1
|
Yadav A, Sharma A, Moulick M, Ghatak S. Nanomanaging Chronic Wounds with Targeted Exosome Therapeutics. Pharmaceutics 2025; 17:366. [PMID: 40143030 PMCID: PMC11945274 DOI: 10.3390/pharmaceutics17030366] [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: 02/12/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic wounds pose a significant healthcare challenge, impacting millions of patients worldwide and burdening healthcare systems substantially. These wounds often occur as comorbidities and are prone to infections. Such infections hinder the healing process, complicating clinical management and proving recalcitrant to therapy. The environment within the wound itself poses challenges such as lack of oxygen, restricted blood flow, oxidative stress, ongoing inflammation, and bacterial presence. Traditional systemic treatment for such chronic peripheral wounds may not be effective due to inadequate blood supply, resulting in unintended side effects. Furthermore, topical applications are often impervious to persistent biofilm infections. A growing clinical concern is the lack of effective therapeutic modalities for treating chronic wounds. Additionally, the chemically harsh wound microenvironment can reduce the effectiveness of treatments, highlighting the need for drug delivery systems that can deliver therapies precisely where needed with optimal dosages. Compared to cell-based therapies, exosome-based therapies offer distinct advantages as a cell-free approach for chronic wound treatment. Exosomes are of endosomal origin and enable cell-to-cell communications, and they possess benefits, including biocompatibility and decreased immunogenicity, making them ideal vehicles for efficient targeting and minimizing off-target damage. However, exosomes are rapidly cleared from the body, making it difficult to maintain optimal therapeutic concentrations at wound sites. The hydrogel-based approach and development of biocompatible scaffolds for exosome-based therapies can be beneficial for sustained release and prolong the presence of these therapeutic exosomes at chronic wound sites. Engineered exosomes have been shown to possess stability and effectiveness in promoting wound healing compared to their unmodified counterparts. Significant progress has been made in this field, but further research is essential to unlock their clinical potential. This review seeks to explore the benefits and opportunities of exosome-based therapies in chronic wounds, ensuring sustained efficacy and precise delivery despite the obstacles posed by the wound environment.
Collapse
Affiliation(s)
| | | | | | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA; (A.Y.); (A.S.); (M.M.)
| |
Collapse
|
2
|
Mishra I, Chaudhary K, Sharma V, Krishna G, Mishra R. Electroceuticals: Unlocking the promise of therapies. Daru 2024; 33:7. [PMID: 39694935 PMCID: PMC11655896 DOI: 10.1007/s40199-024-00549-4] [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: 09/18/2023] [Accepted: 10/09/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVES Electroceuticals refers to the constantly growing disciplines of bioelectric and bioelectronic medication. These include a broad variety of devices that have been invented and are now being utilized in medical implants, wearable medical electronics, and bioelectronics. The primary aim of this study is to encompass several facets of electroceuticals, their applications, and recent advancements in the field of medical challenges. EVIDENCE ACQUISITIONS A complete literature study was conducted, which included a comprehensive review of globally recognized scientific research databases. RESULTS The progressive refinement and diminution of technology, in conjunction with swift advancements in comprehending the role of electrical pathways in the human body, have rendered it progressively viable to manipulate these pathways for therapeutic purposes. DISCUSSION AND CONCLUSION Electrical stimulation impacts and modifies biological functioning and pathological processes in the body. In the contemporary era of medicine, health care practitioners from a variety of fields utilize electricity to cure disease or injury or to assess and diagnose using a variety of electrically driven medical tools.
