1
|
Guo YP, Wang ZX, Guo SL. Giant cutaneous ulcer in Epstein-Barr virus positive T-cell/NK-cell lymphoproliferative disorder: A case report. World J Clin Cases 2025; 13:104258. [DOI: 10.12998/wjcc.v13.i22.104258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/30/2025] [Accepted: 04/15/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Epstein-Barr virus (EBV)-positive T-cell/natural killer (NK)-cell lymphoproliferative disorder is a rare but challenging condition that requires multidisciplinary teamwork, including co-management by infection medicine, radiotherapy, rehabilitation, and psychology experts, as well as wound specialist nurses.
CASE SUMMARY The patient was a 33-year-old female who presented with an erythema-like lesion on the left upper extremity that became desquamated and then blistered and eventually became a giant ulcer with exposed nerves and muscles. The left wrist had a fixed posture, and pathology tests showed cutaneous EBV-positive NK/T-cell proliferative disease. We employed a multidisciplinary collaborative treatment approach, and after 13 wound changes over 45 days, combined with radiation therapy, dietary supplementation, and psychosocial therapy, the left upper extremity wound healed.
CONCLUSION Giant ulcers caused by cutaneous EBV-positive NK/T-cell proliferative disease can be treated using a multidisciplinary collaborative approach.
Collapse
Affiliation(s)
- Ya-Ping Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhi-Xin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Shu-Li Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
2
|
Sørensen S, Kvich L, Xu Y, Thomsen TR, Bjarnsholt T, Thaarup I. Development of a tri-species wound model for studying fungal-bacterial interactions and antimicrobial therapies. Biofilm 2025; 9:100256. [PMID: 39927095 PMCID: PMC11804781 DOI: 10.1016/j.bioflm.2025.100256] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 01/17/2025] [Indexed: 02/11/2025] Open
Abstract
Chronic wounds are increasing in numbers and biofilm-producing bacteria are highly prevalent in these wounds and often create resilient polymicrobial infections. Moreover, estimates suggest that up to 23 % of wounds contain fungi, particularly Candida albicans. Currently, inter-kingdom chronic wound models are scarce; thus, this study presents one of the few in vitro models that incorporate both bacterial and fungal species in a wound-relevant environment, addressing a critical gap in current biofilm research. The newly developed model contained the commonly isolated wound bacteria Pseudomonas aeruginosa and Staphylococcus aureus, and the fungus Candida albicans. Inter-species interactions were investigated through selective plate counting and pH and oxygen measurements, as well as confocal microscopy. Investigations were carried out before and after exposure to commonly used clinical antimicrobial treatments, including silver-infused bandages. When grown in a tri-species consortium, P. aeruginosa and S. aureus exhibited a higher tolerance towards silver-infused bandages than when they were grown individually. This suggests that C. albicans plays a protective role for the bacteria. In addition, the treatment also caused a shift in species ratios, moving from a P. aeruginosa-dominated consortium to a S. aureus-dominated consortium. Moreover, confocal microscopy revealed a change in biofilm architecture when comparing single-species models to tri-species models. Finally, we observed that silver-infused bandages increased the pH in the tri-species model as well as partially restoring the oxygenation within the wound model. In conclusion, our novel model exemplifies how inter-kingdom interactions in fungal-bacterial infections can complicate both the microenvironment and treatment efficacy.
Collapse
Affiliation(s)
- Stine Sørensen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lasse Kvich
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Center for Surgical Science, Zealand University Hospital, Køge, Denmark
| | - Yijuan Xu
- SEGES Innovation P/S, Aarhus, Denmark
| | - Trine R. Thomsen
- Department of Chemistry and Biotechnology, Aalborg University, Aalborg, Denmark
- Danish Technology Institute, Aarhus, Denmark
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ida Thaarup
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Habib MB, Batool G, Shah NA, Muhammad T, Akbar NS, Shahid A. Biofilm-mediated infections; novel therapeutic approaches and harnessing artificial intelligence for early detection and treatment of biofilm-associated infections. Microb Pathog 2025; 203:107497. [PMID: 40118297 DOI: 10.1016/j.micpath.2025.107497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 12/04/2024] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
A biofilm is a group of bacteria that have self-produced a matrix and are grouped together in a dense population. By resisting the host's immune system's phagocytosis process and attacking with anti-microbial chemicals such as reactive oxygen and nitrogen species, a biofilm enables pathogenic bacteria to evade elimination. One of the major problems in managing chronic injuries is treating wounds colonized by biofilms. These days, a major issue is the biofilms, which exacerbate infection pathogenesis and severity. Numerous investigators have already discovered cutting-edge methods for biofilm manipulation. Using phytochemicals is a practical tactic to control and prevent the production of biofilms. Numerous studies conducted in the last few years have demonstrated the antibacterial and antibiofilm qualities of nanoparticles (NPs) against bacteria, fungi, and protozoa. Because hydrogel has antibiofilm properties, it has been employed extensively in wound care recently. It may be removed with ease and without causing trauma. Today, artificial intelligence (AI) is being used to improve these tactics by providing customized treatment alternatives and predictive analytics. Artificial intelligence (AI) algorithms have the capability to examine extensive datasets and detect trends in biofilm formation and resistance mechanisms. This can aid in the creation of more potent antimicrobial drugs. AI models analyze complex datasets, predict biofilm formation, and guide the design of personalized treatment strategies by identifying resistance mechanisms and therapeutic targets with exceptional precision. This review provides an integrative perspective on biofilm formation mechanisms and their role in infections, highlighting the innovative applications of AI in this domain. By integrating data from diverse biological systems, AI accelerates drug discovery, optimizes treatment regimens, and enables real-time monitoring of biofilm dynamics. From predictive analytics to personalized care, we explore how AI enhances biofilm diagnostics and introduces precision medicine in biofilm-associated infections. This approach not only addresses the limitations of traditional methods but also paves the way for revolutionary advancements in infection control, antimicrobial resistance management, and improved patient outcomes.
Collapse
Affiliation(s)
| | - Ghanwa Batool
- Department of Computer Science, Comsats University Islamabad, Abbottabad, 22060, Pakistan
| | - Naseer Ali Shah
- Department of Biosciences, COMSATS University, Islamabad, 44000, Pakistan
| | - Taseer Muhammad
- Department of Mathematics, College of Science, King Khalid University, Abha, 61413, Saudi Arabia
| | - Noreen Sher Akbar
- Department of Mechanical Engineering, College of Engineering, Prince Mohammad Bin Fahd University, Al Khobar, 31952, Saudi Arabia
| | - Ameera Shahid
- National Institute of Health, Islamabad, 44000, Pakistan
| |
Collapse
|
4
|
Seol Y, Ganguly K, Patil TV, Dutta SD, Park H, Lee J, Randhawa A, Kim H, Lim KT. Zinc Oxide@Tetracycline Spiky Microparticles Design for Persistent Antibacterial Therapy. J Biomed Mater Res A 2025; 113:e37915. [PMID: 40269589 DOI: 10.1002/jbm.a.37915] [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: 01/19/2025] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Antibiotics have revolutionized medical treatment by effectively combating bacterial infections, particularly those associated with chronic wounds and implant complications. Nevertheless, the persistent use of these drugs has resulted in an increase in antibiotic-resistant bacteria and biofilm infections, highlighting the urgent need for alternative therapies. This study presents an approach for combating persistent bacterial and biofilm infections through the integration of biomimetic design and advanced nanotechnology. Inspired by the natural defense mechanisms of pollen grains and lotus leaves, we engineered zinc oxide spiky microparticles combined with tetracycline-loaded beads mimicking the structure of lotus leaf papillae. This biomimetic design exhibits a multifaceted antimicrobial strategy, leveraging hierarchical micro/nanostructures and the inherent antibacterial properties of their natural counterparts. ZnO microparticles, which mimic the morphology of pollen grains, provide topological cues to rupture adhered bacteria, whereas tetracycline beads, inspired by lotus leaf papillae, deliver a controlled release of antibiotics to target persistent bacteria. Using a synergistic multimodal approach, our biomimetic materials demonstrated exceptional efficacy in eradicating persistent methicillin-resistant Staphylococcus aureus and Escherichia coli infections, offering promising prospects for the development of advanced antibacterial therapies. This study not only underscores the importance of biomimicry in material design but also highlights the potential of integrating nature-inspired strategies with nanotechnology for biomedical applications.
Collapse
Affiliation(s)
- Youjin Seol
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia, USA
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Hyeonseo Park
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Jieun Lee
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Hojin Kim
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon University, Chuncheon, Gangwon-do, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| |
Collapse
|
5
|
Zhong H, Chen Z, Huang J, Yu X, Wang C, Zheng Y, Peng M, Yuan Z. Spray-drying-engineered CS/HA-bilayer microneedles enable sequential drug release for wound healing. J Mater Chem B 2025; 13:4819-4829. [PMID: 40152787 DOI: 10.1039/d5tb00121h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
High incidence and mortality rates of chronic wounds place a heavy burden on global healthcare systems. Achieving phased delivery of antimicrobial and regenerative drugs is crucial for promoting chronic wound healing. Herein, a microneedle (MN) patch with a biphasic release system was developed using a combination of solvent casting and spraying methods. Additionally, a copper/PDMS mold was introduced to address the issue of deformation in the chitosan material during drying on polydimethylsiloxane (PDMS). The MNs have a bilayer structure, with a hyaluronic acid (HA) coating loaded with doxycycline (DOX) for antibacterial action and a chitosan (CS) core loaded with vascular endothelial growth factor (VEGF) for promoting cell migration and proliferation. Notably, in vitro drug release studies showed that the coating drug was released by 98.8% within 10 hours, while the release of the core drug could be sustained for up to 70 hours. In vivo studies showed that chronic wounds on C57 mice treated with CS/HA-bilayer MNs achieved nearly complete healing by day 9. These wounds exhibited reduced inflammatory cell infiltration, increased epithelial tissue regeneration, and enhanced collagen deposition. This work integrates the staged management of bacterial infection and angiogenesis and offers promising prospects for enhancing chronic wound healing.
Collapse
Affiliation(s)
- Haowen Zhong
- School of Electro-mechanical Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
- Guangdong Provincial Key Laboratory of Minimally Invasive Surgical Instruments and Manufacturing Technology, Guangdong University of Technology, Guangzhou, 510006, China
- State Key Laboratory for High Performance Tools, Guangdong University of Technology, Guangzhou, 510006, China
- Smart Medical Innovation Technology Center, Guangdong University of Technology, Guangzhou, 510006, China
| | - Zongyou Chen
- School of Electro-mechanical Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
- Guangdong Provincial Key Laboratory of Minimally Invasive Surgical Instruments and Manufacturing Technology, Guangdong University of Technology, Guangzhou, 510006, China
- State Key Laboratory for High Performance Tools, Guangdong University of Technology, Guangzhou, 510006, China
- Smart Medical Innovation Technology Center, Guangdong University of Technology, Guangzhou, 510006, China
| | - Jiahao Huang
- School of Electro-mechanical Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
- Guangdong Provincial Key Laboratory of Minimally Invasive Surgical Instruments and Manufacturing Technology, Guangdong University of Technology, Guangzhou, 510006, China
- State Key Laboratory for High Performance Tools, Guangdong University of Technology, Guangzhou, 510006, China
- Smart Medical Innovation Technology Center, Guangdong University of Technology, Guangzhou, 510006, China
| | - Xiao Yu
- School of Electro-mechanical Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
- Guangdong Provincial Key Laboratory of Minimally Invasive Surgical Instruments and Manufacturing Technology, Guangdong University of Technology, Guangzhou, 510006, China
- State Key Laboratory for High Performance Tools, Guangdong University of Technology, Guangzhou, 510006, China
- Smart Medical Innovation Technology Center, Guangdong University of Technology, Guangzhou, 510006, China
| | - Chengyong Wang
- School of Electro-mechanical Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
- Guangdong Provincial Key Laboratory of Minimally Invasive Surgical Instruments and Manufacturing Technology, Guangdong University of Technology, Guangzhou, 510006, China
- State Key Laboratory for High Performance Tools, Guangdong University of Technology, Guangzhou, 510006, China
- Smart Medical Innovation Technology Center, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yue Zheng
- Nanfang Hospital, Southern Medical University, Guangzhou, 510006, China
| | - Mengran Peng
- Department of Dermatology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhishan Yuan
- School of Electro-mechanical Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
- Guangdong Provincial Key Laboratory of Minimally Invasive Surgical Instruments and Manufacturing Technology, Guangdong University of Technology, Guangzhou, 510006, China
- State Key Laboratory for High Performance Tools, Guangdong University of Technology, Guangzhou, 510006, China
- Smart Medical Innovation Technology Center, Guangdong University of Technology, Guangzhou, 510006, China
| |
Collapse
|
6
|
Allkja J, Bakri A, Short B, Gilmour A, Brown JL, Bal AM, Newby KJM, Jenkins T, Short RD, Williams C, Ramage G. Investigating the Prevalence of Fungi in Diabetic Ulcers: An Under-Recognised Contributor to Polymicrobial Biofilms. APMIS 2025; 133:e70025. [PMID: 40264255 PMCID: PMC12015384 DOI: 10.1111/apm.70025] [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: 09/06/2024] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025]
Abstract
Diabetic foot ulcers (DFUs) are common complications for diabetic patients, often exacerbated by complex polymicrobial biofilm infections. While the majority of DFU studies are bacterial focused, fungi have also been identified. This study aims to investigate the prevalence of fungi in DFUs, as well as their potential role and influence on persistence and wound healing. Consecutive DFU swabs were collected from 128 patients (n = 349). Fungal positivity was assessed using enhanced culture and real-time qPCR. Routine microbiology cultures were carried out as part of standard care in the clinics, and their results were then compared to our laboratory investigation. Routine and enhanced culture resulted in similar rates of fungal detection (~9%), whereas qPCR resulted in a higher rate of detection (31%). Notably, the predominant yeast Candida parapsilosis was present in ischaemic and penetrating bone wounds. These findings support existing evidence of fungal presence in DFUs. We demonstrated that routine diagnostic methods are sufficient for fungal detection, but enhanced culture methods allow for more precise fungal identification. Finally, while fungal presence does not appear to impact patient outcomes in our study, their role within these infections remains poorly understood, and further studies are needed to fully understand their relationship to the microbiome.
