition of DNA replication and ATP synthesis; inhibition of quorum sensing; inhibition of AHL or oligopeptide signal generation, broadcasting, and reception; inhibition of the formation of biofilm; disruption of pathogenicity activities; and generation of reactive oxygen species. The aim of this review is to highlight some promising traditional medicinal plants found in Africa and provide insights into their secondary metabolites as alternative options in antibiotic therapy against multi-drug-resistant bacteria. Additionally, synergism between plant secondary metabolites and antibiotics has been discussed.
Collapse
Affiliation(s)
| | | | | | - Teddie O. Rahube
- Department of Biological Sciences and Biotechnology, Faculty of Science, Botswana International University of Science and Technology (BIUST), Private Bag 16, Palapye, Botswana; (B.N.M.); (K.P.P.M.); (G.R.)
| | - Abdullah Makhzoum
- Department of Biological Sciences and Biotechnology, Faculty of Science, Botswana International University of Science and Technology (BIUST), Private Bag 16, Palapye, Botswana; (B.N.M.); (K.P.P.M.); (G.R.)
| |
Collapse
|
25
|
Kong J, Wang Y, Yao Z, Lin Y, Zhang Y, Han Y, Zhou T, Ye J, Cao J. Eugenol works synergistically with colistin against colistin-resistant Pseudomonas aeruginosa and Klebsiella pneumoniae isolates by enhancing membrane permeability. Microbiol Spectr 2023; 11:e0366622. [PMID: 37707450 PMCID: PMC10581171 DOI: 10.1128/spectrum.03666-22] [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: 09/16/2022] [Accepted: 07/20/2023] [Indexed: 09/15/2023] Open
Abstract
Colistin is a potent antibiotic for the treatment of carbapenem-resistant Gram-negative bacteria and is considered a last-resort drug. Unfortunately, the incidence of colistin-resistant bacteria isolated from patients is continuously growing due to clinical reuse of colistin. In this study, we found that the combination of colistin and eugenol has a significant synergistic antibacterial effect and reverses the sensitivity of colistin-resistant Pseudomonas aeruginosa and Klebsiella pneumoniae against colistin, as confirmed by checkerboard and time-kill assays. Crystal violet staining and scanning electron microscopy revealed colistin and eugenol's synergistic antibiofilm action. Concerning the synergy mechanism, the results revealed that the combination of eugenol and colistin increases membrane permeability and causes considerable membrane damage, further inhibiting bacteria synergistically. Meanwhile, up to 500 µg/mL of eugenol is non-toxic to RAW 264.7 cells, and the colistin/eugenol combination is also efficacious in vivo, as demonstrated by the Galleria mellonella infection model. Our findings indicate that the colistin/eugenol combination is a viable treatment option for colistin-resistant P. aeruginosa and K. pneumoniae clinical infections. IMPORTANCE Colistin is used as a last resort for severe infections caused by multidrug-resistant Gram-negative bacteria, however, colistin resistance is increasing. As a result, we investigated the synergistic effect of eugenol/colistin combination, and the results revealed significant antibacterial and antibiofilm action. Eugenol may help clinical colistin-resistant Pseudomonas aeruginosa and Klebsiella pneumoniae recover their susceptibility. These findings suggest that combining eugenol and colistin may be a viable treatment option for colistin-resistant pathogen clinical infections.
Collapse
Affiliation(s)
- Jingchun Kong
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yue Wang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zhuocheng Yao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yishuai Lin
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yi Zhang
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yijia Han
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Tieli Zhou
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jianzhong Ye
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Jianming Cao
- Department of Medical Lab Science, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
26
|
Nesse LL, Osland AM, Asal B, Mo SS. Evolution of antimicrobial resistance in E. coli biofilm treated with high doses of ciprofloxacin. Front Microbiol 2023; 14:1246895. [PMID: 37731931 PMCID: PMC10509014 DOI: 10.3389/fmicb.2023.1246895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/07/2023] [Indexed: 09/22/2023] Open
Abstract
The evolution of antimicrobial resistance (AMR) has mainly been studied in planktonic bacteria exposed to sub-inhibitory antimicrobial (AM) concentrations. However, in a number of infections that are treated with AMs the bacteria are located in biofilms where they tolerate high doses of AM. In the present study, we continuously exposed biofilm residing E. coli at body temperature to high ciprofloxacin (CIP) concentrations increasing from 4 to 130 times the minimal inhibitory concentration (MIC), i.e., from 0.06 to 2.0 mg/L. After 1 week, the biofilms were full of CIP resistant bacteria. The evolutionary trajectory observed was the same as described in the literature for planktonic bacteria, i.e., starting with a single mutation in the target gene gyrA followed by mutations in parC, gyrB, and parE, as well as in genes for regulation of multidrug efflux pump systems and outer membrane porins. Strains with higher numbers of these mutations also displayed higher MIC values. Furthermore, the evolution of CIP resistance was more rapid, and resulted in strains with higher MIC values, when the bacteria were biofilm residing than when they were in a planktonic suspension. These results may indicate that extensive clinical AM treatment of biofilm-residing bacteria may not only fail to eradicate the infection but also pose an increased risk of AMR development.
Collapse
Affiliation(s)
- Live L. Nesse
- Department of Food Safety and Animal Health Research, Norwegian Veterinary Institute, Ås Municipality, Norway
| | - Ane Mohr Osland
- Department of Microbiology, Norwegian Veterinary Institute, Ås Municipality, Norway
| | - Basma Asal
- Department of Bacteriology, Norwegian Veterinary Institute, Ås Municipality, Norway
| | - Solveig Sølverød Mo
- Department of Bacteriology, Norwegian Veterinary Institute, Ås Municipality, Norway
| |
Collapse
|
27
|
Abu Rached N, Kley S, Storck M, Meyer T, Stücker M. Cold Plasma Therapy in Chronic Wounds-A Multicenter, Randomized Controlled Clinical Trial (Plasma on Chronic Wounds for Epidermal Regeneration Study): Preliminary Results. J Clin Med 2023; 12:5121. [PMID: 37568525 PMCID: PMC10419810 DOI: 10.3390/jcm12155121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic wounds (CWs) pose a significant health challenge in clinical practice. Standard wound therapy (SWT) is currently considered the gold standard. However, recent evidence suggests that cold plasma therapy (CPT) holds promise for improving CWs. In light of this, the POWER study was conducted as a multicenter, randomized clinical trial to investigate the effect of large-area plasma application compared with SWT in patients with chronic, non-healing arterial or venous wounds on the lower leg. To analyze the interim results, we employed a comprehensive range of statistical tests, including both parametric and non-parametric methods, as well as GLS model regression and an ordinal mixed model. Our findings clearly demonstrate that CPT therapy significantly accelerates wound closure compared with SWT. In fact, complete wound closure was exclusively observed in the CPT group during the intervention period. Additionally, the CPT group required significantly less antibiotic therapy (4%) compared with the SWT group (23%). Furthermore, CPT led to a significant reduction in wound pain and improved quality of life compared with SWT. In conclusion, the study highlights that the combination of CPT and SWT surpasses monotherapy with SWT alone.