Collapse
Affiliation(s)
- Isha Mishra
- Galgotias College of Pharmacy, 201310, Greater Noida, Uttar Pradesh, India
| | - Kajal Chaudhary
- IEC Group of Institutions, Knowledge Park-I, Greater Noida, Uttar Pradesh, 201310, India
| | - Vikram Sharma
- Galgotias College of Pharmacy, 201310, Greater Noida, Uttar Pradesh, India
| | | | - Raghav Mishra
- Lloyd School of Pharmacy, 201306, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
3
|
Zhen M, Zhu Y, Wang P, Liu X, Zhu J, Liu H, Li J, Zhao J, Shu B. HMGB1 Accelerates Wound Healing by Promoting the Differentiation of Epidermal Stem Cells via the "HMGB1-TLR4-Wnt/Notch" Axis. Adv Wound Care (New Rochelle) 2024. [PMID: 39694535 DOI: 10.1089/wound.2023.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
Objective: Impairments in the differentiation and migratory capacity of epidermal stem cells (ESCs) are pivotal factors contributing to delayed wound healing. High mobility group box1 (HMGB1) has recently emerged as a potential target for tissue repair. Therefore, we aimed to investigate the role and molecular mechanisms of HMGB1 in ESCs during the wound-healing process. Approach: Initially, we examined the expression of HMGB1 and the differentiation of ESCs in normal skin, normal wounds and chronic wounds. Then, we assessed the ESC migration and differentiation, and the key markers in the Wnt/Notch signaling pathways, after treatment of HMGB1 and inhibitor, and the knockdown of toll-like receptor 4 (TLR4), using scratch assay, qPCR, western blotting, and immunofluorescence. Finally, we conducted mice models to analyze the healing rates and quality in vivo. Results: HMGB1 was decreased across all epidermal layers, and the differentiation of ESCs was hindered in diabetic foot ulcer. In vitro, HMGB1 enhanced both the migration and differentiation of ESCs while stimulating the expression of the Wnt/Notch pathway within ESCs. However, the downregulation of TLR4 negated these effects. Finally, our in vivo experiments provided evidence that HMGB1 facilitates wound healing and epidermis differentiation via TLR4 and Wnt/Notch signaling pathways. Innovation: This study innovatively introduces HMGB1 as a novel target for skin wound healing and elucidates its mechanisms of action. Conclusions: HMGB1 accelerated wound healing by promoting the differentiation of epidermal stem cells through the "HMGB1-TLR4-Wnt/Notch" axis, which reveals a new potential mechanism and target to expedite wound healing.
Collapse
Affiliation(s)
- Miao Zhen
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yongkang Zhu
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Peng Wang
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaogang Liu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Junyou Zhu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hengdeng Liu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingting Li
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingling Zhao
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Shu
- Department of Burns and Wound Repair, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Chattopadhyay D, Sinha M, Kapoor A, Kumar M, Singh K, Mathew-Steiner SS, Sen CK. Deficient functional wound closure as measured by elevated trans-epidermal water loss predicts chronic wound recurrence: An exploratory observational study. Sci Rep 2024; 14:23593. [PMID: 39384891 PMCID: PMC11464781 DOI: 10.1038/s41598-024-74426-0] [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: 06/12/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024] Open
Abstract
A single-center, prospective, observational pilot study was performed to evaluate wound healing endpoint and recurrence by measuring transepidermal water loss (TEWL) post-closure at the site of wound repair. Patients with clinically-defined chronic wounds (such as pressure ulcers, diabetic ulcers, and trauma wounds) who visited the Plastic Surgery outpatient department or were in-patients at the All India Institute of Medical Sciences, Rishikesh, India, and were referred for chronic wound management, were enrolled. Non-invasive point-of-care TEWL measurements were obtained, from closed wound-site and contralateral healthy skin site, starting from confirmation of closure (post-closure, V0) continuing every 2 weeks for a maximum of five visits or until the wound recurred. Statistical analyses of the data involved logistic regression and likelihood ratio chi-square tests to assess differences in TEWL at visit 0 (V0) between the closed wound site and reference skin, with the TEWL score as the sole predictor of recurrence. Of the 72 subjects that completed the study, 44 (61%) showed no recurrence and 28 (39%) had wounds that recurred over a period of 12 weeks. A significant association was found between the V0 (post-closure) TEWL score and the odds of wound recurrence, both in univariate analysis (OR [95%CI] = 1.26[1.14,1.42] (p < 0.001) and after adjusting for covariates in multivariable analysis (OR [95%CI] = 1.34[1.19,1.61] (p < 0.001). The likelihood ratio chi-square analysis demonstrated that the V0 TEWL score is a significant universal predictor of recurrence across all wound types studied. Cases of closed wounds with subsequent recurrence showed an overall higher post-closure V0 TEWL score, compared to those who did not have a wound recurrence, across visits. The TEWL score cut-off value predictive of recurrence was 24.1 g.m-2.h-1 (AUC = 0.967). The outcome of this pilot study on a wide range of chronic wounds leads to the hypothesis that post-closure TEWL at the site of wound healing is a reliable biomarker of wound recurrence. It also raises the question whether the clinical endpoint of wound closure should include re-establishment of skin barrier function as additional criterion. The current standard of care wound closure endpoint calls for re-epithelialization of the wound with no discharge for two consecutive weeks disregarding the functional parameter of restoration of skin barrier function at the wound-site.