Collapse
Affiliation(s)
- Jontana Allkja
- Safeguarding Health Through Infection Prevention (SHIP) Research Group, Research Centre for Health, School of Health and Life SciencesGlasgow Caledonian UniversityGlasgowUK
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Ahmed Bakri
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | - Bryn Short
- Safeguarding Health Through Infection Prevention (SHIP) Research Group, Research Centre for Health, School of Health and Life SciencesGlasgow Caledonian UniversityGlasgowUK
| | - Andrew Gilmour
- Safeguarding Health Through Infection Prevention (SHIP) Research Group, Research Centre for Health, School of Health and Life SciencesGlasgow Caledonian UniversityGlasgowUK
| | - Jason L. Brown
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUK
| | | | - Kelly J. M. Newby
- Pharmacy DepartmentUniversity Hospitals of Morecambe Bay NHS Foundation TrustUK
| | - Toby Jenkins
- Department of ChemistryThe University of SheffieldSheffieldUK
| | | | - Craig Williams
- Microbiology Department, Lancaster Royal InfirmaryUniversity of LancasterLancasterUK
| | - Gordon Ramage
- Safeguarding Health Through Infection Prevention (SHIP) Research Group, Research Centre for Health, School of Health and Life SciencesGlasgow Caledonian UniversityGlasgowUK
| |
Collapse
|
7
|
Hickerson N, Gruzmark F, Danker S, Lev-Tov H. Addressing the Dressings: Wound Care in Hidradenitis Suppurativa. Dermatol Clin 2025; 43:261-272. [PMID: 40023626 DOI: 10.1016/j.det.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Hidradenitis suppurativa (HS) wounds are complex and have unique characteristics that complicate dressing selection. Proper wound care assists with wound healing and contributes to improved quality of life. Ideal dressings are comfortable while maintaining a wound-healing environment. Antimicrobial dressings should target dysbiosis and biofilms. Absorptive dressings are needed for control of exudate. Pain control is best achieved with nonadhesive dressings and contact layers. Agents targeting anaerobic bacteria are effective at odor reduction. Surgery may be indicated for management of refractory wounds. Overall, there is a need for increased research and education on both routine and postsurgical wound care in HS.
Collapse
Affiliation(s)
- Natalie Hickerson
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami, 1600 NW 10th Avenue RMSB 2023A, Miami, FL 33136, USA
| | - Fiona Gruzmark
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami, 1600 NW 10th Avenue RMSB 2023A, Miami, FL 33136, USA
| | - Sara Danker
- Division of Plastic Surgery, University of Miami Miller School of Medicine, 1120 NW 14th Street Suite 9, Miami, FL 33136, USA
| | - Hadar Lev-Tov
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami, 1600 NW 10th Avenue RMSB 2023A, Miami, FL 33136, USA.
| |
Collapse
|
8
|
Sakib S, Andoy NMO, Yang JYC, Galang A, Sullan RMA, Zou S. Antimicrobial and anti-inflammatory effects of polyethyleneimine-modified polydopamine nanoparticles on a burn-injured skin model. Biomater Sci 2025; 13:1770-1783. [PMID: 39995391 DOI: 10.1039/d4bm01530d] [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: 02/26/2025]
Abstract
Chronic infections involving bacterial biofilms pose significant treatment challenges due to the resilience of biofilms against existing antimicrobials. Here, we introduce a nanomaterial-based platform for treating Staphylococcus epidermidis biofilms, both in isolation and within a biofilm-infected burn skin model. Our approach leverages biocompatible and photothermal polydopamine nanoparticles (PDNP), functionalized with branched polyethyleneimine (PEI) and loaded with the antibiotic rifampicin, to target bacteria dwelling within biofilms. A key innovation of our method is its ability to not only target planktonic S. epidermidis but also effectively tackle biofilm-embedded bacteria. We demonstrated that PDNP-PEI interacts effectively with the bacterial surface, facilitating laser-activated photothermal eradication of planktonic S. epidermidis. In a 3D skin burn injury model, PDNP-PEI demonstrates anti-inflammatory and reactive oxygen species (ROS)-scavenging effects, reducing inflammatory cytokine levels and promoting healing. The rifampicin-loaded PDNP-PEI (PDNP-PEI-Rif) platform further shows significant efficacy against bacteria inside biofilms. The PDNP-PEI-Rif retained its immunomodulatory activity and efficiently eradicated biofilms grown on our burn-injured 3D skin model, effectively addressing the challenges of biofilm-related infections. This achievement marks a significant advancement in infection management, with the potential for a transformative impact on clinical practice.
Collapse
Affiliation(s)
- Sadman Sakib
- Metrology Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1N 5A2, Canada.
| | - Nesha May O Andoy
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
| | - Jessica Y C Yang
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
| | - Anna Galang
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON, M5S 3H6, Canada.
| | - Ruby May A Sullan
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, 1065 Military Trail, Toronto, ON, M1C 1A4 Canada
- Department of Chemistry, University of Toronto, 80 St. George St., Toronto, ON, M5S 3H6, Canada.
| | - Shan Zou
- Metrology Research Centre, National Research Council Canada, 100 Sussex Drive, Ottawa, Ontario, K1N 5A2, Canada.
| |
Collapse
|
9
|
Beraldo S, Ljungqvist J, Rodger R, Hanson B, Saavedra C. Effectiveness of an enhanced silver-containing dressing in hard-to-heal venous leg ulcers: a randomised controlled trial. J Wound Care 2025; 34:170-178. [PMID: 40047822 DOI: 10.12968/jowc.2025.0023] [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: 05/13/2025]
Abstract
OBJECTIVE To assess the efficacy and safety of a carboxymethylcellulose dressing containing ionic silver, ethylenediaminetetraacetic acid and benzethonium chloride (CISEB) versus a dialkylcarbamoyl chloride-coated dressing (DACC) in hard-to-heal venous leg ulcers (VLUs). METHOD In a multinational, multicentre, randomised controlled trial, patients with hard-to-heal VLUs were randomised 1:1 to receive CISEB (n=100) or DACC (n=103) for up to four weeks. VLUs that were not healed by week 4 were managed with standard of care for up to 12 weeks or until healed (whichever was sooner). The primary endpoint was complete wound closure at week 12. Additional endpoints included time to complete wound closure and incidence of adverse events (AEs). RESULTS The trial cohort included 203 patients. CISEB achieved a higher rate of complete wound closure by week 12 compared to DACC (74.8% versus 55.6%, respectively; p<0.0031), and was associated with a 35% increased likelihood of healing (risk ratio, 1.35; 95% confidence interval: 1.10-1.65). Median time to complete wound closure was shorter in the CISEB arm (56 days) compared to the DACC arm (70 days; p<0.0272). A smaller proportion of patients experienced an AE with CISEB compared to DACC (5.0% versus 17.6%, respectively). CONCLUSION Management of hard-to-heal VLUs with CISEB was associated with improved healing outcomes compared to DACC, without additional safety concerns. CISEB is a gelling fibre dressing with antimicrobial, metal-chelating and surfactant components that may promote an optimal healing environment to address the challenge of hard-to-heal wounds.
Collapse
|
10
|
Davis SC, Avery JT, Gil J, Solis MR, Jozic I, Kimmerling KA, Mowry KC. Protection with a collagen wound matrix containing polyhexamethylene biguanide supports innate wound healing in biofilm-infected porcine wounds. Wound Repair Regen 2025; 33:e70025. [PMID: 40251887 PMCID: PMC12008732 DOI: 10.1111/wrr.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/25/2025] [Indexed: 04/21/2025]
Abstract
Over 90% of chronic wounds have biofilm infections, making the need for inhibiting reformation of biofilm post-debridement paramount to support progression through the normal phases of wound healing. Herein, we describe a porcine wound model infected with methicillin-resistant Staphylococcus aureus (MRSA) and examine the ability of an antimicrobial barrier composed of native type I collagen and polyhexamethylene biguanide (PCMP) to serve as a barrier to protect wounds and support progression through the innate wound healing cascade. Wounds were inoculated with MRSA and allowed to form a biofilm for 72 h, subjected to standard of care sharp debridement, then either left untreated or received PCMP for 5, 10, 15 or 20 days. Wounds were assessed for bioburden, wound closure and expression of genes related to wound healing. Wounds treated with PCMP exhibited statistically lower MRSA levels compared to untreated controls and achieved 90% closure by 2 weeks of treatment. Gene expression analysis demonstrated that by reducing bacterial load, wounds progressed through the innate wound healing cascade, while untreated wounds exhibited a dampening of the immune response. Additionally, for randomly assigned wounds, PCMP was not reapplied at dressing changes to assess the impact of inconsistent wound protection. At all timepoints, a resurgence in bioburden was observed following removal of PCMP if the wounds had not fully closed. This study highlights the value of PCMP as an antimicrobial barrier and the importance of protecting wounds through closure and resolution.
Collapse
Affiliation(s)
- Stephen C. Davis
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miller School of MedicineUniversity of MiamiFloridaUSA
| | - Justin T. Avery
- Research & DevelopmentOrganogenesis Discovery CenterBirminghamAlabamaUSA
| | - Joel Gil
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miller School of MedicineUniversity of MiamiFloridaUSA
| | - Michael R. Solis
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miller School of MedicineUniversity of MiamiFloridaUSA
| | - Ivan Jozic
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miller School of MedicineUniversity of MiamiFloridaUSA
| | | | - Katie C. Mowry
- Research & DevelopmentOrganogenesis Discovery CenterBirminghamAlabamaUSA
| |
Collapse
|
11
|
Gerges BZ, Rosenblatt J, Truong YL, Jiang Y, Raad II. The Antifungal Activity of a Polygalacturonic and Caprylic Acid Ointment in an In Vitro, Three-Dimensional Wound Biofilm Model. J Fungi (Basel) 2025; 11:178. [PMID: 40137216 PMCID: PMC11943374 DOI: 10.3390/jof11030178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/31/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
Candida colonization and biofilms are significant contributors to impaired wound healing. Consequently, improved treatments are needed to eradicate Candida biofilms in wounds. Wounds present complex biofilm extracellular matrix environments, with microbial cells frequently enmeshed in matrices comprising wound exudate macromolecular gels. We evaluated the ability of a polygalacturonic and caprylic acid (PG + CAP) ointment to eradicate Candida albicans, C. parapsilosis, C. glabrata, C. tropicalis, and C. auris biofilms in a fibrin gel wound biofilm model of the complex wound biofilm environment. Hypochlorous acid (HOCl) is a disinfecting antimicrobial agent that is widely used as wound irrigant, and this was used as a comparator. A single treatment with PG + CAP reduced the number of viable organisms in the C. albicans and C. glabrata biofilms by over 5 log10, in the C. parapsilosis and C. auris biofilms by over 4 log10, and in the C. tropicalis biofilm by 3.85 log10. PG + CAP was superior (p < 0.01) to HOCl in eradicating all Candida species biofilms, except for C. auris, for which both treatments fully eradicated all viable organisms. The use of HOCl in Candida-colonized wounds should include consideration of the extracellular matrix load in the wound bed. PG + CAP warrants further study in wounds compromised by Candida biofilms.
Collapse
Affiliation(s)
- Bahgat Z. Gerges
- Department of Infectious Diseases, Infection Control and Employee Health Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (J.R.); (Y.-L.T.); (Y.J.); (I.I.R.)
| | | | | | | | | |
Collapse
|
12
|
Balukoff NC, Houk G, Gonzalez T, Berton Y, Ronfard V, Pastar I, Tomic-Canic M. Out of this World: Wound Healing on Earth and in Space. J Invest Dermatol 2025:S0022-202X(25)00027-2. [PMID: 39955658 DOI: 10.1016/j.jid.2024.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 02/17/2025]
Abstract
Impaired wound healing is a significant concern for humans in space, where the unique microgravity environment poses challenges to the natural healing processes of the body. Similar to chronic wounds on earth, such as diabetic foot ulcers and venous leg ulcers, wounds inflicted in space exhibit delayed or impaired healing responses. These wounds share common features, including dysregulated cellular signaling, altered cytokine profiles, and impaired tissue regeneration. Little is known about the mechanisms underlying wound healing under microgravity. In this review, we focused on exploring the parallels between wound healing in space and chronic wounds on earth as a fundamental approach for developing effective countermeasures to promote healing and mitigate associated health risks during long-space missions.