Collapse
Affiliation(s)
- Nessr Abu Rached
- Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany;
| | - Susanne Kley
- Scientific Institute for Health Economics and Health Research, Markt 9, 04109 Leipzig, Germany;
| | - Martin Storck
- Municipal Hospital Karlsruhe gGmbH, Moltkestraße 90, 76133 Karlsruhe, Germany;
| | - Thomas Meyer
- Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany;
| | - Markus Stücker
- Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Gudrunstr. 56, 44791 Bochum, Germany;
| |
Collapse
|
28
|
Rippon M, Rogers AA, Westgate S, Ousey K. Effectiveness of a polyhexamethylene biguanide-containing wound cleansing solution using experimental biofilm models. J Wound Care 2023; 32:359-367. [PMID: 37300862 DOI: 10.12968/jowc.2023.32.6.359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
OBJECTIVE Antiseptics are widely used in wound management to prevent or treat wound infections, and have been shown to have antibiofilm efficacy. The objective of this study was to assess the effectiveness of a polyhexamethylene biguanide (PHMB)-containing wound cleansing and irrigation solution on model biofilm of pathogens known to cause wound infections compared with a number of other antimicrobial wound cleansing and irrigation solutions. METHOD Staphylococcus aureus and Pseudomonas aeruginosa single-species biofilms were cultured using microtitre plate and Centers for Disease Control and Prevention (CDC) biofilm reactor methods. Following a 24-hour incubation period, the biofilms were rinsed to remove planktonic microorganisms and then challenged with wound cleansing and irrigation solutions. Following incubation of the biofilms with a variety of concentrations of the test solutions (50%, 75% or 100%) for 20, 30, 40, 50 or 60 minutes, remaining viable organisms from the treated biofilms were quantified. RESULTS The six antimicrobial wound cleansing and irrigation solutions used were all effective in eradicating Staphylococcus aureus biofilm bacteria in both test models. However, the results were more variable for the more tolerant Pseudomonas aeruginosa biofilm. Only one of the six solutions (sea salt and oxychlorite/NaOCl-containing solution) was able to eradicate Pseudomonas aeruginosa biofilm using the microtitre plate assay. Of the six solutions, three (a solution containing PHMB and poloxamer 188 surfactant, a solution containing hypochlorous acid (HOCl) and a solution containing NaOCl/HOCl) showed increasing levels of eradication of Pseudomonas aeruginosa biofilm microorganisms with increasing concentration and exposure time. Using the CDC biofilm reactor model, all six cleansing and irrigation solutions, except for the solution containing HOCl, were able to eradicate Pseudomonas aeruginosa biofilms such that no viable microorganisms were recovered. CONCLUSION This study demonstrated that a PHMB-containing wound cleansing and irrigation solution was as effective as other antimicrobial wound irrigation solutions for antibiofilm efficacy. Together with the low toxicity, good safety profile and absence of any reported acquisition of bacterial resistance to PHMB, the antibiofilm effectiveness data support the alignment of this cleansing and irrigation solution with antimicrobial stewardship (AMS) strategies.
Collapse
Affiliation(s)
- Mark Rippon
- Visiting Clinical Research Associate, Huddersfield University, Huddersfield, UK
- Medical Marketing Consultant, Daneriver Consultancy Ltd, Holmes Chapel, Cheshire, UK
| | - Alan A Rogers
- Independent Wound Care Consultant, Flintshire, North Wales, UK
| | | | - Karen Ousey
- Professor of Skin Integrity, Director for the Institute of Skin Integrity and Infection Prevention, University of Huddersfield Department of Nursing and Midwifery, Huddersfield, UK
- Adjunct Professor, School of Nursing, Faculty of Health at the Queensland University of Technology, Australia
- Visiting Professor, RCSI, Dublin, Ireland
- Chair IWII
- President Elect ISTAP
| |
Collapse
|
29
|
Kışla D, Gökmen GG, Akdemir Evrendilek G, Akan T, Vlčko T, Kulawik P, Režek Jambrak A, Ozogul F. Recent developments in antimicrobial surface coatings: Various deposition techniques with nanosized particles, their application and environmental concerns. Trends Food Sci Technol 2023. [DOI: 10.1016/j.tifs.2023.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
30
|
Heydarian N, Wouters CL, Neel A, Ferrell M, Panlilio H, Haight T, Gu T, Rice CV. Eradicating Biofilms of Carbapenem-Resistant Enterobacteriaceae by Simultaneously Dispersing the Biomass and Killing Planktonic Bacteria with PEGylated Branched Polyethyleneimine. ChemMedChem 2023; 18:e202200428. [PMID: 36542457 PMCID: PMC9899318 DOI: 10.1002/cmdc.202200428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/21/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Carbapenem-resistant Enterobacteriaceae (CRE) are emerging pathogens that cause variety of severe infections. CRE evade antibiotic treatments because these bacteria produce enzymes that degrade a wide range of antibiotics including carbapenems and β-lactams. The formation of biofilms aggravates CRE infections, especially in a wound environment. These difficulties lead to persistent infection and non-healing wounds. This creates the need for new compounds to overcome CRE antimicrobial resistance and disrupt biofilms. Recent studies in our lab show that 600 Da branched polyethyleneimine (BPEI) and its derivative PEG350-BPEI can overcome antimicrobial resistance and eradicate biofilms in methicillin-resistant S. aureus, methicillin-resistant S. epidermidis, P. aeruginosa, and E. coli. In this study, the ability of 600 Da BPEI and PEG350-BPEI to eradicate carbapenem-resistant Enterobacteriaceae bacteria and their biofilms is demonstrated. We show that both BPEI and PEG350-BPEI have anti-biofilm efficacy against CRE strains expressing Klebsiella pneumoniae carbapenemases (KPCs) and metallo-β-lactamases (MBLs), such as New Delhi MBL (NDM-1). Furthermore, our results illustrate that BPEI affects planktonic CRE bacteria by increasing bacterial length and width from the inability to proceed with normal cell division processes. These data demonstrate the multi-functional properties of 600 Da BPEI and PEG350-BPEI to reduce biofilm formation and mitigate virulence in carbapenem-resistant Enterobacteriaceae.