Collapse
Affiliation(s)
| | - Mithun Sinha
- Department of Surgery, Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Akshay Kapoor
- All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Manoj Kumar
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Economic Studies and Planning, Jawaharlal Nehru University, New Delhi, India
| | - Kanhaiya Singh
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Chandan K Sen
- Department of Surgery, Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Piazzesi A, Scanu M, Ciprandi G, Putignani L. Modulations of the skin microbiome in skin disorders: A narrative review from a wound care perspective. Int Wound J 2024; 21:e70087. [PMID: 39379177 PMCID: PMC11461044 DOI: 10.1111/iwj.70087] [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: 06/26/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
The cutaneous microbiome represents a highly dynamic community of bacteria, fungi and viruses. Scientific evidence, particularly from the last two decades, has revealed that these organisms are far from being inconsequential microscopic hitchhikers on the human body, nor are they all opportunistic pathogens waiting for the chance to penetrate the skin barrier and cause infection. In this review, we will describe how dermatological diseases have been found to be associated with disruptions and imbalances in the skin microbiome and how this new evidence had shaped the diagnosis and clinical practice relating to these disorders. We will identify the microbial agents which have been found to directly exacerbate skin diseases, as well as those which can ameliorate many of the symptoms associated with dermatological disorders. Furthermore, we will discuss the studies which suggest that bacteriotherapy, either by topical use of probiotics or by bacteria-derived compounds, can rectify skin microbial imbalances, thereby offering a promising alternative to antibiotic treatment and reducing the risks of antibiotic resistance.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Guido Ciprandi
- Research Institute Division of Plastic and Maxillofacial Surgery, Department of SurgeryBambino Gesu' Children's Hospital, IRCCSRomeItaly
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics; and Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| |
Collapse
|
6
|
Dinić M, Burgess JL, Lukić J, Catanuto P, Radojević D, Marjanović J, Verpile R, Thaller SR, Gonzalez T, Golić N, Strahinić I, Tomic-Canic M, Pastar I. Postbiotic lactobacilli induce cutaneous antimicrobial response and restore the barrier to inhibit the intracellular invasion of Staphylococcus aureus in vitro and ex vivo. FASEB J 2024; 38:e23801. [PMID: 39018106 PMCID: PMC11258854 DOI: 10.1096/fj.202400054rr] [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: 01/08/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Intracellular pathogens including Staphylococcus aureus contribute to the non-healing phenotype of chronic wounds. Lactobacilli, well known as beneficial bacteria, are also reported to modulate the immune system, yet their role in cutaneous immunity remains largely unknown. We explored the therapeutic potential of bacteria-free postbiotics, bioactive lysates of lactobacilli, to reduce intracellular S. aureus colonization and promote healing. Fourteen postbiotics derived from various lactobacilli species were screened, and Latilactobacillus curvatus BGMK2-41 was selected for further analysis based on the most efficient ability to reduce intracellular infection by S. aureus diabetic foot ulcer clinical isolate and S. aureus USA300. Treatment of both infected keratinocytes in vitro and infected human skin ex vivo with BGMK2-41 postbiotic cleared S. aureus. Keratinocytes treated in vitro with BGMK2-41 upregulated expression of antimicrobial response genes, of which DEFB4, ANG, and RNASE7 were also found upregulated in treated ex vivo human skin together with CAMP exclusively upregulated ex vivo. Furthermore, BGMK2-41 postbiotic treatment has a multifaceted impact on the wound healing process. Treatment of keratinocytes stimulated cell migration and the expression of tight junction proteins, while in ex vivo human skin BGMK2-41 increased expression of anti-inflammatory cytokine IL-10, promoted re-epithelialization, and restored the epidermal barrier via upregulation of tight junction proteins. Together, this provides a potential therapeutic approach for persistent intracellular S. aureus infections.