Collapse
Affiliation(s)
- Nathan C Balukoff
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Garrett Houk
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Tammy Gonzalez
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.
| |
Collapse
|
13
|
Indrakumar S, Gugulothu SB, Joshi A, Dash TK, Mishra V, Tandon B, Chatterjee K. Silk Composite-Based Multifunctional Pellets for Controlled Release. Macromol Biosci 2025; 25:e2400410. [PMID: 39427344 DOI: 10.1002/mabi.202400410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/06/2024] [Indexed: 10/22/2024]
Abstract
Chronic wounds present significant clinical challenges due to the high risk of infections and persistent inflammation. While personalized treatments in point-of-care settings are crucial, they are limited by the complex fabrication techniques of the existing products. The calcium sulfate hemihydrate (CSH)-based drug delivery platform enables rapid fabrication but lacks antioxidant and antibacterial properties, essential to promote healing. To develop a multifunctional platform, a tannic acid (TA)-silk fibroin (SF) complex is engineered and incorporated as an additive in CSH cement. This cement is then cast into pellets to create silk/bioceramic-based composite drug delivery systems, designed for point-of-care use. Compared to neat CSH pellets, the composite pellets exhibit a 7.5-fold increase in antioxidant activity and prolonged antibacterial efficacy (up to 13 d). Moreover, the subcutaneous implantation of the pellets shows no hallmarks of local or systemic toxicity in a rodent model. The pellets are optimized in composition and fabrication to ease market translation. Clinically, the pellets have the potential to be further developed into products to place on wound beds or fill into bone cavities that are designed to deliver the intended therapeutic effect. The developed multifunctional system proves to be a promising solution for personalized treatment in point-of-care settings.
Collapse
Affiliation(s)
- Sushma Indrakumar
- Department of Materials Engineering, Indian Institute of Science, Bangalore, 560012, India
| | | | - Akshat Joshi
- Department of Materials Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Tapan Kumar Dash
- Fibroheal Woundcare Pvt. Ltd., Yelahanka New Town, Bangalore, 560064, India
| | - Vivek Mishra
- Fibroheal Woundcare Pvt. Ltd., Yelahanka New Town, Bangalore, 560064, India
| | - Bharat Tandon
- Fibroheal Woundcare Pvt. Ltd., Yelahanka New Town, Bangalore, 560064, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
14
|
Wang JL, Pan X, Li X, Liu KM, Yao M, An JY, Wan Y, Yu XQ, Feng S, Wu MY. Photoimmunologic Therapy of Stubborn Biofilm via Inhibiting Bacteria Revival and Preventing Reinfection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411468. [PMID: 39723739 DOI: 10.1002/adma.202411468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/11/2024] [Indexed: 12/28/2024]
Abstract
Stubborn biofilm infections pose serious threats to public health. Clinical practices highly rely on mechanical debridement and antibiotics, which often fail and lead to persistent and recurrent infections. The main culprits are 1) persistent bacteria reviving, colonizing, and rejuvenating biofilms, and 2) secondary pathogen exposure, particularly in individuals with chronic diseases. Addressing how to inhibit persistent bacteria revival and prevent reinfection simultaneously is still a major challenge. Herein, an oligo-ethylene glycol-modified lipophilic cationic photosensitizer (PS), TBTCP-PEG7, is developed. It effectively eradicates Methicillin-Resistant Staphylococcus aureus (MRSA) under light irradiation. Furthermore, TBTCP-PEG7-mediated photodynamic therapy (PDT) not only conquers stubborn biofilm infections by downregulating the two-component system (TCS), quorum sensing (QS), and virulence factors, thereby reducing intercellular communication, inhibiting persistent bacterial regrowth and biofilm remodeling but also prevents reinfection by upregulating heat shock protein-related genes to induce immunogenetic cell death (ICD) and establish immune memory. In vivo, TBTCP-PEG7 efficiently eradicates MRSA biofilm adhered to medical catheters, stimulates angiogenesis, reduces inflammatory factor expression, and accelerates wound healing. Furthermore, ICD promotes short-term immune and long-term immunological memory for coping with secondary infections. This two-pronged strategy not only effectively overcomes stubborn, persistent and recurrent biofilm infection, but also provides theoretical guidance for designing the next generation of antibacterial materials.
Collapse
Affiliation(s)
- Jia-Li Wang
- School of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xiu Pan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xin Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Kun-Mei Liu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Mei Yao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jin-Yu An
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yu Wan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xiao-Qi Yu
- Asymmetric Synthesis and Chiral Technology Key Laboratory of Sichuan Province, Department of Chemistry, Xihua University, Chengdu, 610039, P. R. China
| | - Shun Feng
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Ming-Yu Wu
- School of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
15
|
Ojeh N, Vecin NM, Pastar I, Volk SW, Wilgus T, Griffiths S, Ramey‐Ward AN, Driver VR, DiPietro LA, Gould LJ, Tomic‐Canic M. The Wound Reporting in Animal and Human Preclinical Studies (WRAHPS) Guidelines. Wound Repair Regen 2025; 33:e13232. [PMID: 39639458 PMCID: PMC11621255 DOI: 10.1111/wrr.13232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/02/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024]
Abstract
Preclinical studies for wound healing disorders are an essential step in translating discoveries into therapies. Also, they are an integral component of initial safety screening and gaining mechanistic insights using an in vivo approach. Given the complexity of the wound healing process, existing guidelines for animal testing do not capture key information due to the inevitable variability in experimental design. Variations in study interpretation are increased by complexities associated with wound aetiology, wounding procedure, multiple treatment conditions, wound assessment, and analysis, as well as lack of acknowledgement of limitation of the model used. Yet, no standards exist to guide reporting crucial experimental information required to interpret results in translational studies of wound healing. Consistency in reporting allows transparency, comparative, and meta-analysis studies and avoids repetition and redundancy. Therefore, there is a critical and unmet need to standardise reporting for preclinical wound studies. To aid in reporting experimental conditions, The Wound Reporting in Animal and Human Preclinical Studies (WRAHPS) Guidelines have now been created by the authors working with the Wound Care Collaborative Community (WCCC) GAPS group to provide a checklist and reporting template for the most frequently used preclinical models in support of development for human clinical trials for wound healing disorders. It is anticipated that the WRAHPS Guidelines will standardise comprehensive methods for reporting in scientific manuscripts and the wound healing field overall. This article is not intended to address regulatory requirements but is intended to provide general guidelines on important scientific considerations for such studies.
Collapse
Affiliation(s)
- Nkemcho Ojeh
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Preclinical and Health Sciences, Faculty of Medical SciencesThe University of the West IndiesBridgetownBarbados
| | - Nicole M. Vecin
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Susan W. Volk
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Traci Wilgus
- Department of PathologyThe Ohio State UniversityColumbusOhioUSA
| | | | | | - Vickie R. Driver
- School of MedicineWashington State UniversitySpokaneWashingtonUSA
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue RegenerationUniversity of Illinois ChicagoChicagoIllinoisUSA
| | - Lisa J. Gould
- South Shore Hospital Center for Wound HealingWeymouthMassachusettsUSA
| | - Marjana Tomic‐Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
16
|
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
|
17
|
Liu M, Jiang J, Wang Y, Liu H, Lu Y, Wang X. Smart drug delivery and responsive microneedles for wound healing. Mater Today Bio 2024; 29:101321. [PMID: 39554838 PMCID: PMC11567927 DOI: 10.1016/j.mtbio.2024.101321] [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: 05/08/2024] [Revised: 09/25/2024] [Accepted: 10/29/2024] [Indexed: 11/19/2024] Open
Abstract
Wound healing is an ongoing concern for the medical community. The limitations of traditional dressings are being addressed by materials and manufacturing technology. Microneedles (MNs) are a novel type of drug delivery system that has been widely used in cancer therapy, dermatological treatment, and insulin and vaccine delivery. MNs locally penetrate necrotic tissue, eschar, biofilm and epidermis into deep tissues, avoiding the possibility of drug dilution and degradation and greatly improving administration efficiency with less pain. MNs represent a new direction for wound treatment and transdermal delivery. In this study, we summarise the skin wound healing process and the mechanical stimulation of MNs in the context of the wound healing process. We also introduce the structural design and manufacture of MNs. Subsequently, MNs are categorised according to the loaded drugs, where the design of the MNs according to the traumatic biological/biochemical microenvironment (pH, glucose, and bacteria) and the physical microenvironment (temperature, light, and ultrasound) is emphasised. Finally, the advantages of MNs are compared with traditional drug delivery systems and their prospects are discussed.
Collapse
Affiliation(s)
- Meixuan Liu
- Department of Burns & Wound Care Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Jing Jiang
- Department of Burns & Wound Care Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yiran Wang
- Department of Burns & Wound Care Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Huan Liu
- Department of Burns & Wound Care Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yiping Lu
- Senior once Class 5, Shanghai Pinghe School, Shanghai, 200000, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
18
|
Jeong GJ, Khan F, Kim DK, Cho KJ, Tabassum N, Choudhury A, Hassan MI, Jung WK, Kim HW, Kim YM. Marine polysaccharides for antibiofilm application: A focus on biomedical fields. Int J Biol Macromol 2024; 283:137786. [PMID: 39577534 DOI: 10.1016/j.ijbiomac.2024.137786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Microbial pathogens such as bacteria and fungi form biofilms, which represent substantial hurdles in treating human illness owing to their adaptive resistance mechanism to conventional antibiotics. Biofilm may cause persistent infection in a variety of bodily areas, including wounds, oral cavity, and vaginal canal. Using invasive devices such as implants and catheters contributes significantly to developing healthcare-associated infections because they offer an ideal surface for biofilm formation. Marine organisms produce a variety of polysaccharides, which have recently attracted worldwide attention due to their biochemical features, various applications, and advantageous properties such as bioactivity, biodegradability, and biocompatibility. Because of their antimicrobial and antibiofilm features, several polysaccharides such as chitosan, fucoidan, carrageenan, alginate, and hyaluronic acid have been used to treat infected wounds as well as ophthalmic, oral, and vaginal infections. In addition, marine polysaccharides are currently employed as coatings on medical devices and implant materials, alone or in combination with other bioactive substances or nanomaterials, to protect the materials' undertones from microbial contamination. This review discussed the recent advancements in marine polysaccharides and their derivatives as a therapeutic potential against biofilm-associated diseases. The potential obstacles in the scalability of their production, clinical translation, and/or regulatory hurdles have also been discussed.
Collapse
Affiliation(s)
- Geum-Jae Jeong
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Fazlurrahman Khan
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Ocean and Fisheries Development International Cooperation Institute, Pukyong National University, Busan 48513, Republic of Korea; International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea.
| | - Do-Kyun Kim
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Kyung-Jin Cho
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Nazia Tabassum
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Hyun-Woo Kim
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea; Department of Marine Biology, Pukyong National University, Busan 48513, Republic of Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan 48513, Republic of Korea; Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan 48513, Republic of Korea; Research Center for Marine Integrated Bionics Technology, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
19
|
Roshni PT, Rekha PD. Essential oils: a potential alternative with promising active ingredients for pharmaceutical formulations in chronic wound management. Inflammopharmacology 2024; 32:3611-3630. [PMID: 39312099 DOI: 10.1007/s10787-024-01571-3] [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: 07/30/2024] [Accepted: 09/11/2024] [Indexed: 11/10/2024]
Abstract
Chronic wound is a major clinical challenge that complicates wound healing, mainly associated with bacterial biofilms. Bacterial burden damages tissue and persists inflammation, failing to granulate, leading to morbidity and mortality. Various therapeutic strategies and approaches have been developed for chronic wound healing in clinical practice. As treating biofilm infection is crucial in chronic wounds, a potent antibiofilm agent, essential oils have been explored extensively for their therapeutic properties and as a replacement for antibiotic therapy. Currently, several studies on essential oils and their active compounds in therapeutics, such as adjunctive therapies, nanotechnology-based treatment and their drug delivery systems, help heal chronic wounds. The antimicrobial, anti-inflammatory and antioxidant properties of essential oils make them distinct and are renowned as natural remedies to improve the healing of infected chronic wounds. Consequently, it accelerates wound closure by reducing inflammation, increasing angiogenesis and tissue regeneration. This review focuses on different essential oils and their active compounds that are exploited for the treatment of biofilm infection, chronic inflammation and wound healing. Thus, an effective novel treatment can be developed to improve the current treatment strategy to overcome multidrug resistance bacteria or antibiotic resistance in various chronic wound infections that support wound healing.