Collapse
Affiliation(s)
- Neda Heydarian
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019
| | - Cassandra L. Wouters
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019
| | - Andrew Neel
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019
| | - Maya Ferrell
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019
| | - Hannah Panlilio
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019
| | - Tristan Haight
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019
| | - Tingting Gu
- Department of Biology, 730 Van Vleet Oval, Room 314, University of Oklahoma, Norman, OK 73019, USA
| | - Charles V. Rice
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019
| |
Collapse
|
31
|
Hamdan N, Khodir WKWA, Hamid SA, Nasir MHM, Hamzah AS, Cruz-Maya I, Guarino V. PCL/Gelatin/Graphene Oxide Electrospun Nanofibers: Effect of Surface Functionalization on In Vitro and Antibacterial Response. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:488. [PMID: 36770449 PMCID: PMC9921190 DOI: 10.3390/nano13030488] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/09/2023] [Accepted: 01/21/2023] [Indexed: 06/18/2023]
Abstract
The emergence of resistance to pathogenic bacteria has resulted from the misuse of antibiotics used in wound treatment. Therefore, nanomaterial-based agents can be used to overcome these limitations. In this study, polycaprolactone (PCL)/gelatin/graphene oxide electrospun nanofibers (PGO) are functionalized via plasma treatment with the monomeric groups diallylamine (PGO-M1), acrylic acid (PGO-M2), and tert-butyl acrylate (PGO-M3) to enhance the action against bacteria cells. The surface functionalization influences the morphology, surface wettability, mechanical properties, and thermal stability of PGO nanofibers. PGO-M1 and PGO-M2 exhibit good antibacterial activity against Staphylococcus aureus and Escherichia coli, whereas PGO-M3 tends to reduce their antibacterial properties compared to PGO nanofibers. The highest proportion of dead bacteria cells is found on the surface of hydrophilic PGO-M1, whereas live cells are colonized on the surface of hydrophobic PGO-M3. Likewise, PGO-M1 shows a good interaction with L929, which is confirmed by the high levels of adhesion and proliferation with respect to the control. All the results confirm that surface functionalization can be strategically used as a tool to engineer PGO nanofibers with controlled antibacterial properties for the fabrication of highly versatile devices suitable for different applications (e.g., health, environmental pollution).
Collapse
Affiliation(s)
- Nazirah Hamdan
- Department of Chemistry, Kulliyyah of Science, International Islamic University Malaysia Kuantan Campus, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Wan Khartini Wan Abdul Khodir
- Department of Chemistry, Kulliyyah of Science, International Islamic University Malaysia Kuantan Campus, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia
- SYNTOF, Kulliyyah of Science, International Islamic University Malaysia Kuantan Campus, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Shafida Abd Hamid
- Department of Chemistry, Kulliyyah of Science, International Islamic University Malaysia Kuantan Campus, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia
- SYNTOF, Kulliyyah of Science, International Islamic University Malaysia Kuantan Campus, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Mohd Hamzah Mohd Nasir
- Department of Biotechnology, Kulliyyah of Science, International Islamic University Malaysia Kuantan Campus, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia
| | - Ahmad Sazali Hamzah
- Institute of Science, Universiti Teknologi MARA, Shah Alam 40450, Selangor, Malaysia
| | - Iriczalli Cruz-Maya
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Mostra d’Oltremare Pad.20, V.le J.F.Kennedy 54, 80125 Naples, Italy
| | - Vincenzo Guarino
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Mostra d’Oltremare Pad.20, V.le J.F.Kennedy 54, 80125 Naples, Italy
| |
Collapse
|
32
|
Rouillard KR, Markovetz MR, Kissner WJ, Boone WL, Plott LM, Hill DB. Altering the viscoelastic properties of mucus-grown Pseudomonas aeruginosa biofilms affects antibiotic susceptibility. Biofilm 2023; 5:100104. [PMID: 36711323 PMCID: PMC9880403 DOI: 10.1016/j.bioflm.2023.100104] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023] Open
Abstract
The viscoelastic properties of biofilms are correlated with their susceptibility to mechanical and chemical stress, and the airway environment in muco-obstructive pulmonary diseases (MOPD) facilitates robust biofilm formation. Hyperconcentrated, viscoelastic mucus promotes chronic inflammation and infection, resulting in increased mucin and DNA concentrations. The viscoelastic properties of biofilms are regulated by biopolymers, including polysaccharides and DNA, and influence responses to antibiotics and phagocytosis. We hypothesize that targeted modulation of biofilm rheology will compromise structural integrity and increase antibiotic susceptibility and mucociliary transport. We evaluate biofilm rheology on the macro, micro, and nano scale as a function of treatment with a reducing agent, a biopolymer, and/or tobramycin to define the relationship between the viscoelastic properties of biofilms and susceptibility. Disruption of the biofilm architecture is associated with altered macroscopic and microscopic moduli, rapid vector permeability, increased antibiotic susceptibility, and improved mucociliary transport, suggesting that biofilm modulating therapeutics will improve the treatment of chronic respiratory infections in MOPD.
Collapse
Affiliation(s)
- Kaitlyn R. Rouillard
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Matthew R. Markovetz
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - William J. Kissner
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - William L. Boone
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lucas M. Plott
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - David B. Hill
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Joint Department of Biomedical Engineering, North Carolina State University and the University of North Carolina, Chapel Hill, NC, 27599, USA,Department of Physics and Astronomy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Corresponding author. Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
33
|
Jiang F, Cai C, Gao L, Su X, Han S. Peptidoglycan-Directed Chemical Ligation for Selective Inhibition on Gram-Positive Bacteria. ACS OMEGA 2023; 8:2485-2490. [PMID: 36687063 PMCID: PMC9850734 DOI: 10.1021/acsomega.2c06964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Microbicides with distinct antibacterial mechanisms show potential to combat multi-drug resistance bacteria. We herein report peptidoglycan-directed chemical ligation (PGCL) between alkyne-bearing vancomycin and an azide-tagged cationic polymer. The former binds peptidoglycan and inhibits peptidoglycan crosslinking, while the latter interferes the integrity of the bacterial membrane. PGCL results in enhanced bactericidal activity against Gram-positive Staphylococcus aureus (S. aureus) over Gram-negative Escherichia coli (E. coli). These data indicate the potential of PGCL to selectively and synergistically inhibit Gram-positive pathogens via dual modality antibacterial mechanisms of peptidoglycan-inhibiting antibiotics and bacterial membrane-disrupting polycations.