Collapse
Affiliation(s)
- Pulukkunadu Thekkeveedu Roshni
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka 575018, India
| | - Punchappady Devasya Rekha
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka 575018, India.
| |
Collapse
|
20
|
Yang F, Shu R, Dai W, Li B, Liu C, Yang H, Johnson HM, Yu S, Bai D, Yang W, Deng Y. H 2Se-evolving bio-heterojunctions promote cutaneous regeneration in infected wounds by inhibiting excessive cellular senescence. Biomaterials 2024; 311:122659. [PMID: 38861831 DOI: 10.1016/j.biomaterials.2024.122659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024]
Abstract
Pathogenic infection leads to excessive senescent cell accumulation and stagnation of wound healing. To address these issues, we devise and develop a hydrogen selenide (H2Se)-evolving bio-heterojunction (bio-HJ) composed of graphene oxide (GO) and FeSe2 to deracinate bacterial infection, suppress cellular senescence and remedy recalcitrant infected wounds. Excited by near-infrared (NIR) laser, the bio-HJ exerts desired photothermal and photodynamic effects, resulting in rapid disinfection. The crafted bio-HJ could also evolve gaseous H2Se to inhibit cellular senescence and dampen inflammation. Mechanism studies reveal the anti-senescence effects of H2Se-evolving bio-HJ are mediated by selenium pathway and glutathione peroxidase 1 (GPX1). More critically, in vivo experiments authenticate that the H2Se-evolving bio-HJ could inhibit cellular senescence and potentiate wound regeneration in rats. As envisioned, our work not only furnishes the novel gasotransmitter-delivering bio-HJ for chronic infected wounds, but also gets insight into the development of anti-senescence biomaterials.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Shu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenyu Dai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chuang Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Hang Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China
| | - Hannah M Johnson
- Department of Chemistry, Washington State University, Washington, USA
| | - Sheng Yu
- Department of Chemistry, Washington State University, Washington, USA
| | - Ding Bai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weizhong Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China.
| | - Yi Deng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, College of Biomedical Engineering, School of Chemical Engineering, Sichuan University, Chengdu, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
21
|
Zhang YS, Ke S, Hu X, Wang SY, Peng WQ, Qian XH, Tian LH, Wu HJ, Li BH, Zeng XT, Zhang LL. Enhancing wound healing through sonodynamic silver/barium titanate heterostructures-loading gelatin/PCL nanodressings. Int J Biol Macromol 2024; 283:137648. [PMID: 39547623 DOI: 10.1016/j.ijbiomac.2024.137648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/28/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Bacterial infections present a formidable challenge in surgical procedures, and are a major threat to wound healing. Sonodynamic therapy (SDT) is a non-invasive approach for fighting pathogens; however, it is hindered by the efficiency of sonosensitizers and effective antibacterial time. In this study, we developed a biocompatible nanodressing to improve the antibacterial efficacy and accelerate wound healing via SDT. Silver nanoparticles (NPs) were synthesized on tetragonal barium titanate (BTO) NPs to create an Ag@BTO heterostructure of sonosensitizers to improve their piezocatalytic activities, which were then incorporated into gelatin/polycaprolactone (PCL) to form Ag@BTO-gelatin/PCL nanofiber (ABT-gP NFs) dressings. The loading of Ag@BTO NPs resulted in ABT-gP NFs with better mechanical properties and excellent piezocatalysis, which produced reactive oxygen species and Ag+ to kill bacteria during ultrasound (US) irradiation. Additionally, nanodressing released moderate amounts of silver ions without US, prolonging the antibacterial time, while promoting fibroblast migration. This approach was effective in killing Gram-positive and Gram-negative bacteria (100 % and 90.8 %, respectively), promoting cell migration in vitro and accelerating wound healing without adverse effects in vivo. This study extends the potential applications of ultrasound-triggered nanodressing to the field of wound healing.
Collapse
Affiliation(s)
- Yu-Sen Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China
| | - Shuai Ke
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China
| | - Xiao Hu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China
| | - Shuang-Ying Wang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wan-Qi Peng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xin-Hang Qian
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ling-Hui Tian
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui-Jun Wu
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Bing-Hui Li
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xian-Tao Zeng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Department of Urology, Zhongnan Hosptial of Wuhan University, Wuhan 430071, China.
| | - Ling-Ling Zhang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
22
|
Chingizova EA, Chingizov AR, Menchinskaya ES, Pislyagin EA, Kuzmich AS, Leshchenko EV, Borkunov GV, Guzhova IV, Aminin DL, Yurchenko EA. The influence of marine fungal meroterpenoid meroantarctine A toward HaCaT keratinocytes infected with Staphylococcus aureus. J Antibiot (Tokyo) 2024; 77:812-822. [PMID: 39256545 DOI: 10.1038/s41429-024-00771-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/17/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
A new biological activity was discovered for marine fungal meroterpenoid meroantarctine A with unique 6/5/6/6 polycyclic system. It was found that meroantarctine A can significantly reduce biofilm formation by Staphylococcus aureus with an IC50 of 9.2 µM via inhibition of sortase A activity. Co-cultivation of HaCaT keratinocytes with a S. aureus suspension was used as an in vitro model of skin infection. Treatment of S. aureus-infected HaCaT cells with meroantarctine A at 10 µM caused a reduction in the production of TNF-α, IL-18, NO, and ROS, as well as LDH release and caspase 1 activation in these cells and, finally, recovered the proliferation and migration of HaCaT cells in an in vitro wound healing assay up to the control level. Thus, meroantarctine A is a new promising antibiofilm compound which can effective against S. aureus caused skin infection.
Collapse
Affiliation(s)
- Ekaterina A Chingizova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia.
| | - Artur R Chingizov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia
| | | | - Evgeny A Pislyagin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia
| | - Aleksandra S Kuzmich
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia
| | - Elena V Leshchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia
| | - Gleb V Borkunov
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia
| | | | - Dmitry L Aminin
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, 80708, Kaohsiung, Taiwan
| | - Ekaterina A Yurchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry FEB RAS, 690022, Vladivostok, Russia.
| |
Collapse
|
23
|
Truong YL, Rosenblatt J, Gerges B, Jiang Y, Raad I. Eradication of Candida auris biofilm in vitro by a polygalacturonic and caprylic acid wound ointment. J Mycol Med 2024; 34:101519. [PMID: 39549521 DOI: 10.1016/j.mycmed.2024.101519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
Candida auris is a rapidly spreading virulent pathogen frequently resistant to multiple antifungal drugs that can form biofilms and infect wounds. Hence, there is a need for C. auris wound treatments not posing risks for developing antifungal resistance. We tested the ability of a polygalacturonic and caprylic acid ointment (PG+CAP) to rapidly eradicate C. auris biofilms within 2-hour exposures in an in vitro model. Medical-grade honey (MediHoney) wound ointment was used as a comparator. Nine different C. auris strains were tested. PG+CAP eradicated biofilms of 8 of the 9 tested strains and produced a > 5-log10 reduction of the ninth. MediHoney produced reductions ranging from 2 to 4 log10 without fully eradicating any strains. The differences between PG+CAP and MediHoney were statistically significant (p < 0.05). These results suggest that PG+CAP is a promising antimicrobial ointment warranting further in vivo study in wounds which may be colonized by C. auris biofilms.
Collapse
Affiliation(s)
- Y-Lan Truong
- Department of Infectious Disease, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, United States.
| | - Joel Rosenblatt
- Department of Infectious Disease, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, United States
| | - Bahgat Gerges
- Department of Infectious Disease, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, United States
| | - Ying Jiang
- Department of Infectious Disease, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, United States
| | - Issam Raad
- Department of Infectious Disease, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd., Houston, TX, United States
| |
Collapse
|
24
|
Osiro KO, Hashemi N, Brango-Vanegas J, Oliveira SMD, Franco OL. Emerging peptide-based technology for biofilm control. Expert Opin Biol Ther 2024:1-5. [PMID: 39548688 DOI: 10.1080/14712598.2024.2430623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Affiliation(s)
- Karen O Osiro
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Nona Hashemi
- Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, USA
| | - José Brango-Vanegas
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Samuel M D Oliveira
- Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, Greensboro, NC, USA
| | - Octavio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| |
Collapse
|
25
|
Visvalingam J, Yakandawala N, Regmi S, Adeniji A, Sharma P, Sailer M. Wound Gel Formulations Containing Poloxamer 407 and Polyhexanide Have In Vitro Antimicrobial and Antibiofilm Activity Against Wound-Associated Microbial Pathogens. Microorganisms 2024; 12:2362. [PMID: 39597749 PMCID: PMC11596435 DOI: 10.3390/microorganisms12112362] [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: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024] Open
Abstract
Chronic wounds are often caused or exacerbated by microbial biofilms that are highly resistant to antimicrobial treatments and that prevent healing. This study compared the antimicrobial and antibiofilm activity of nine topical wound treatments, comprising gels with different concentrations of poloxamer 407 (20-26%) and different pH levels (4-6) and containing polyhexanide (PHMB) as an antimicrobial agent; the effects of pH on wound gels containing this agent have not been previously reported. The wound gel formulations were tested against six common wound-associated microbial pathogens: Staphylococcus aureus, S. epidermidis, Pseudomonas aeruginosa, Escherichia coli, Acinetobacter baumannii, and Candida albicans. Time-kill assays were used to assess antimicrobial activity against planktonic forms of each species, and a colony biofilm model was used to assess antibiofilm activity against existing biofilms as well as inhibition of new biofilm formation. Biofilm inhibition activity was also assessed in the presence of common wound dressing materials. Wound gels with higher pH levels exhibited stronger antimicrobial activity, while poloxamer 407 concentrations >20% negatively impacted antimicrobial activity. Wound gel formulations were identified that had antimicrobial, antibiofilm, and biofilm inhibition activity against all tested species in vitro. Biofilm inhibition activity was not affected by contact with common wound dressings. Further development of these wound gels may provide a valuable new option for the treatment and prevention of chronic wounds.
Collapse
|
26
|
Tesfaye S, Tafere W, Abebe W, Wondimeneh Y. Prevalence of Extended-Spectrum Beta-Lactamase Producer Gram-Negative Rods and Associated Factors Among Patients With Wound Infection at University of Gondar Comprehensive Specialized Hospital, Northwest Ethiopia. Int J Microbiol 2024; 2024:1478975. [PMID: 39559163 PMCID: PMC11573444 DOI: 10.1155/2024/1478975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024] Open
Abstract
Background: Multidrug-resistant (MDR) bacteria have significantly affected the management and treatment of wound infections globally. Data on the prevalence of MDR bacterial profiles that cause wound infections in Ethiopia are scarce. Therefore, this study aimed to determine MDR as well as extended-spectrum beta-lactamase production profiles of Gram-negative rods that are difficult to treat with conventional antibiotics and that cause wound infections. Objective: The aim of the study was to determine the prevalence of extended-spectrum beta-lactamase producer Gram-negative rods and associated factors among patients with wound infection at University of Gondar Comprehensive Specialized Hospital, northwest Ethiopia. Materials and Methods: This hospital-based cross-sectional study was conducted at University of Gondar Comprehensive Specialized Hospital between May and July 2022. Convenience sampling was used to recruit 228 participants. Swabs from different wound types were inoculated onto the MacConkey agar and blood agar plates and incubated overnight at 37°C for 24 h. Biochemical tests were performed on isolated colonies for the identification of bacterial species based on their biochemical reaction. Antimicrobial susceptibility tests were performed using the disk diffusion technique as per the standard Kirby-Bauer method by using Muller-Hinton agar, and the zone of inhibition was interpreted as resistant, intermediate, and sensitive as per the recommendation of Clinical Laboratory Standard Institute. Isolates were tested against ceftriaxone, cefotaxime, and ceftazidime for extended-spectrum beta-lactamase screening using the Kirby-Bauer disk diffusion method, and combined disk tests were applied for phenotypic confirmatory test of extended-spectrum beta-lactamase producing isolates. Result: Of 228 study participants, 162 (71.1%) were culture-positive. Among culture-positive patients, 165 Gram-negative bacteria were recovered. The most common Gram-negative isolates were Pseudomonas aeruginosa (47; 28.5%), followed by Escherichia coli (43; 26.1%) and Klebsiella pneumoniae (24; 14.5%). The susceptibility rates of the isolate for imipenem and tobramycin were 97.0% and 73.2%, respectively, and the overall multidrug resistance rate was 80.5%. Extended-spectrum beta-lactamase producer bacteria were also isolated. Besides, all (100%) of extended-spectrum beta-lactamase producer bacteria were MDR. Living in rural areas (AOR 5.8; 95% CI [2.01-16.7]), hospital admission (AOR 3.95; 95% CI [1.13-13.83]), antibiotic use (AOR 2.83; 95% CI [1.03-7.72]), and comorbidity (AOR 0.25; 95% CI [0.07-0.59]) were significantly associated with wound infection. Conclusions and Recommendations: There was a high prevalence of Gram-negative bacterial isolates in this study. Pseudomonas aeruginosa (28.5%) was the predominant isolate. In addition, high rates of multidrug resistance were observed. The high level of multidrug resistance in this study implies that definitive therapy should be based on culture and susceptibility analysis to promote the rational use of antibiotics and to reduce the emergence of bacterial resistance to antimicrobials.
Collapse
Affiliation(s)
- Sara Tesfaye
- Department of Medical Microbiology, School of Medicine, College of Medicine and Health Sciences, Jigjiga University, Jigjiga, Ethiopia
| | - Wudu Tafere
- Department of Medical Microbiology, Amhara National Regional State Public Health Institute, Bahirdar, Ethiopia
| | - Wondwossen Abebe
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayih Wondimeneh
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
27
|
Leaper D, Bond-Smith G, Edmiston C. Biofilms and antibacterial sutures. J Wound Care 2024; 33:850-851. [PMID: 39480733 DOI: 10.12968/jowc.2024.0302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Affiliation(s)
- David Leaper
- University of Newcastle, UK; University of Huddersfield, UK
| | | | | |
Collapse
|
28
|
Stewart PS, Kim J, James G, Yi F, Stechmiller J, Weaver M, Kelly DL, Fisher S, Schultz G, Lyon D. Association of biofilm and microbial metrics with healing rate in older adults with chronic venous leg ulcers. Wound Repair Regen 2024; 32:858-871. [PMID: 39425525 PMCID: PMC11585430 DOI: 10.1111/wrr.13230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/19/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
The presence of microbial biofilms in many human chronic wounds led to the hypothesis that biofilms delay healing of these wounds. We tested this hypothesis in a population of 117 older individuals with venous leg ulcers who were receiving standardised therapy, including frequent debridement. Debridement specimens were analysed for the amount of bacterial biomass by two independent methods: a microscopic approach that scored the relative size and number of bacterial aggregates, interpreted as a biofilm metric, and conventional enumeration by agar plating for viable bacteria. The plating protocol yielded three distinct values: the total viable bacterial count, bleach-tolerant bacteria, and the log reduction in viable bacteria upon bleach treatment. Wound healing rates over an 8-week observation period were calculated as the rate of decrease of the equivalent diameter of the wound. There was no statistically significant association between wound healing and the biofilm metric in any of the three analyses performed (p ≥0.15). In all three statistical tests, wound healing was associated with the log reduction caused by bleach treatment (p ≤0.004); wounds that harboured bacteria that were more bleach-susceptible healed more slowly. A refinement of the model of chronic wound infection pathogenesis is proposed in which dormant bacteria constitute a persistent nidus and outgrowth of metabolically active cells impairs healing. This model constitutes a new hypothesis as metabolic activity was not directly measured in this investigation.
Collapse
Affiliation(s)
- Philip S. Stewart
- Center for Biofilm Engineering, Montana State University
- Department of Chemical and Biological Engineering, Montana State University
| | - Junglyun Kim
- Chungnam National University College of Nursing
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Garth James
- Center for Biofilm Engineering, Montana State University
- Department of Chemical and Biological Engineering, Montana State University
| | - Fan Yi
- Department of Mathematics and Statistical Science, University of Idaho
| | - Joyce Stechmiller
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Michael Weaver
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Debra L. Kelly
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| | - Steve Fisher
- Center for Biofilm Engineering, Montana State University
| | | | - Debra Lyon
- Department of Biobehavioral Nursing Science, University of Florida College of Nursing
| |
Collapse
|
29
|
Gunasegaran N, Goh WT, Tan WX, Saipollah H, Chong HR, Sunari RN, Chong TT, Ang SY, Aloweni F. Patients with venous leg ulcers can be managed safely in the community ----results of an observational comparison study in Singapore. J Tissue Viability 2024; 33:561-570. [PMID: 39164151 DOI: 10.1016/j.jtv.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/22/2024]
Abstract
AIM To examine the healing outcomes of patients with venous leg ulcers requiring compression bandaging in community care versus tertiary care. METHOD This was an analytical observational cohort study. Venous leg ulcer (VLU) patients who required compression bandaging were recruited from an outpatient vascular clinic between May 2021 and August 2022. Eligible patients received two-or four-layer compression bandaging and followed up with the community care or tertiary care centre nurses. The primary outcome was the difference in the total surface area of the VLU after 12 weeks, and the secondary outcome was the patient's quality of life, as measured by the Cardiff Wound Impact Schedule (CWIS). RESULTS Forty-seven VLU patients were recruited; 27 received compression bandaging in the community care and 20 by the tertiary care centre. Mean age 70 years old (SD 11.04). The two most prevalent comorbidities were hypertension (51.06 %) and diabetes mellitus (38.29 %). Among those who completed follow-up (12 weeks), the median difference of the total surface area of the VLU between community-based care (p = 0.02) versus tertiary-based care (0.003) was significant. However, there was no difference in the healing status between community and tertiary-based care (p = 0.68). There was no difference in the quality of life of patients between groups. CONCLUSION This first tropical study comparing VLU healing outcomes between community and tertiary care found no significant difference in healing with compression bandaging by nurses in either setting. However, the small sample size and high dropout rate limit the generalizability of the findings, necessitating a larger-scale study with longer follow-up. Despite these limitations, the study is a crucial step toward improving wound care services in Singapore, and highlights the need for further research to guide future community wound care implementation.
Collapse
Affiliation(s)
| | - Wee Ting Goh
- Nursing Division, Singapore General Hospital, Singapore.
| | | | | | - Hui Ru Chong
- Nursing Division, Singapore General Hospital, Singapore.
| | | | - Tze Tec Chong
- Department of Vascular Surgery, Singapore General Hospital, Singapore.
| | - Shin Yuh Ang
- Nursing Division, Singapore General Hospital, Singapore.
| | - Fazila Aloweni
- Nursing Division, Singapore General Hospital, Singapore.
| |
Collapse
|
30
|
Coluccio A, Lopez Palomera F, Spero MA. Anaerobic bacteria in chronic wounds: Roles in disease, infection and treatment failure. Wound Repair Regen 2024; 32:840-857. [PMID: 39129662 DOI: 10.1111/wrr.13208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/09/2024] [Accepted: 07/29/2024] [Indexed: 08/13/2024]
Abstract
Infection is among the most common factors that impede wound healing, yet standard treatments routinely fail to resolve chronic wound infections. The chronic wound environment is largely hypoxic/anoxic, and wounds are predominantly colonised by facultative and obligate anaerobic bacteria. Oxygen (O2) limitation is an underappreciated driver of microbiota composition and behaviour in chronic wounds. In this perspective article, we examine how anaerobic bacteria and their distinct physiologies support persistent, antibiotic-recalcitrant infections. We describe the anaerobic energy metabolisms bacteria rely on for long-term survival in the wound environment, and why many antibiotics become less effective under hypoxic conditions. We also discuss obligate anaerobes, which are among the most prevalent taxa to colonise chronic wounds, yet their potential roles in influencing the microbial community and wound healing have been overlooked. All of the most common obligate anaerobes found in chronic wounds are opportunistic pathogens. We consider how these organisms persist in the wound environment and interface with host physiology to hinder wound healing processes or promote chronic inflammation. Finally, we apply our understanding of anaerobic physiologies to evaluate current treatment practices and to propose new strategies for treating chronic wound infections.
Collapse
Affiliation(s)
- Alison Coluccio
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, USA
| | | | - Melanie A Spero
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, USA
| |
Collapse
|
31
|
Frison SS, Borges EL, Guedes ACM, Honorato-Sampaio K. Biofilm and Its Characteristics in Venous Ulcers. J Wound Ostomy Continence Nurs 2024; 51:445-453. [PMID: 39588812 DOI: 10.1097/won.0000000000001123] [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: 11/27/2024]
Abstract
PURPOSE The aim of the study was to analyze the characteristics of the biofilm of venous ulcers in terms of location and formation and to relate the presence of the biofilm to ulcer characteristics including duration, injured area, and necrotic tissue. DESIGN Descriptive clinical study. MATERIALS AND METHODS We obtained 2 biopsy fragments (tissue samples) from 44 patients with venous ulcers treated at a public outpatient clinic in a university hospital in Belo Horizonte, Brazil. Ulcers were photographed and classified according to the duration. In addition, the wound size and proportion of wound surface covered by necrotic tissue were measured. One fragment from each ulcer underwent microbiological analysis, while the other was analyzed using transmission electron microscopy. Data analysis was limited to fragments from patients with bacteria in the microbiological analysis. RESULTS Data analysis is based on samples obtained from 21 ulcers in 21 patients who had bacteria in their ulcer based on microbiologic analysis of a tissue sample. Most ulcers were open for 2 to 10 years, 57% (n = 12) were 16 cm2 or smaller, and the proportion of the wound bed covered by necrotic tissue coverage varied widely. Of the 21/44 patients (48%) with bacteria in their ulcers, only 3 patients had bacterial biofilm present in the transmission electron microscopy, corresponding to 7% of the 44 patients. Pseudomonas aeruginosa was the most frequent bacterium, identified in 10 fragments. The biofilm was not present on the surface but in a layer slightly below it. The detection of biofilms was not directly related to the duration of the ulcer. It was not possible to establish a correlation between the size of the lesion and the presence of these microorganisms due to the small sample size. CONCLUSIONS Our findings indicate that detecting biofilm in venous ulcers is challenging, as it does not uniformly occur throughout the wound bed, can occur at different depths, and is often not present on the wound surface. There is a need to develop studies that can contribute to the detection of biofilm in clinical practice.
Collapse
Affiliation(s)
- Susiane Sucasas Frison
- Susiane Sucasas Frison, MSc, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Eline Lima Borges, PhD, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Antônio Carlos Martins Guedes, PhD, MD, Department of Internal Medicine, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Kinulpe Honorato-Sampaio, PhD, Faculdade de Medicina da Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brazil
| | - Eline Lima Borges
- Susiane Sucasas Frison, MSc, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Eline Lima Borges, PhD, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Antônio Carlos Martins Guedes, PhD, MD, Department of Internal Medicine, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Kinulpe Honorato-Sampaio, PhD, Faculdade de Medicina da Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brazil
| | - Antônio Carlos Martins Guedes
- Susiane Sucasas Frison, MSc, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Eline Lima Borges, PhD, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Antônio Carlos Martins Guedes, PhD, MD, Department of Internal Medicine, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Kinulpe Honorato-Sampaio, PhD, Faculdade de Medicina da Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brazil
| | - Kinulpe Honorato-Sampaio
- Susiane Sucasas Frison, MSc, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Eline Lima Borges, PhD, Department of Basic Nursing, Escola de Enfermagem da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Antônio Carlos Martins Guedes, PhD, MD, Department of Internal Medicine, Faculdade de Medicina da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Kinulpe Honorato-Sampaio, PhD, Faculdade de Medicina da Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brazil
| |
Collapse
|
32
|
Kulayta K, Zerdo Z, Seid M, Dubale A, Manilal A, Kebede T, Alahmadi RM, Raman G, Akbar I. Biofilm formation and antibiogram profile of bacteria from infected wounds in a general hospital in southern Ethiopia. Sci Rep 2024; 14:26359. [PMID: 39487302 PMCID: PMC11530625 DOI: 10.1038/s41598-024-78283-9] [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: 03/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
Biofilm-producing bacteria associated with wound infections exhibit exceptional drug resistance, leading to an escalation in morbidity, worse clinical outcomes (including delay in the healing process), and an increase in health care cost, burdening the whole system. This study is an attempt to estimate the prevalence and the relationship between the biofilm-forming capacity and multi-drug resistance of wound bacterial isolates. The findings intended to help clinicians, healthcare providers and program planners and to formulate an evidence-based decision-making process, especially in resource-limited healthcare settings. This study was done to assess the prevalence of bacterial infections in wounds and the antibiogram and biofilm-forming capacity of those bacteria in patients with clinical signs and symptoms, attending a General Hospital in southern Ethiopia. A cross-sectional study was performed in Arba Minch General Hospital from June to November 2021. The study participants comprised 201 patients with clinically infected wounds. Demographic and clinical data were gathered via a structured questionnaire. Specimens from wounds were taken from each participant and inoculated onto a series of culture media, namely MacConkey agar, mannitol salt agar, and blood agar, and different species were identified using a number of biochemical tests. Antimicrobial susceptibility tests were performed by means of the Kirby-Bauer disc diffusion technique following the guidelines of the Clinical and Laboratory Standards Institute. A micro-titer plate method was employed to detect the extent of biofilm formation. Bivariable and multivariable logistic regression models were applied to analyse the association between dependent and independent variables, and P values ≤ 0.05 were considered as statistically significant. Data analyses were done with Statistical Package for the Social Sciences version 25. Out of the 201 clinically infected wounds, 165 were found culture-positive with an overall prevalence of 82% (95% CI: 75.9-86.9). In total, 188 bacteria were recovered; 53.1% of them were Gram-positive cocci. The often-isolated bacterial species were Staphylococcus aureus, 38.3% (n = 72), and Pseudomonas aeruginosa, 16.4% (n = 31). The Gram-positive isolates showed considerable resistance against penicillin, 70%, and somewhat strong resistance against tetracycline, 57.7%. Gram-negative isolates showed severe resistance to ampicillin, 80.68%. The overall multi-drug resistance (MDR) among isolates was 48.4%. Extended beta-lactamase (ESBL)-producing Gram-negatives and methicillin-resistant Staphylococcus aureus (MRSA) accounted for 49 and 41.67%, respectively; 62.2% of the isolates were biofilm formers and were correlated statistically with MDR, ESBL producers, and MRSA (P < 0.005). The extent of biofilm formation and the prevalence of MDR bacteria associated with infected wounds hint at a public health threat that needs immediate attention. Thus, a more balanced and comprehensive wound management approach and antimicrobial stewardship program are essential in the study setting.