Collapse
Affiliation(s)
- Feng Jiang
- Department
of Chemical Biology, College of Chemistry and Chemical Engineering,
State Key Laboratory for Physical Chemistry of Solid Surfaces, State
Key Laboratory of Cellular Stress Biology, the Key Laboratory for
Chemical Biology of Fujian Province, and the MOE Key Laboratory of
Spectrochemical Analysis & Instrumentation, Xiamen University, Xiamen 361005, China
| | - Chengteng Cai
- Department
of Chemical Biology, College of Chemistry and Chemical Engineering,
State Key Laboratory for Physical Chemistry of Solid Surfaces, State
Key Laboratory of Cellular Stress Biology, the Key Laboratory for
Chemical Biology of Fujian Province, and the MOE Key Laboratory of
Spectrochemical Analysis & Instrumentation, Xiamen University, Xiamen 361005, China
| | - Lei Gao
- Department
of Chemical Biology, College of Chemistry and Chemical Engineering,
State Key Laboratory for Physical Chemistry of Solid Surfaces, State
Key Laboratory of Cellular Stress Biology, the Key Laboratory for
Chemical Biology of Fujian Province, and the MOE Key Laboratory of
Spectrochemical Analysis & Instrumentation, Xiamen University, Xiamen 361005, China
| | - Xinhui Su
- PET
center, Department of Nuclear Medicine, The First Affiliated Hospital,
College of Medicine, Zhejiang University, Hangzhou 310027, China
| | - Shoufa Han
- Department
of Chemical Biology, College of Chemistry and Chemical Engineering,
State Key Laboratory for Physical Chemistry of Solid Surfaces, State
Key Laboratory of Cellular Stress Biology, the Key Laboratory for
Chemical Biology of Fujian Province, and the MOE Key Laboratory of
Spectrochemical Analysis & Instrumentation, Xiamen University, Xiamen 361005, China
| |
Collapse
|
34
|
Caputo WJ, Monterosa P, Beggs D. Antibiotic Misuse in Wound Care: Can Bacterial Localization through Fluorescence Imaging Help? Diagnostics (Basel) 2022; 12:3207. [PMID: 36553214 PMCID: PMC9778012 DOI: 10.3390/diagnostics12123207] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
(1) Background: Systemic antibiotic use in chronic wounds is alarmingly high worldwide. Between 53% to 71% of patients are prescribed at least one course per chronic wound. Systemic antibiotic use should follow antibiotic stewardship guidelines and ought to be reserved for situations where their use is deemed supported by clinical indications. Unfortunately, in the field of wound care, indiscriminate and often inadequate use of systemic antibiotics is leading to both patient complications and worsening antibiotic resistance rates. Implementing novel tools that help clinicians prevent misuse or objectively determine the true need for systemic antibiotics is essential to reduce prescribing rates. (2) Methods: We present a compendium of available systemic antibiotic prescription rates in chronic wounds. The impact of various strategies used to improve these rates, as well as preliminary data on the impact of implementing fluorescence imaging technology to finesse wound status diagnosis, are presented. (3) Results: Interventions including feedback from wound care surveillance and treatment data registries as well as better diagnostic strategies can ameliorate antibiotic misuse. (4) Conclusions: Interventions that mitigate unnecessary antibiotic use are needed. Effective strategies include those that raise awareness of antibiotic overprescribing and those that enhance diagnosis of infection, such as fluorescence imaging.
Collapse
Affiliation(s)
- Wayne J. Caputo
- Director of the Wound Care Center at Clara Maass Medical Center, Belleville, NJ 07109, USA
| | | | - Donald Beggs
- Infectious Disease, Clara Maass Medical Center, Belleville, NJ 07109, USA
| |
Collapse
|
35
|
Ratia C, Sueiro S, Soengas RG, Iglesias MJ, López-Ortiz F, Soto SM. Gold(III) Complexes Activity against Multidrug-Resistant Bacteria of Veterinary Significance. Antibiotics (Basel) 2022; 11:antibiotics11121728. [PMID: 36551386 PMCID: PMC9774617 DOI: 10.3390/antibiotics11121728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
The emergence and spread of multidrug-resistant bacteria are a global concern. The lack of new antibiotics in the pipeline points to the need for developing new strategies. In this sense, gold(III) complexes (G3Cs) could be a promising alternative due to their recently described antibacterial activity. The aim of this study was to evaluate the antimicrobial activity of G3Cs alone and in combination with colistin against pathogenic bacteria from veterinary sources. Minimal inhibitory concentration (MIC) values were determined by broth microdilution and compared with clinically relevant antibiotics. Antibiofilm activity was determined by crystal violet staining. Combinations of selected G3Cs with colistin and cytotoxicity in commercial human cell lines were evaluated. Four and seven G3Cs showed antibacterial effect against Gram-negative and Gram-positive strains, respectively, with this activity being higher among Gram-positive strains. The G3Cs showed antibiofilm activity against Gram-negative species at concentrations similar or one to four folds higher than the corresponding MICs. Combination of G3Cs with colistin showed a potential synergistic antibacterial effect reducing concentrations and toxicity of both agents. The antimicrobial and antibiofilm activity, the synergistic effect when combined with colistin and the in vitro toxicity suggest that G3Cs would provide a new therapeutic alternative against multidrug-resistant bacteria from veterinary origin.
Collapse
Affiliation(s)
- Carlos Ratia
- ISGlobal, Hospital Clínic—Universitat de Barcelona, 08036 Barcelona, Spain
| | - Sara Sueiro
- ISGlobal, Hospital Clínic—Universitat de Barcelona, 08036 Barcelona, Spain
| | - Raquel G. Soengas
- Departamento de Química Orgánica e Inorgánica, Universidad de Oviedo, Julián Clavería 7, 33006 Oviedo, Spain
| | - María José Iglesias
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, 04120 Almería, Spain
| | - Fernando López-Ortiz
- Área de Química Orgánica, Centro de Investigación CIAIMBITAL, Universidad de Almería, 04120 Almería, Spain
- Correspondence: (F.L.-O.); (S.M.S.)
| | - Sara María Soto
- ISGlobal, Hospital Clínic—Universitat de Barcelona, 08036 Barcelona, Spain
- CIBER Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (F.L.-O.); (S.M.S.)
| |
Collapse
|
36
|
Li J, Fan Q, Zuo J, Xue B, Zhang X, Wei Y, Sun L, Grenier D, Yi L, Hou X, Wang Y. Paeoniflorin combined with norfloxacin ameliorates drug-resistant Streptococcus suis infection. J Antimicrob Chemother 2022; 77:3275-3282. [PMID: 36173390 DOI: 10.1093/jac/dkac313] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 08/29/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The increased resistance of bacterial pathogens to fluoroquinolones (FQs), such as norfloxacin and ciprofloxacin, supports the need to develop new antibacterial drugs and combination therapies using conventional antibiotics. The LuxS/AI-2 quorum sensing (QS) system can regulate the complex group behaviour of Streptococcus suis and impact its susceptibility to FQs. OBJECTIVES We investigated the combination of paeoniflorin and norfloxacin as a novel and effective strategy against FQ-resistant S. suis. METHODS FIC, AI-2 activity assay, real-time RT-PCR and biofilm inhibition assays were performed to investigate the in vitro effect of paeoniflorin combined with norfloxacin. Mouse protection and mouse anti-infection assays were performed to investigate the in vivo effect of paeoniflorin combined with norfloxacin. RESULTS FIC results showed that paeoniflorin and norfloxacin exert a synergistic bactericidal effect. Evidence was brought that paeoniflorin reduces the S. suis AI-2 activity and significantly down-regulates the transcription of the FQ efflux pump gene. In addition, paeoniflorin can inhibit biofilm formation, thereby promoting the ability of norfloxacin to kill S. suis. Finally, we showed in a mouse model that paeoniflorin in association with norfloxacin is effective to treat S. suis infections. CONCLUSIONS This study highlighted the inhibitory potential of paeoniflorin on the LuxS/AI-2 QS system of S. suis, and provided evidence that it can inhibit the FQ efflux pump and prevent biofilm formation to cooperate with norfloxacin in the treatment of resistant S. suis-related infections.