Collapse
Affiliation(s)
- Kebede Kulayta
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
- Department of Medical Laboratory Science, Arba Minch College of Health Sciences, Arba Minch, Ethiopia
| | - Zerihun Zerdo
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
| | - Mohammed Seid
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia.
| | - Amanuel Dubale
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
| | - Aseer Manilal
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia.
| | - Teshome Kebede
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Arba Minch University, Arba Minch, southern, Ethiopia
- Arba Minch General Hospital, Arba Minch, Ethiopia
| | - Reham M Alahmadi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box 2455, 11451, Riyadh, Saudi Arabia
| | - Gurusamy Raman
- Department of Life Sciences, Yeungnam University, Gyeongsan, Gyeongbuk-Do, South Korea.
| | - Idhayadhulla Akbar
- Research Department of Chemistry, Nehru Memorial College (Affiliated to Bharathidasan University), Puthanampatti, Tiruchirappalli District, 621007, Tamil Nadu, India
| |
Collapse
|
33
|
Ma X, Dong Z, Mao R, Tian X, Yang N, Ren W, Hao Y, Shen W, Teng D, Li X, Wang J. Therapeutic Potential of Insect Defensin DLP4 Against Staphylococcus hyicus-Infected Piglet Exudative Epidermitis. Pharmaceutics 2024; 16:1350. [PMID: 39598475 PMCID: PMC11597436 DOI: 10.3390/pharmaceutics16111350] [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: 09/20/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: The emergence of resistance to Staphylococcus hyicus (S. hyicus), the major cause of exudative epidermatitis (EE) in piglets, has led to the need for new antimicrobial agents. The study aimed to evaluate the potential efficacy of the insect defensin DLP4 against EE in piglets caused by clinically isolated S. hyicus ACCC 61734. Methods and Results: DLP4 showed strong antibacterial activity against S. hyicus ACCC 61734 (minimum inhibitory concentration, MIC: 0.92 μM, median effect concentration, EC50: 3.158 μM). DLP4 could effectively inhibit the formation of S. hyicus early biofilm with an inhibition rate of 95.10-98.34% and eradicate mature biofilm with a clearance rate of 82.09-86.41%, which was significantly superior to that of ceftriaxone sodium (CRO). Meanwhile, DLP4 could efficiently inhibit bacteria in early and mature biofilm, killing up to 95.3% of bacteria in early biofilm and 87.2-90.3% of bacteria in mature biofilm. The results showed that DLP4 could be effective in alleviating the clinical symptoms of EE by down-regulating the nuclear factor κB (NF-κB) signaling pathway, balancing cytokines, inhibiting bacterial proliferation, and reducing organ tissue damage. Conclusions: This study firstly demonstrated the potential efficacy of DLP4 against EE caused by S. hyicus ACCC 61734 infection in piglets, which may be used as an alternative to antibiotics in treating EE.
Collapse
Affiliation(s)
- Xuanxuan Ma
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.M.); (R.M.); (N.Y.); (Y.H.); (W.S.)
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zhimin Dong
- Tianjin Animal Science and Veterinary Research Institute, Tianjin 300381, China (X.T.); (W.R.); (X.L.)
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.M.); (R.M.); (N.Y.); (Y.H.); (W.S.)
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiangxue Tian
- Tianjin Animal Science and Veterinary Research Institute, Tianjin 300381, China (X.T.); (W.R.); (X.L.)
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.M.); (R.M.); (N.Y.); (Y.H.); (W.S.)
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Weike Ren
- Tianjin Animal Science and Veterinary Research Institute, Tianjin 300381, China (X.T.); (W.R.); (X.L.)
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.M.); (R.M.); (N.Y.); (Y.H.); (W.S.)
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Wenluan Shen
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.M.); (R.M.); (N.Y.); (Y.H.); (W.S.)
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.M.); (R.M.); (N.Y.); (Y.H.); (W.S.)
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiuli Li
- Tianjin Animal Science and Veterinary Research Institute, Tianjin 300381, China (X.T.); (W.R.); (X.L.)
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (X.M.); (R.M.); (N.Y.); (Y.H.); (W.S.)
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
34
|
Iversen AKS, Lichtenberg M, Fritz BG, Díaz-Pinés Cort I, Al-Zoubaidi DF, Gottlieb H, Kirketerp-Møller K, Bjarnsholt T, Jakobsen TH. The chronic wound characterisation study and biobank: a study protocol for a prospective observational cohort investigation of bacterial community composition, inflammatory responses and wound-healing trajectories in non-healing wounds. BMJ Open 2024; 14:e084081. [PMID: 39419618 PMCID: PMC11487800 DOI: 10.1136/bmjopen-2024-084081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
INTRODUCTION Chronic wounds affect 1%-2% of the global population, with rising incidence due to ageing and lifestyle-related diseases. Bacterial biofilms, found in 80% of chronic wounds, and scattered single-cell bacteria may hinder healing. Microbes are believed to negatively impact healing by exacerbating inflammation and host immune response. METHODS AND ANALYSIS The primary objective of the chronic wound characterisation (CWC) study is to investigate chronic wounds through a prospective observational cohort study exploring bacterial community composition, inflammatory responses and the influence of bacteria on wound-healing trajectories. The CWC study will be investigated through two cohorts: the predictive and in-depth.The predictive cohort includes patients with a chronic wound scheduled for mechanical debridement. The debrided material will be collected for dual RNA sequencing and 16s ribosomal RNA gene sequencing, as well as samples for microbial culturing and a photo to assess the wound. Clinical data is recorded, and healing and/or other clinical endpoints are established through medical records.The in-depth cohort includes and follows patients undergoing split-thickness skin grafting. Extensive sampling (ESwabs, biopsies, tape strips, debrided material and a sample of the skin graft) will be performed on surgery and patients will be seen at two follow-up visits. Samples will be analysed through culturing and next-generation sequencing methods. A biobank will be established comprising longitudinal clinical samples and clinical data. ETHICS AND DISSEMINATION The study has been approved by the board of health ethics, Capital Region of Denmark, under protocol number H-20032214. The study findings will be disseminated through peer-reviewed publications and showcased at both national and international conferences and meetings within the domains of microbiology, wound healing and infection.
Collapse
Affiliation(s)
| | - Mads Lichtenberg
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Blaine Gabriel Fritz
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Isabel Díaz-Pinés Cort
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Dania Firas Al-Zoubaidi
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Hans Gottlieb
- Department of Orthopaedic Surgery, Herlev Hospital, Herlev, Denmark
| | - Klaus Kirketerp-Møller
- Copenhagen Wound Healing Centre, University Hospital of Copenhagen, Bispebjerg, Copenhagen, Denmark
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Tim Holm Jakobsen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Liu JD, VanTreeck KE, Marston WA, Papadopoulou V, Rowe SE. Ultrasound-Mediated Antibiotic Delivery to In Vivo Biofilm Infections: A Review. Chembiochem 2024; 25:e202400181. [PMID: 38924307 PMCID: PMC11483220 DOI: 10.1002/cbic.202400181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 06/28/2024]
Abstract
Bacterial biofilms are a significant concern in various medical contexts due to their resilience to our immune system as well as antibiotic therapy. Biofilms often require surgical removal and frequently lead to recurrent or chronic infections. Therefore, there is an urgent need for improved strategies to treat biofilm infections. Ultrasound-mediated drug delivery is a technique that combines ultrasound application, often with the administration of acoustically-active agents, to enhance drug delivery to specific target tissues or cells within the body. This method involves using ultrasound waves to assist in the transportation or activation of medications, improving their penetration, distribution, and efficacy at the desired site. The advantages of ultrasound-mediated drug delivery include targeted and localized delivery, reduced systemic side effects, and improved efficacy of the drug at lower doses. This review scrutinizes recent advances in the application of ultrasound-mediated drug delivery for treating biofilm infections, focusing on in vivo studies. We examine the strengths and limitations of this technology in the context of wound infections, device-associated infections, lung infections and abscesses, and discuss current gaps in knowledge and clinical translation considerations.
Collapse
Affiliation(s)
- Jamie D. Liu
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Kelly E. VanTreeck
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - William A. Marston
- Department of Surgery, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Virginie Papadopoulou
- Joint Department of Biomedical Engineering, The University of North Carolina and North Carolina State University, Chapel Hill, North Carolina 27599, USA
- Department of Radiology, The University of North Carolina at Chapel Hill, NC, USA
| | - Sarah E. Rowe
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
36
|
Abdollahi M, Baharian A, Mohamadhoseini M, Hassanpour M, Makvandi P, Habibizadeh M, Jafari B, Nouri R, Mohamadnia Z, Nikfarjam N. Advances in ionic liquid-based antimicrobial wound healing platforms. J Mater Chem B 2024; 12:9478-9507. [PMID: 39206539 DOI: 10.1039/d4tb00841c] [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: 09/04/2024]
Abstract
Wound infections, marked by the proliferation of microorganisms at surgical sites, necessitate the development of innovative wound dressings with potent bactericidal properties to curb microbial growth and prevent bacterial infiltration. This study explores the recent strides in utilizing ionic liquid-based polymers as highly promising antimicrobial agents for advanced wound healing applications. Specifically, cationic polymers containing quaternary ammonium, imidazolium, guanidinium, pyridinium, triazolium, or phosphonium groups have emerged as exceptionally effective antimicrobial compounds. Their mechanism of action involves disrupting bacterial membranes, thereby preventing the development of resistance and minimizing toxicity to mammalian cells. This comprehensive review not only elucidates the intricate dynamics of the skin's immune response and the various stages of wound healing but also delves into the synthesis methodologies of ionic liquid-based polymers. By spotlighting the practical applications of antimicrobial wound dressings, particularly those incorporating ionic liquid-based materials, this review aims to lay the groundwork for future research endeavors in this burgeoning field. Through a nuanced examination of these advancements, this article seeks to contribute to the ongoing progress in developing cutting-edge wound healing platforms that can effectively address the challenges posed by microbial infections in surgical wounds.
Collapse
Affiliation(s)
- Mahin Abdollahi
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Aysan Baharian
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Masoumeh Mohamadhoseini
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Mahnaz Hassanpour
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Pooyan Makvandi
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3JL, UK
| | - Mina Habibizadeh
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Bahman Jafari
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Roya Nouri
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Zahra Mohamadnia
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
| | - Nasser Nikfarjam
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 451951159, Iran.
- Department of Chemical Engineering, College of Engineering and Computing, University of South Carolina, Columbia 29208, SC, USA
| |
Collapse
|
37
|
Münter KC, Lázaro-Martínez JL, Kanya S, Sawade L, Schwenke C, Pegalajar-Jurado A, Swanson T, Leaper D. Clinical efficacy and safety of a silver ion-releasing foam dressing on hard-to-heal wounds: a meta-analysis. J Wound Care 2024; 33:726-736. [PMID: 39388210 DOI: 10.12968/jowc.2024.0149] [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: 10/12/2024]
Abstract
OBJECTIVE Delayed or stalled healing in open wounds can result from persisting chronic inflammation related to infection and/or persistent bacterial colonisation and biofilm. Treatment of hard-to-heal wounds focuses on debridement and exudate management, but also on infection prevention and control. Silver dressings have been evaluated in randomised clinical trials (RCTs); this meta-analysis evaluated the efficacy and safety of a silver ion-releasing foam dressing (Biatain Ag; Coloplast A/S, Denmark) to treat hard-to-heal wounds. METHOD Literature databases (PubMed and Cochrane Library) were searched for studies on silver ion-releasing foam dressings in the treatment of hard-to-heal wounds. Individual patient data from four RCTs were obtained and included in the meta-analysis. RESULTS Findings showed that treatment with the silver ion-releasing foam dressing was associated with a significantly higher relative reduction in wound area after four (least squares-mean difference (LS-MD): -12.55%, 95% confidence interval (CI): (-15.95, -9.16); p<0.01) and six weeks of treatment (LS-MD: -11.94%, 95%CI: (-17.21, -6.68); p<0.01) compared with controls. Significant benefits were also observed for time to disappearance of odour (hazard ratio: 1.61, 95%CI: (1.31, 1.98); p<0.01), relative reduction of exudate (LS-MD: -5.15, 95%CI: (-7.36, -2.94); p<0.01), proportion of patients with periwound erythema (relative risk (RR): 0.81, 95%CI: (0.69; 0.94); p<0.01), and less pain at dressing removal (LS-MD: -0.35, 95%CI: (-0.63, -0.06); p=0.02). No differences regarding safety outcomes were identified. CONCLUSION This meta-analysis has demonstrated beneficial outcomes and a good tolerability profile for silver ion-releasing foam dressings in the treatment of moderate-to-highly exuding wounds with delayed healing compared with control dressings.
Collapse
Affiliation(s)
| | | | - Susanne Kanya
- Wound and Skin Care Region DACH, Coloplast GmbH, Hamburg, Germany
| | | | | | | | - Terry Swanson
- South West Healthcare, Warrnambool, Victoria, Australia
| | - David Leaper
- University of Newcastle, UK
- University of Huddersfield, UK
| |
Collapse
|
38
|
Schwarzer S, Martinez JL, Killeen A, Alves P, Gledhill A, Nygren E, Lavery LA, Malone M. Does the use of DACC-coated dressings improve clinical outcomes for hard to heal wounds: A systematic review. Int Wound J 2024; 21:e70053. [PMID: 39362798 PMCID: PMC11449527 DOI: 10.1111/iwj.70053] [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: 06/14/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
Reports of overuse and antimicrobial resistance have fuelled some clinicians to adopt alternative wound dressings termed to be non-medicated or non-antimicrobials, which still claim antimicrobial or antibacterial activity. In this PROSPERO-registered systematic review, we evaluated the in vivo clinical evidence for the effectiveness of DACC-coated dressings in chronic, hard to heal wound-related outcomes. The Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) Framework was adopted as the template in constructing this systematic review. The PICO format (Population [or patients], Intervention, Comparison [control], Outcome/s) was used to identify key clinical questions in determining patient outcomes under two domains (infection control and wound healing). A systematic search was performed in PubMed, OVID, Cochrane Library, clinical trial registries and data sources from independent committees. Abstracts of all studies were screened independently by two reviewers, with six further reviewers independently assessing records proceeding to full review. The authors rated the quality of evidence for each of the outcomes critical to decision making. After excluding duplicates, 748 records were screened from the databases, and 13 records were sought for full review. After full review, we excluded a further three records, leaving ten records for data extraction. Three records were narrative reviews, three systematic reviews, two prospective non-comparative before/after studies, one prospective head-to-head comparator cohort study and one retrospective head-to-head comparator cohort study. No RCTs or case versus control studies were identified. The overall quality of clinical evidence for the use of DACC-coated dressing to improve wound infection and wound healing outcomes was assessed as very low. There is an urgent unmet need to perform appropriately designed RCTs or case-control studies. The extracted data provide no clarity and have limited to no evidence to support that using a DACC-coated dressing improves wound infection or wound healing outcomes. Further, there is no evidence to suggest this therapy is either superior to standard of wound care or equivocal to topical antimicrobial agents in the management of infected hard to heal wounds.
Collapse
Affiliation(s)
- Saskia Schwarzer
- South Western Sydney Local Health DistrictLiverpool HospitalSydneyNew South WalesAustralia
- South West Sydney Limb Preservation and Wound Research Academic UnitIngham Institute of Applied Medical ResearchSydneyNew South WalesAustralia
| | - Jose Lazaro Martinez
- Complutense University of Madrid, Diabetic Foot UnitUniversity Clinic of PodiatryMadridSpain
| | - Amanda Killeen
- School of Podiatric MedicineUniversity of Texas Rio Grande ValleyHarlingenTexasUSA
| | - Paulo Alves
- Wounds Research Laboratory, Institute of Health Sciences, Center for Interdisciplinary Research in HealthUniversidade Católica PortuguesaLisboaPortugal
| | - Andrea Gledhill
- Great Western Hospital NHS Foundation TrustDepartment of Trauma and OrthopaedicsSwindonUK
| | - Erik Nygren
- Wound Care Research and Development, Mölnlycke Health Care ABGothenburgSweden
| | - Lawrence A. Lavery
- Department of Plastic SurgeryUniversity of Texas Southwestern Medical CentreDallasTexasUSA
| | - Matthew Malone
- Wound Care Research and Development, Mölnlycke Health Care ABGothenburgSweden
- Infectious Diseases and Microbiology, School of MedicineWestern Sydney UniversitySydneyNew South WalesAustralia
| |
Collapse
|
39
|
Tyagi A, Kumar V, Joshi N, Dhingra HK. Combinatorial Effects of Ursodeoxycholic Acid and Antibiotic in Combating Staphylococcus aureus Biofilm: The Roles of ROS and Virulence Factors. Microorganisms 2024; 12:1956. [PMID: 39458266 PMCID: PMC11509559 DOI: 10.3390/microorganisms12101956] [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: 08/26/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Staphylococcus aureus is a biofilm-forming bacterium responsible for various human infections, one particularly challenging to treat due to its antibiotic resistance. Biofilms can form on both soft tissues and medical devices, leading to persistent and hard-to-treat infections. Combining multiple antimicrobials is a potential approach to overcoming this resistance. This study explored the effects of ursodeoxycholic acid (UDCA) combined with the antibiotic ciprofloxacin against S. aureus biofilms, aiming to evaluate any synergistic effects. Results showed that UDCA and ciprofloxacin co-treatment significantly reduced biofilm formation and disrupted pre-formed biofilms more effectively than either agent alone (p < 0.01). The combination also displayed a slight synergistic effect, with a fractional inhibitory concentration of 0.65. Additionally, the treatment reduced the production of extracellular polymeric substances, increased reactive oxygen species production, decreased metabolic activity, altered cell membrane permeability, and lowered cell surface hydrophobicity in S. aureus. Furthermore, it diminished biofilm-associated pathogenic factors, including proteolytic activity and staphyloxanthin production. Overall, the UDCA-ciprofloxacin combination shows considerable promise as a strategy to combat infections related to staphylococcal biofilms, offering a potential solution to the healthcare challenges posed by antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Anuradha Tyagi
- Department of Biosciences, School of Liberal Arts and Sciences, Mody University of Science and Technology, Lakshmangarh 332311, Rajasthan, India; (A.T.); (N.J.)
| | - Vinay Kumar
- Department of Medicine, Pennsylvania State University, Hershey Medical Center, Hershey, PA 17033, USA
| | - Navneet Joshi
- Department of Biosciences, School of Liberal Arts and Sciences, Mody University of Science and Technology, Lakshmangarh 332311, Rajasthan, India; (A.T.); (N.J.)
| | - Harish Kumar Dhingra
- Department of Biosciences, School of Liberal Arts and Sciences, Mody University of Science and Technology, Lakshmangarh 332311, Rajasthan, India; (A.T.); (N.J.)
| |
Collapse
|
40
|
El Zein S, Melin MM, Suh GA, Tran NV, Rose PS, Berbari EF. Evaluation and Management of Pelvic Osteomyelitis in Stage IV Pressure Injuries: A Multidisciplinary Collaborative Approach. Clin Infect Dis 2024; 79:e11-e26. [PMID: 39325647 DOI: 10.1093/cid/ciae394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Indexed: 09/28/2024] Open
Abstract
Managing pelvic osteomyelitis (POM) in the setting of stage IV pressure injuries requires multidisciplinary evaluation as well as patient and caregiver engagement and is complicated by the lack of high-evidence data to guide best practices. In this review, we describe our approach to pressure injury and POM evaluation and management through multidisciplinary collaboration and highlight areas of future research that are necessary to enhance patient outcomes, reduce healthcare costs, and improve the quality of life of those affected by POM.
Collapse
Affiliation(s)
- Said El Zein
- Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew M Melin
- Gonda Vascular Center, Wound Clinic, Mayo Clinic, Rochester, Minnesota, USA
| | - Gina A Suh
- Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - N V Tran
- Department of Plastic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter S Rose
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Elie F Berbari
- Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
41
|
Mayer P, Smith AC, Hurlow J, Morrow BR, Bohn GA, Bowler PG. Assessing Biofilm at the Bedside: Exploring Reliable Accessible Biofilm Detection Methods. Diagnostics (Basel) 2024; 14:2116. [PMID: 39410520 PMCID: PMC11475494 DOI: 10.3390/diagnostics14192116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 10/20/2024] Open
Abstract
INTRODUCTION Biofilm is linked through a variety of mechanisms to the pathogenesis of chronic wounds. However, accurate biofilm detection is challenging, demanding highly specialized and technically complex methods rendering it unapplicable for most clinical settings. This study evaluated promising methods of bedside biofilm localization, fluorescence imaging of wound bacterial loads, and biofilm blotting by comparing their performance against validation scanning electron microscopy (SEM). METHODS In this clinical trial, 40 chronic hard-to-heal wounds underwent the following assessments: (1) clinical signs of biofilm (CSB), (2) biofilm blotting, (3) fluorescence imaging for localizing bacterial loads, wound scraping taken for (4) SEM to confirm matrix encased bacteria (biofilm), and (5) PCR (Polymerase Chain Reaction) and NGS (Next Generation Sequencing) to determine absolute bacterial load and species present. We used a combination of SEM and PCR microbiology to calculate the diagnostic accuracy measures of the CSB, biofilm blotting assay, and fluorescence imaging. RESULTS Study data demonstrate that 62.5% of wounds were identified as biofilm-positive based on SEM and microbiological assessment. By employing this method to determine the gold truth, and thus calculate accuracy measures for all methods, fluorescence imaging demonstrated superior sensitivity (84%) and accuracy (63%) compared to CSB (sensitivity 44% and accuracy 43%) and biofilm blotting (sensitivity 24% and accuracy 40%). Biofilm blotting exhibited the highest specificity (64%), albeit with lower sensitivity and accuracy. Using SEM alone as the validation method slightly altered the results, but all trends held constant. DISCUSSION This trial provides the first comparative assessment of bedside methods for wound biofilm detection. We report the diagnostic accuracy measures of these more feasibly implementable methods versus laboratory-based SEM. Fluorescence imaging showed the greatest number of true positives (highest sensitivity), which is clinically relevant and provides assurance that no pathogenic bacteria will be missed. It effectively alerted regions of biofilm at the point-of-care with greater accuracy than standard clinical assessment (CSB) or biofilm blotting paper, providing actionable information that will likely translate into enhanced therapeutic approaches and better patient outcomes.
Collapse
Affiliation(s)
- Perry Mayer
- The Mayer Institute (TMI), Hamilton, ON L8R 2R3, Canada
| | - Allie Clinton Smith
- Department of Honors Studies, Texas Tech University, Lubbock, TX 79409, USA;
| | - Jennifer Hurlow
- Consultant Wound Care Specialized Nurse Practitioner, Memphis, TN 38120, USA;
| | - Brian R. Morrow
- College of Dentistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Gregory A. Bohn
- The American Professional Wound Care Association (APWCA), American Board of Wound Healing, Milwaukee, WI 53214, USA
| | | |
Collapse
|
42
|
Ball C, Jones H, Thomas H, Woodmansey E, Cole W, Schultz G. Impact of continuous topical oxygen therapy on biofilm gene expression in a porcine tissue model. J Wound Care 2024; 33:702-707. [PMID: 39287037 DOI: 10.12968/jowc.2024.0213] [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: 09/19/2024]
Abstract
OBJECTIVE The effect of continuous topical oxygen therapy (cTOT) on Pseudomonas aeruginosa biofilm gene transcription profiles following inoculation onto porcine skin, using a customised molecular assay was determined. METHOD Sterilised porcine skin explants were inoculated with Pseudomonas aeruginosa in triplicate: 0 hours as negative control; 24 hours cTOT device on; 24 hours cTOT device off. The oxygen delivery system of the cTOT device was applied to the inoculated tissue and covered with a semi-occlusive dressing. All samples were incubated at 37±2°C for 24 hours, with the 0 hours negative control inoculated porcine skin samples recovered immediately. Planktonic suspensions and porcine skin biopsy samples were taken at 0 hours and 24 hours. Samples were processed and quantifiably assessed using gene specific reverse transcription-quantitative polymerase chain reaction assays for a panel of eight Pseudomonas aeruginosa genes (16S, pelA, pslA, rsaL, pcrV, pscQ, acpP, cbrA) associated with biofilm formation, quorum sensing, protein secretion/translocation and metabolism. RESULTS Transcriptional upregulation of pelA, pcrV and acpP, responsible for intracellular adhesion, needletip protein production for type-3 secretion systems and fatty acid synthesis during proliferation, respectively, was observed when the cTOT device was switched on compared to when the device was switched off. Data suggest increased metabolic activity within bacterial cells following cTOT treatment. CONCLUSION cTOT is an adjunctive therapy that supports faster healing and pain reduction in non-healing hypoxic wounds. Oxygen has previously been shown to increase susceptibility of biofilms to antibiotics through enhancing metabolism. Observed gene expression changes highlighted the impact of cTOT on biofilms, potentially influencing antimicrobial treatment success in wounds. Further in vitro and clinical investigations are warranted.
Collapse
Affiliation(s)
| | | | | | | | - Windy Cole
- Natrox Wound Care (Inotec AMD Ltd.), Cambridge, UK
- College of Podiatric Medicine, Kent State University, OH, US
| | | |
Collapse
|
43
|
Cometta S, Hutmacher DW, Chai L. In vitro models for studying implant-associated biofilms - A review from the perspective of bioengineering 3D microenvironments. Biomaterials 2024; 309:122578. [PMID: 38692146 DOI: 10.1016/j.biomaterials.2024.122578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/01/2024] [Accepted: 04/13/2024] [Indexed: 05/03/2024]
Abstract
Biofilm research has grown exponentially over the last decades, arguably due to their contribution to hospital acquired infections when they form on foreign body surfaces such as catheters and implants. Yet, translation of the knowledge acquired in the laboratory to the clinic has been slow and/or often it is not attempted by research teams to walk the talk of what is defined as 'bench to bedside'. We therefore reviewed the biofilm literature to better understand this gap. Our search revealed substantial development with respect to adapting surfaces and media used in models to mimic the clinical settings, however many of the in vitro models were too simplistic, often discounting the composition and properties of the host microenvironment and overlooking the biofilm-implant-host interactions. Failure to capture the physiological growth conditions of biofilms in vivo results in major differences between lab-grown- and clinically-relevant biofilms, particularly with respect to phenotypic profiles, virulence, and antimicrobial resistance, and they essentially impede bench-to-bedside translatability. In this review, we describe the complexity of the biological processes at the biofilm-implant-host interfaces, discuss the prerequisite for the development and characterization of biofilm models that better mimic the clinical scenario, and propose an interdisciplinary outlook of how to bioengineer biofilms in vitro by converging tissue engineering concepts and tools.