Collapse
Affiliation(s)
- Jinpeng Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| | - Qingying Fan
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| | - Jing Zuo
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| | - Bingqian Xue
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| | - Xiaoling Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| | - Ying Wei
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| | - Liyun Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, Quebec, Canada
| | - Li Yi
- Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China.,College of Life Science, Luoyang Normal University, Luoyang, Henan, China
| | - Xiaogai Hou
- College of Agriculture/College of Tree Peony, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, China.,Key Laboratory of Molecular Pathogen and Immunology of Animal of Luoyang, Luoyang, Henan, China
| |
Collapse
|
37
|
Yang X, Wang B, Peng D, Nie X, Wang J, Yu CY, Wei H. Hyaluronic Acid‐Based Injectable Hydrogels for Wound Dressing and Localized Tumor Therapy: A Review. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200124] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Xu Yang
- Postdoctoral Mobile Station of Basic Medical Sciences Hengyang Medical School University of South China Hengyang 421001 China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science University of South China Hengyang Hunan 421001 China
| | - Bin Wang
- Postdoctoral Mobile Station of Basic Medical Sciences Hengyang Medical School University of South China Hengyang 421001 China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science University of South China Hengyang Hunan 421001 China
| | - Dongdong Peng
- Postdoctoral Mobile Station of Basic Medical Sciences Hengyang Medical School University of South China Hengyang 421001 China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science University of South China Hengyang Hunan 421001 China
| | - Xiaobo Nie
- Postdoctoral Mobile Station of Basic Medical Sciences Hengyang Medical School University of South China Hengyang 421001 China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science University of South China Hengyang Hunan 421001 China
| | - Jun Wang
- Postdoctoral Mobile Station of Basic Medical Sciences Hengyang Medical School University of South China Hengyang 421001 China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science University of South China Hengyang Hunan 421001 China
| | - Cui-Yun Yu
- Postdoctoral Mobile Station of Basic Medical Sciences Hengyang Medical School University of South China Hengyang 421001 China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science University of South China Hengyang Hunan 421001 China
| | - Hua Wei
- Postdoctoral Mobile Station of Basic Medical Sciences Hengyang Medical School University of South China Hengyang 421001 China
- Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science University of South China Hengyang Hunan 421001 China
| |
Collapse
|
38
|
Islam S, Mahmud ML, Almalki WH, Biswas S, Islam MA, Mortuza MG, Hossain MA, Ekram MAE, Uddin MS, Zaman S, Saleh MA. Cell-Free Supernatants (CFSs) from the Culture of Bacillus subtilis Inhibit Pseudomonas sp. Biofilm Formation. Microorganisms 2022; 10:2105. [PMID: 36363697 PMCID: PMC9692604 DOI: 10.3390/microorganisms10112105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/12/2022] [Accepted: 10/16/2022] [Indexed: 05/29/2024] Open
Abstract
Biofilm inhibition has been identified as a novel drug target for the development of broad-spectrum antibiotics to combat infections caused by drug-resistant bacteria. Although several plant-based compounds have been reported to have anti-biofilm properties, research on the anti-biofilm properties of bacterial bioactive compounds has been sparse. In this study, the efficacy of compounds from a cell-free supernatant of Bacillus subtilis against a biofilm formation of Pseudomonas sp. was studied through in vitro, in vivo and in silico studies. Here, in well diffusion method, Bacillus subtilis demonstrated antibacterial activity, and more than 50% biofilm inhibition activity against Pseudomonas sp. was exhibited through in vitro studies. Moreover, molecular docking and molecular dynamics (MD) simulation gave insights into the possible mode of action of the bacterial volatile compounds identified through GC-MS to inhibit the biofilm-formation protein (PDB ID: 7M1M) of Pseudomonas sp. The binding energy revealed from docking studies ranged from -2.3 to -7.0 kcal mol-1. Moreover, 1-(9H-Fluoren-2-yl)-2-(1-phenyl-1H-ttetrazole5-ylsulfanyl)-ethanone was found to be the best-docked compound through ADMET and pharmacokinetic properties. Furthermore, MD simulations further supported the in vitro studies and formed a stable complex with the tested protein. Thus, this study gives an insight into the development of new antibiotics to combat multi-drug-resistant bacteria.
Collapse
Affiliation(s)
- Shirmin Islam
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Liton Mahmud
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Waleed H. Almalki
- Department of Pharmacology and Toxicology, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Suvro Biswas
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Ariful Islam
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Golam Mortuza
- Department of Science and Humanities, Bangladesh Army International University of Science and Technology, Cumilla 3500, Bangladesh
| | - Mohammad Akbar Hossain
- Department of Pharmacology and Toxicology, Faculty of Medicine in Al-Qunfudah, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Md. Akhtar-E Ekram
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Salah Uddin
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Shahriar Zaman
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Md. Abu Saleh
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| |
Collapse
|
39
|
Rembe JD, Thompson VD, Stuermer EK. Antimicrobials cetylpyridinium-chloride and miramistin demonstrate non-inferiority and no "protein-error" compared to established wound care antiseptics in vitro. AIMS Microbiol 2022; 8:372-387. [PMID: 36694590 PMCID: PMC9834083 DOI: 10.3934/microbiol.2022026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/02/2022] [Accepted: 10/17/2022] [Indexed: 01/26/2023] Open
Abstract
Concern about microbial tolerance and resistance to established antimicrobials drives research into alternatives for local antiseptic wound treatment. Precise efficacy profiles are thereby important in the evaluation of potential alternative antimicrobials, and protein interference ("protein error") is a key factor. Here, the antimicrobial efficacy of cetylpyridinium chloride (CPC) and miramistin (MST) was compared to the established antimicrobials octenidine (OCT), povidon-iodine (PVP-I), polyhexamethylene-biguanide (PHMB) and chlorhexidine (CHX). Efficacy was evaluated after 0.5, 1, 3, 5 and 10 min against Staphylococcus aureus, Pseudomonas aeruginosa, Escherichia coli, Enterococcus faecium and Candida albicans using an in vitro quantitative suspension method (based on DIN EN 13727). To investigate protein interference, 0.3% or 3% bovine albumin was used as the challenge. OCT and PVP-I demonstrated a significant efficacy within 0.5 min, regardless of the microbial organism and protein challenge (p < 0.01). CPC and MST showed no inferiority in efficacy, with only MST needing up to 3 min to achieve the same microbial reduction. PHMB and CHX also achieved significant reduction rates over the tested time-course, yet demonstrated a necessity for prolonged exposure (up to 10 min) for comparable reduction. A protein interference was predominantly observed for PHMB against S. aureus, but without statistically significant differences in antimicrobial efficacy between the 0.3% and 3% protein challenges. All other tested agents showed no relevant interference with the presence of protein. CPC and MST proved to be non-inferior to established wound antiseptics agents in vitro. In fact, CPC showed a more efficient reduction than PHMB and CHX despite there being an introduced protein challenge. Both agents demonstrated no significant "protein error" under challenging conditions (3% albumin), posing them as valid potential candidates for alternative antimicrobials in wound management.
Collapse
Affiliation(s)
- Julian-Dario Rembe
- Department of Vascular and Endovascular Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University, Duesseldorf, North-Rhine Westphalia, Germany
| | - Vivian-Denise Thompson
- Chair for Translational Wound Research, Center for Biomedical Education and Research, Witten/Herdecke University, Witten, North-Rhine Westphalia, Germany
| | - Ewa Klara Stuermer
- Department of Vascular Medicine, University Heart Center, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
40
|
Burnet M, Metcalf DG, Milo S, Gamerith C, Heinzle A, Sigl E, Eitel K, Haalboom M, Bowler PG. A Host-Directed Approach to the Detection of Infection in Hard-to-Heal Wounds. Diagnostics (Basel) 2022; 12:2408. [PMID: 36292097 PMCID: PMC9601189 DOI: 10.3390/diagnostics12102408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 01/08/2023] Open
Abstract
Wound infection is traditionally defined primarily by visual clinical signs, and secondarily by microbiological analysis of wound samples. However, these approaches have serious limitations in determining wound infection status, particularly in early phases or complex, chronic, hard-to-heal wounds. Early or predictive patient-derived biomarkers of wound infection would enable more timely and appropriate intervention. The observation that immune activation is one of the earliest responses to pathogen activity suggests that immune markers may indicate wound infection earlier and more reliably than by investigating potential pathogens themselves. One of the earliest immune responses is that of the innate immune cells (neutrophils) that are recruited to sites of infection by signals associated with cell damage. During acute infection, the neutrophils produce oxygen radicals and enzymes that either directly or indirectly destroy invading pathogens. These granular enzymes vary with cell type but include elastase, myeloperoxidase, lysozyme, and cathepsin G. Various clinical studies have demonstrated that collectively, these enzymes, are sensitive and reliable markers of both early-onset phases and established infections. The detection of innate immune cell enzymes in hard-to-heal wounds at point of care offers a new, simple, and effective approach to determining wound infection status and may offer significant advantages over uncertainties associated with clinical judgement, and the questionable value of wound microbiology. Additionally, by facilitating the detection of early wound infection, prompt, local wound hygiene interventions will likely enhance infection resolution and wound healing, reduce the requirement for systemic antibiotic therapy, and support antimicrobial stewardship initiatives in wound care.
Collapse
Affiliation(s)
- Michael Burnet
- Synovo GmbH, Paul Ehrlich Straße 15, 72076 Tuebingen, Germany
| | - Daniel G. Metcalf
- ConvaTec Ltd., First Avenue, Deeside Industrial Park, Deeside CH5 2NU, UK
| | - Scarlet Milo
- ConvaTec Ltd., First Avenue, Deeside Industrial Park, Deeside CH5 2NU, UK
| | - Clemens Gamerith
- Austrian Centre of Industrial Biotechnology, Krennagsse 37, A-8010 Graz, Austria
| | - Andrea Heinzle
- Qualizyme Diagnostics GmbH & Co. KG, Neue Stiftingtalstrasse 2, A-8010 Graz, Austria
| | - Eva Sigl
- Qualizyme Diagnostics GmbH & Co. KG, Neue Stiftingtalstrasse 2, A-8010 Graz, Austria
| | - Kornelia Eitel
- Synovo GmbH, Paul Ehrlich Straße 15, 72076 Tuebingen, Germany
| | - Marieke Haalboom
- Medical School Twente, Medisch Spectrum Twente, 7512 KZ Enschede, The Netherlands
| | | |
Collapse
|
41
|
Huang C, Xu X, Fu J, Yu DG, Liu Y. Recent Progress in Electrospun Polyacrylonitrile Nanofiber-Based Wound Dressing. Polymers (Basel) 2022; 14:3266. [PMID: 36015523 PMCID: PMC9415690 DOI: 10.3390/polym14163266] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 02/07/2023] Open
Abstract
Bleeding control plays a very important role in worldwide healthcare, which also promotes research and development of wound dressings. The wound healing process involves four stages of hemostasis, inflammation, proliferation and remodeling, which is a complex process, and wound dressings play a huge role in it. Electrospinning technology is simple to operate. Electrospun nanofibers have a high specific surface area, high porosity, high oxygen permeability, and excellent mechanical properties, which show great utilization value in the manufacture of wound dressings. As one of the most popular reactive and functional synthetic polymers, polyacrylonitrile (PAN) is frequently explored to create nanofibers for a wide variety of applications. In recent years, researchers have invested in the application of PAN nanofibers in wound dressings. Research on spun nanofibers is reviewed, and future development directions and prospects of electrospun PAN nanofibers for wound dressings are proposed.
Collapse
Affiliation(s)
- Chang Huang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xizi Xu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Junhao Fu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Deng-Guang Yu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yanbo Liu
- School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, China
| |
Collapse
|
42
|
Preliminary Study on Phytochemical Constituents and Biological Activities of Essential Oil from Myriactis nepalensis Less. Molecules 2022; 27:molecules27144631. [PMID: 35889501 PMCID: PMC9324352 DOI: 10.3390/molecules27144631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
In response to the need for novel therapeutic strategies to combat the development of microbial resistance, plant essential oils may represent a promising alternative source. This study set out to characterize the chemical composition and assess the antibacterial potential of Myriactis nepalensis Less. essential oil (MNEO). Essential oil isolated from M. nepalensis by hydrodistillation was analyzed using a GC–MS technique. The antibacterial properties of MNEO alone and combined with antibiotics (chloramphenicol and streptomycin) were tested via the disc diffusion, microbroth dilution, and checkerboard methods. MNEO was represented by oxygenated sesquiterpenes (60.3%) and sesquiterpene hydrocarbons (28.6%), with caryophyllene oxide, spathulenol, humulene epoxide II, β-elemene, neointermedeol, and β-caryophyllene as the main compounds. MNEO exhibited a strong antibacterial effect against Gram-positive bacteria, with MIC and MBC values of 0.039 mg/mL and 0.039–0.156 mg/mL, respectively, and synergistic effects were observed in both combinations with chloramphenicol and streptomycin. Furthermore, the antibiofilm and cytotoxic activities of MNEO were also evaluated. The crystal violet assay was used for quantification of Staphylococcus aureus biofilm formation, and an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was conducted to determine cell viability. The results revealed MNEO could dose-dependently inhibit Staphylococcus aureus biofilm formation and possessed potential cytotoxic on both normal and cancer cells (IC50 values from 13.13 ± 1.90 to 35.22 ± 8.36 μg/mL). Overall, the results indicate that MNEO may have promising applications in the field of bacterial infections.