Collapse
Affiliation(s)
- Silvia Cometta
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD 4000, Australia; Faculty of Engineering, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia; Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia.
| | - Dietmar W Hutmacher
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD 4000, Australia; Faculty of Engineering, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, QLD 4000, Australia; Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia; Australian Research Council Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Brisbane, QLD 4059, Australia.
| | - Liraz Chai
- Max Planck Queensland Centre, Queensland University of Technology, Brisbane, QLD 4000, Australia; The Hebrew University of Jerusalem, Institute of Chemistry, Jerusalem, 91904, Israel; The Harvey M. Krueger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, 9190401, Israel.
| |
Collapse
|
44
|
Brown A. The use of PICO ™ single-use negative pressure wound therapy in the community settings. Br J Community Nurs 2024; 29:S8-S26. [PMID: 39240814 DOI: 10.12968/bjcn.2024.0094] [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: 09/08/2024]
Abstract
The prevalence and incidence of wounds is predicted to rise due to an ageing population, that is also likely to have an increasing number of comorbidities (Dowsett et al, 2017). This trend will invariably result in increased costs to the NHS. The estimated annual cost of wound management in 2017/2018 was £8.3 billion. The cost of managing 70% of wounds which healed was £2.7 billion while it cost £5.6 billion managing only 30% of unhealed wounds (Guest et al, 2020). In view of these figures, it is important that health professionals (HPs) recognise wounds that are not progressing to healing at an early stage and implement all available treatment modalities to ensure that the wound does not become non-healing or stalled. Therefore, this article defines non-healing wounds, how to identify wounds at risk of becoming non-healing and the timely implementation of advanced treatment modalities, such as single use negative pressure wound therapy (sNPWT).
Collapse
|
45
|
Wan C, Ju X, Xu D, Ou J, Zhu M, Lu G, Li K, Jiang W, Li C, Hu X, Tian Y, Niu Z. Escherichia coli exopolysaccharides disrupt Pseudomonas aeruginosa biofilm and increase its antibiotic susceptibility. Acta Biomater 2024; 185:215-225. [PMID: 39067645 DOI: 10.1016/j.actbio.2024.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a major pathogen that causes infectious diseases. It has high tendency to form biofilms, resulting in the failure of traditional antibiotic therapies. Inspired by the phenomenon that co-culture of Escherichia coli (E. coli) and P. aeruginosa leads to a biofilm reduction, we reveal that E. coli exopolysaccharides (EPS) can disrupt P. aeruginosa biofilm and increase its antibiotic susceptibility. The results show that E. coli EPS effectively inhibit biofilm formation and disrupt mature biofilms in P. aeruginosa, Staphylococcus aureus, and E. coli itself. The maximal inhibition and disruption rates against P. aeruginosa biofilm are 40 % and 47 %, respectively. Based on the biofilm-disrupting ability of E. coli EPS, we develop an E. coli EPS/antibiotic combining strategy for the treatment of P. aeruginosa biofilms. The combination with E. coli EPS increases the antibacterial efficiency of tobramycin against P. aeruginosa biofilms in vitro and in vivo. This study provides a promising strategy for treating biofilm infections. STATEMENT OF SIGNIFICANCE: Biofilm formation is a leading cause of chronic infections. It blocks antibiotics, increases antibiotic-tolerance, and aids in immune evasion, thus representing a great challenge in clinic. This study proposes a promising approach to combat pathogenic Pseudomonas aeruginosa (P. aeruginosa) biofilms by combining Escherichia coli exopolysaccharides with antibiotics. This strategy shows high efficiency in different P. aeruginosa stains, including two laboratory strains, PAO1 and ATCC 10145, as well as a clinically acquired carbapenem-resistant strain. In addition, in vivo experiments have shown that this approach is effective against implanted P. aeruginosa biofilms and can prevent systemic inflammation in mice. This strategy offers new possibilities to address the clinical failure of conventional antibiotic therapies for microbial biofilms.
Collapse
Affiliation(s)
- Chenxiao Wan
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyan Ju
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Dandan Xu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinzhao Ou
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Zhu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Guojun Lu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kejia Li
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Jiang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunyan Li
- Department of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Xiaohua Hu
- Department of Burns and Plastic Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - Ye Tian
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Zhongwei Niu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
46
|
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
|
47
|
Musuroi C, Musuroi SI, Baditoiu L, Crainiceanu Z, Muntean D, Voinescu A, Izmendi O, Sirmon A, Licker M. The Profile of Bacterial Infections in a Burn Unit during and after the COVID-19 Pandemic Period. Antibiotics (Basel) 2024; 13:823. [PMID: 39334997 PMCID: PMC11428671 DOI: 10.3390/antibiotics13090823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Infections represent a major complication for burn-injured patients. The aim of this study was to highlight the changes in the incidence and antimicrobial resistance of bacterial strains isolated from burn patients, at the end of the COVID-19 pandemic, in relation to the antibiotics used during the pandemic. A comparative analysis of the demographic data and the microorganisms identified in the clinical samples of two groups of burn patients admitted to a university hospital in Romania was carried out. The first group consisted of 48 patients and the second of 69 patients, hospitalized in January-August 2020 and 2023, respectively. The bacterial species with the highest incidence were S. aureus, A. baumannii, Pseudomonas spp. The significant changes between 2023 and 2020 are reflected in the increase in the frequency of non-fermentative Gram-negative bacteria, especially S. maltophilia, and the increase in antimicrobial resistance of Pseudomonas and Klebsiella spp. Klebsiella spp. did not change in frequency (7%), but there was a significant increase in the incidence of K. pneumoniae strains with pan-drug resistant behaviour to antibiotics (40%), including colistin. The phenomenon can be explained by the selection of specimens carrying multiple resistance genes, as a result of antibiotic treatment during the COVID-19 period. The post-pandemic antimicrobial resistance detected in burn patients indicates the need for permanent surveillance of the resistance trends, primarily due to the limited therapeutic options available for these patients.
Collapse
Affiliation(s)
- Corina Musuroi
- Multidisciplinary Research Center of Antimicrobial Resistance, Microbiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Microbiology Laboratory, "Pius Brinzeu" County Clinical Emergency Hospital, 300723 Timisoara, Romania
| | - Silvia-Ioana Musuroi
- Doctoral School, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Luminita Baditoiu
- Epidemiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Zorin Crainiceanu
- Department of Plastic Surgery, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Delia Muntean
- Multidisciplinary Research Center of Antimicrobial Resistance, Microbiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Microbiology Laboratory, "Pius Brinzeu" County Clinical Emergency Hospital, 300723 Timisoara, Romania
| | - Adela Voinescu
- Multidisciplinary Research Center of Antimicrobial Resistance, Microbiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Microbiology Laboratory, "Pius Brinzeu" County Clinical Emergency Hospital, 300723 Timisoara, Romania
- Doctoral School, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Oana Izmendi
- Multidisciplinary Research Center of Antimicrobial Resistance, Microbiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Doctoral School, "Victor Babeș" University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Alexandra Sirmon
- Epidemiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Epidemiology Department, "Pius Brinzeu" County Clinical Emergency Hospital, 300723 Timisoara, Romania
| | - Monica Licker
- Multidisciplinary Research Center of Antimicrobial Resistance, Microbiology Department, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Microbiology Laboratory, "Pius Brinzeu" County Clinical Emergency Hospital, 300723 Timisoara, Romania
| |
Collapse
|
48
|
Ghanbari M, Salkovskiy Y, Carlson MA. The rat as an animal model in chronic wound research: An update. Life Sci 2024; 351:122783. [PMID: 38848945 PMCID: PMC11581782 DOI: 10.1016/j.lfs.2024.122783] [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: 01/10/2024] [Revised: 03/29/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
The increasing global prevalence of chronic wounds underscores the growing importance of developing effective animal models for their study. This review offers a critical evaluation of the strengths and limitations of rat models frequently employed in chronic wound research and proposes potential improvements. It explores these models in the context of key comorbidities, including diabetes, venous and arterial insufficiency, pressure-induced blood flow obstruction, and infections. Additionally, the review examines important wound factors including age, sex, smoking, and the impact of anesthetic and analgesic drugs, acknowledging their substantial effects on research outcomes. A thorough understanding of these variables is crucial for refining animal models and can provide valuable insights for future research endeavors.
Collapse
Affiliation(s)
- Mahboubeh Ghanbari
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA.
| | - Yury Salkovskiy
- Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA.
| | - Mark A Carlson
- Department of Surgery, Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
49
|
Uberoi A, McCready-Vangi A, Grice EA. The wound microbiota: microbial mechanisms of impaired wound healing and infection. Nat Rev Microbiol 2024; 22:507-521. [PMID: 38575708 DOI: 10.1038/s41579-024-01035-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
The skin barrier protects the human body from invasion by exogenous and pathogenic microorganisms. A breach in this barrier exposes the underlying tissue to microbial contamination, which can lead to infection, delayed healing, and further loss of tissue and organ integrity. Delayed wound healing and chronic wounds are associated with comorbidities, including diabetes, advanced age, immunosuppression and autoimmune disease. The wound microbiota can influence each stage of the multi-factorial repair process and influence the likelihood of an infection. Pathogens that commonly infect wounds, such as Staphylococcus aureus and Pseudomonas aeruginosa, express specialized virulence factors that facilitate adherence and invasion. Biofilm formation and other polymicrobial interactions contribute to host immunity evasion and resistance to antimicrobial therapies. Anaerobic organisms, fungal and viral pathogens, and emerging drug-resistant microorganisms present unique challenges for diagnosis and therapy. In this Review, we explore the current understanding of how microorganisms present in wounds impact the process of skin repair and lead to infection through their actions on the host and the other microbial wound inhabitants.
Collapse
Affiliation(s)
- Aayushi Uberoi
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amelia McCready-Vangi
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth A Grice
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Rafique A, Amjad F, Janjua MRSA, Naqvi SAR, Hassan SU, Abdullah H, Nazir MS, Ali Z, Alshihri AA, Momenah MA, Mansour AA, Bajaber MA, Alalwiat AA. Chia seed-mediated fabrication of ZnO/Ag/Ag 2O nanocomposites: structural, antioxidant, anticancer, and wound healing studies. Front Chem 2024; 12:1405385. [PMID: 39055045 PMCID: PMC11269097 DOI: 10.3389/fchem.2024.1405385] [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: 03/22/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024] Open
Abstract
Plant extract-mediated fabrication of metal nanocomposites is used in cell proliferation inhibition and topical wound treatment, demonstrating significant effectiveness. Salvia hispanica L. (chia) seed extract (CE) is used as the reaction medium for the green fabrication of ecofriendly ZnO(CE) nanoparticles (NPs) and Ag/Ag2O(CE) and ZnO/Ag/Ag2O(CE) nanocomposites. The resultant nanoparticles and nanocomposite materials were characterized using UV-visible, Fourier-transform infrared (FTIR) spectroscopy, scanning electron microscopy (SEM), X-ray diffraction (XRD), and energy-dispersive X-ray (EDX) techniques. In the context of antioxidant studies, ZnO/Ag/Ag2O(CE) exhibited 57% reducing power and 86% 2,2, diphenyl-1-picrylhydrazyl (DPPH) free radical scavenging. All three materials showed strong antibacterial activity against Staphylococcus aureus (S. aureus), Escherichia coli (E.coli), and Bacillus subtilis (B. subtilis) bacterial strains. Additionally, ZnO(CE), Ag/Ag2O(CE), and ZnO/Ag/Ag2O(CE) also revealed 64.47%, 42.56%, and 75.27% in vitro Michigan Cancer Foundation-7 (MCF7) cancer cell line inhibition, respectively, at a concentration of 100 μg/mL. Selectively, the most effective composite material, ZnO/Ag/Ag2O(CE), was used to evaluate in vivo wound healing potential in rat models. The study revealed 96% wound closure in 10 days, which was quite rapid healing compared to wound healing using clinically available ointment. Therefore, in conclusion, the ZnO/Ag/Ag2O(CE) nanocomposite material could be considered for further testing and formulation as a good anticancer and wound healing agent.
Collapse
Affiliation(s)
- Aisha Rafique
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Fatima Amjad
- Department of Chemistry, COMSATS University Islamabad, Lahore, Pakistan
| | | | - Syed Ali Raza Naqvi
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Sadaf Ul Hassan
- Department of Chemistry, COMSATS University Islamabad, Lahore, Pakistan
| | - Hanzla Abdullah
- Department of Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | | | - Zulfiqar Ali
- Department of Chemistry, COMSATS University Islamabad, Lahore, Pakistan
| | - Abdulaziz A. Alshihri
- Department of Radiological Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Maha Abdullah Momenah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Adel Abo Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Majed A. Bajaber
- Chemistry Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Ahlam A. Alalwiat
- Chemistry Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|