Collapse
|
43
|
Gobin M, Proust R, Lack S, Duciel L, Des Courtils C, Pauthe E, Gand A, Seyer D. A Combination of the Natural Molecules Gallic Acid and Carvacrol Eradicates P. aeruginosa and S. aureus Mature Biofilms. Int J Mol Sci 2022; 23:ijms23137118. [PMID: 35806123 PMCID: PMC9266711 DOI: 10.3390/ijms23137118] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 01/18/2023] Open
Abstract
Wound infection, especially the development of bacterial biofilms, delays wound healing and is a major public health concern. Bacteria in biofilms are more tolerant to antimicrobial agents, and new treatments to eradicate mature biofilms are needed. Combining antimicrobial molecules with different mechanisms of action is an attractive strategy to tackle the heterogeneous nature of microbial communities in biofilms. This study focused on three molecules of natural origin: gallic acid (G), carvacrol (K) and curcumin (Q). Their abilities, individually or in combination, to eradicate biofilms were quantified on mono- and dual-species mature biofilms of Pseudomonas aeruginosa and Staphylococcus aureus, the strains most commonly found in infected wounds. G presented biofilm eradicating activity on P. aeruginosa, whereas K had biofilm eradicating activity on S. aureus and P. aeruginosa. Q had no potent biofilm eradicating activity. The combination of G and K increased the effects previously observed on P. aeruginosa biofilm and led to complete eradication of S. aureus biofilm. This combination was also efficient in eradicating a dual-species biofilm of S. aureus and P. aeruginosa. This work demonstrates that K and G used in combination have a strong and synergistic eradicating activity on both mono- and dual-species mature biofilms of S. aureus and P. aeruginosa and may therefore represent an efficient alternative for the treatment of biofilms in wounds.
Collapse
Affiliation(s)
- Maxime Gobin
- ERRMECe Laboratory, 1 rue Descartes, CY Cergy Paris University, 95031 Neuville sur Oise, France; (M.G.); (E.P.); (A.G.)
| | - Richard Proust
- Les Laboratoires BROTHIER, 41 Rue de Neuilly, 92735 Nanterre, France; (R.P.); (S.L.); (L.D.); (C.D.C.)
| | - Stéphane Lack
- Les Laboratoires BROTHIER, 41 Rue de Neuilly, 92735 Nanterre, France; (R.P.); (S.L.); (L.D.); (C.D.C.)
| | - Laura Duciel
- Les Laboratoires BROTHIER, 41 Rue de Neuilly, 92735 Nanterre, France; (R.P.); (S.L.); (L.D.); (C.D.C.)
| | - Céline Des Courtils
- Les Laboratoires BROTHIER, 41 Rue de Neuilly, 92735 Nanterre, France; (R.P.); (S.L.); (L.D.); (C.D.C.)
| | - Emmanuel Pauthe
- ERRMECe Laboratory, 1 rue Descartes, CY Cergy Paris University, 95031 Neuville sur Oise, France; (M.G.); (E.P.); (A.G.)
| | - Adeline Gand
- ERRMECe Laboratory, 1 rue Descartes, CY Cergy Paris University, 95031 Neuville sur Oise, France; (M.G.); (E.P.); (A.G.)
| | - Damien Seyer
- ERRMECe Laboratory, 1 rue Descartes, CY Cergy Paris University, 95031 Neuville sur Oise, France; (M.G.); (E.P.); (A.G.)
- Correspondence:
| |
Collapse
|
44
|
Neutralizing Staphylococcus aureus Virulence with AZD6389, a Three mAb Combination, Accelerates Closure of a Diabetic Polymicrobial Wound. mSphere 2022; 7:e0013022. [PMID: 35642538 PMCID: PMC9241520 DOI: 10.1128/msphere.00130-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Nonhealing diabetic foot ulcers (DFU), a major complication of diabetes, are associated with high morbidity and mortality despite current standard of care. Since Staphylococcus aureus is the most common pathogen isolated from nonhealing and infected DFU, we hypothesized that S. aureus virulence factors would damage tissue, promote immune evasion and alter the microbiome, leading to bacterial persistence and delayed wound healing. In a diabetic mouse polymicrobial wound model with S. aureus, Pseudomonas aeruginosa, and Streptococcus pyogenes, we report a rapid bacterial proliferation, prolonged pro-inflammatory response and large necrotic lesions unclosed for up to 40 days. Treatment with AZD6389, a three-monoclonal antibody combination targeting S. aureus alpha toxin, 4 secreted leukotoxins, and fibrinogen binding cell-surface adhesin clumping factor A resulted in full skin re-epithelization 21 days after inoculation. By neutralizing multiple virulence factors, AZD6389 effectively blocked bacterial agglutination and S. aureus-mediated cell killing, abrogated S. aureus-mediated immune evasion and targeted the bacteria for opsonophagocytic killing. Neutralizing S. aureus virulence not only facilitated S. aureus clearance in lesions, but also reduced S. pyogenes and P. aeruginosa numbers, damaging inflammatory mediators and markers for neutrophil extracellular trap formation 14 days post initiation. Collectively, our data suggest that AZD6389 holds promise as an immunotherapeutic approach against DFU complications. IMPORTANCE Diabetic foot ulcers (DFU) represent a major complication of diabetes and are associated with poor quality of life and increased morbidity and mortality despite standard of care. They have a complex pathogenesis starting with superficial skin lesions, which often progress to deeper tissue structures up to the bone and ultimately require limb amputation. The skin microbiome of diabetic patients has emerged as having an impact on DFU occurrence and chronicity. DFU are mostly polymicrobial, and the Gram-positive bacterium Staphylococcus aureus detected in more than 95% of cases. S. aureus possess a collection of virulence factors which participate in disease progression and may facilitate growth of other pathogens. Here we show in a diabetic mouse wound model that targeting some specific S. aureus virulence factors with a multimechanistic antibody combination accelerated wound closure and promoted full skin re-epithelization. This work opens promising new avenues for the treatment of DFU.
Collapse
|
45
|
From the environment to the hospital: how plants can help to fight bacteria biofilm. Microbiol Res 2022; 261:127074. [DOI: 10.1016/j.micres.2022.127074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/24/2022]
|
46
|
Kaur H, Kaur A, Soni SK, Rishi P. Microbially-derived cocktail of carbohydrases as an anti-biofouling agents: a 'green approach'. BIOFOULING 2022; 38:455-481. [PMID: 35673761 DOI: 10.1080/08927014.2022.2085566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Enzymes, also known as biocatalysts, display vital properties like high substrate specificity, an eco-friendly nature, low energy inputs, and cost-effectiveness. Among their numerous known applications, enzymes that can target biofilms or their components are increasingly being investigated for their anti-biofouling action, particularly in healthcare, food manufacturing units and environmental applications. Enzymes can target biofilms at different levels like during the attachment of microorganisms, formation of exopolymeric substances (EPS), and their disruption thereafter. In this regard, a consortium of carbohydrases that can target heterogeneous polysaccharides present in the EPS matrix may provide an effective alternative to conventional chemical anti-biofouling methods. Further, for complete annihilation of biofilms, enzymes can be used alone or in conjunction with other antimicrobial agents. Enzymes hold the promise to replace the conventional methods with greener, more economical, and more efficient alternatives. The present article explores the potential and future perspectives of using carbohydrases as effective anti-biofilm agents.
Collapse
Affiliation(s)
- Harmanpreet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Arashdeep Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
47
|
Volatile Organic Compounds in the Early Diagnosis of Non-healing Surgical Wounds: A Systematic Review. World J Surg 2022; 46:1669-1677. [DOI: 10.1007/s00268-022-06548-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2022] [Indexed: 11/27/2022]
|
48
|
Thorn CR, Wignall A, Kopecki Z, Kral A, Prestidge CA, Thomas N. Liquid Crystal Nanoparticles Enhance Tobramycin Efficacy in a Murine Model of Pseudomonas aeruginosa Biofilm Wound Infection. ACS Infect Dis 2022; 8:841-854. [PMID: 35255215 DOI: 10.1021/acsinfecdis.1c00606] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chronic Pseudomonas aeruginosa wound infections are highly prevalent and often untreatable due to biofilm formation, resulting in high antimicrobial tolerance. Standard antibiotic therapy for P. aeruginosa infections involves tobramycin, yet it is highly ineffective as monotherapy as tobramycin cannot penetrate the biofilm to elicit its antimicrobial effect. Lipid liquid crystal nanoparticles (LCNPs) have previously been shown to increase the antimicrobial efficacy and penetration of tobramycin against P. aeruginosa biofilms in vitro and ex vivo. Here, for the first time, we have developed a chronic P. aeruginosa biofilm infection in full-thickness wounds in mice to examine the potential of LCNPs to improve the effect of tobramycin, preclinically. After three doses, administered once a day, tobramycin-LCNPs significantly reduced the P. aeruginosa bacterial load in murine wounds 1000-fold more than unformulated tobramycin, which in turn showed no significant difference to the saline control treatment. Consistent with the improved P. aeruginosa eradication, the tobramycin-LCNPs promoted wound healing. In comparison to previous in vitro and ex vivo data, we show a strong in vitro-in vivo correlation between P. aeruginosa biofilm infection models. The enhanced activity of tobramycin-LCNPs in vivo in the preclinical murine model demonstrates the strong potential of LCNPs as a next-generation formulation approach to improve the efficacy of tobramycin against P. aeruginosa biofilm wound infections.
Collapse
Affiliation(s)
- Chelsea R. Thorn
- University of South Australia, Clinical and Health Science, North Tce, Adelaide, SA 5000, Australia
- The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia
- Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, SA 5000, Australia
| | - Anthony Wignall
- University of South Australia, Clinical and Health Science, North Tce, Adelaide, SA 5000, Australia
| | - Zlatko Kopecki
- University of South Australia, Clinical and Health Science, North Tce, Adelaide, SA 5000, Australia
- Future Industries Institute, UniSA, Mawson Lakes, SA 5095, Australia
| | - Anita Kral
- University of South Australia, Clinical and Health Science, North Tce, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, S.A. Pathology and University of South Australia, Adelaide, SA 5000, Australia
| | - Clive A. Prestidge
- University of South Australia, Clinical and Health Science, North Tce, Adelaide, SA 5000, Australia
| | - Nicky Thomas
- University of South Australia, Clinical and Health Science, North Tce, Adelaide, SA 5000, Australia
- The Basil Hetzel Institute for Translational Health Research, Woodville, SA 5011, Australia
- Biofilm Test Facility, Cancer Research Institute, University of South Australia, North Tce, Adelaide, SA 5000, Australia
| |
Collapse
|
49
|
Shin MK, Lee B, Kim ST, Yoo JS, Sung JS. Designing a Novel Functional Peptide With Dual Antimicrobial and Anti-inflammatory Activities via in Silico Methods. Front Immunol 2022; 13:821070. [PMID: 35432369 PMCID: PMC9010562 DOI: 10.3389/fimmu.2022.821070] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/10/2022] [Indexed: 01/31/2023] Open
Abstract
As spider venom is composed of various bioactive substances, it can be utilized as a platform for discovering future therapeutics. Host defense peptides are great candidates for developing novel antimicrobial agents due to their multifunctional properties. In this study, novel functional peptides were rationally designed to have dual antibacterial and anti-inflammatory activities with high cytocompatibility. Based on a template sequence from the transcriptome of spider Agelena koreana, a series of via in silico analysis were conducted, incorporating web-based machine learning tools along with the alteration of amino acid residues. Two peptides, Ak-N’ and Ak-N’m, were designed and were subjected to functional validation. The peptides inhibited gram-negative and gram-positive bacteria by disrupting the outer and bacterial cytoplasmic membrane. Moreover, the peptides down-regulated the expression of pro-inflammatory mediators, tumor necrosis factor-α, interleukin (IL)-1β, and IL6. Along with low cytotoxicity, Ak-N’m was shown to interact with macrophage surface receptors, inhibiting both Myeloid differentiation primary response 88-dependent and TIR-domain-containing adapter-inducing interferon-β-dependent pathways of Toll-like receptor 4 signaling on lipopolysaccharide-stimulated THP-1-derived macrophages. Here, we rationally designed functional peptides based on the suggested in silico strategy, demonstrating new insights for utilizing biological resources as well as developing therapeutic agents with enhanced properties.
Collapse
Affiliation(s)
- Min Kyoung Shin
- Department of Life Science, Dongguk University-Seoul, Goyang, South Korea
| | - Byungjo Lee
- Department of Life Science, Dongguk University-Seoul, Goyang, South Korea
| | - Seung Tae Kim
- Life and Environment Research Institute, Konkuk University, Seoul, South Korea
| | - Jung Sun Yoo
- Animal Resources Division, National Institute of Biological Resources, Incheon, South Korea
| | - Jung-Suk Sung
- Department of Life Science, Dongguk University-Seoul, Goyang, South Korea
- *Correspondence: Jung-Suk Sung,
| |
Collapse
|
50
|
Zhang Y, Yu J, Zhang H, Li Y, Wang L. Nanofibrous dressing: Potential alternative for fighting against antibiotic‐resistance wound infections. J Appl Polym Sci 2022. [DOI: 10.1002/app.52178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yingjie Zhang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles Donghua University Shanghai China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology Donghua University Shanghai China
| | - Juan Yu
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles Donghua University Shanghai China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology Donghua University Shanghai China
| | - Huiru Zhang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles Donghua University Shanghai China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology Donghua University Shanghai China
| | - Yan Li
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles Donghua University Shanghai China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology Donghua University Shanghai China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles Donghua University Shanghai China
- Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology Donghua University Shanghai China
| |
Collapse
|