Collapse
Affiliation(s)
- Miroslav Dinić
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jamie L. Burgess
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami FL, USA
| | - Jovanka Lukić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Paola Catanuto
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dušan Radojević
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Jelena Marjanović
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Rebecca Verpile
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Seth R. Thaller
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tammy Gonzalez
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nataša Golić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Ivana Strahinić
- Group for Probiotics and Microbiota-Host Interaction, Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Molecular and Cellular Pharmacology Graduate Program, University of Miami Miller School of Medicine, Miami FL, USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
7
|
Fuchs C, Stalnaker KJ, Dalgard CL, Sukumar G, Hupalo D, Dreyfuss JM, Pan H, Wang Y, Pham L, Wu X, Jozic I, Anderson RR, Cho S, Meyerle JH, Tam J. Plantar Skin Exhibits Altered Physiology, Constitutive Activation of Wound-Associated Phenotypes, and Inherently Delayed Healing. J Invest Dermatol 2024; 144:1633-1648.e14. [PMID: 38237729 DOI: 10.1016/j.jid.2023.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 06/24/2024]
Abstract
Wound research has typically been performed without regard for where the wounds are located on the body, despite well-known heterogeneities in physical and biological properties between different skin areas. The skin covering the palms and soles is highly specialized, and plantar ulcers are one of the most challenging and costly wound types to manage. Using primarily the porcine model, we show that plantar skin is molecularly and functionally more distinct from nonplantar skin than previously recognized, with unique gene and protein expression profiles, broad alterations in cellular functions, constitutive activation of many wound-associated phenotypes, and inherently delayed healing. This unusual physiology is likely to play a significant but underappreciated role in the pathogenesis of plantar ulcers as well as the last 25+ years of futility in therapy development efforts. By revealing this critical yet unrecognized pitfall, we hope to contribute to the development of more effective therapies for these devastating nonhealing wounds.
Collapse
Affiliation(s)
- Christiane Fuchs
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine J Stalnaker
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Clifton L Dalgard
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Gauthaman Sukumar
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Daniel Hupalo
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ying Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Linh Pham
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xunwei Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivan Jozic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillp Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - R Rox Anderson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Sunghun Cho
- Department of Dermatology, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA; Department of Dermatology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Jon H Meyerle
- Department of Dermatology, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA; Department of Dermatology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Joshua Tam
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
8
|
Pignet AL, Schellnegger M, Hecker A, Kamolz LP, Kotzbeck P. Modeling Wound Chronicity In Vivo: The Translational Challenge to Capture the Complexity of Chronic Wounds. J Invest Dermatol 2024; 144:1454-1470. [PMID: 38483357 DOI: 10.1016/j.jid.2023.11.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 11/14/2023] [Accepted: 11/19/2023] [Indexed: 06/24/2024]
Abstract
In an aging society with common lifestyle-associated health issues such as obesity and diabetes, chronic wounds pose a frequent challenge that physicians face in everyday clinical practice. Therefore, nonhealing wounds have attracted much scientific attention. Several in vitro and in vivo models have been introduced to deepen our understanding of chronic wound pathogenesis and amplify therapeutic strategies. Understanding how wounds become chronic will provide insights to reverse or avoid chronicity. Although choosing a suitable model is of utmost importance to receive valuable outcomes, an ideal in vivo model capturing the complexity of chronic wounds is still missing and remains a translational challenge. This review discusses the most relevant mammalian models for wound healing studies and provides guidance on how to implement the hallmarks of chronic wounds. It highlights the benefits and pitfalls of established models and maps out future avenues for research.
Collapse
Affiliation(s)
- Anna-Lisa Pignet
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria; COREMED - Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH, Graz, Austria; Research Unit for Tissue Repair and Reconstruction, Medical University of Graz, Graz, Austria
| | - Marlies Schellnegger
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria; COREMED - Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH, Graz, Austria; Research Unit for Tissue Repair and Reconstruction, Medical University of Graz, Graz, Austria.
| | - Andrzej Hecker
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria; COREMED - Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH, Graz, Austria; Research Unit for Tissue Repair and Reconstruction, Medical University of Graz, Graz, Austria
| | - Lars-Peter Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria; COREMED - Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH, Graz, Austria
| | - Petra Kotzbeck
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria; COREMED - Centre for Regenerative and Precision Medicine, JOANNEUM RESEARCH, Graz, Austria; Research Unit for Tissue Repair and Reconstruction, Medical University of Graz, Graz, Austria
| |
Collapse
|
9
|
Lysitsas M, Triantafillou E, Chatzipanagiotidou I, Antoniou K, Spyrou V, Billinis C, Valiakos G. Phenotypic Investigation and Detection of Biofilm-Associated Genes in Acinetobacter baumannii Isolates, Obtained from Companion Animals. Trop Med Infect Dis 2024; 9:109. [PMID: 38787042 PMCID: PMC11125616 DOI: 10.3390/tropicalmed9050109] [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: 03/31/2024] [Revised: 05/01/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Bacteria of the genus Acinetobacter, especially Acinetobacter baumannii (Ab), have emerged as pathogens of companion animals during the last two decades and are commonly associated with hospitalization and multidrug resistance. A critical factor for the distribution of relevant strains in healthcare facilities, including veterinary facilities, is their adherence to both biotic and abiotic surfaces and the production of biofilms. A group of 41 A. baumannii isolates obtained from canine and feline clinical samples in Greece was subjected to phenotypic investigation of their ability to produce biofilms using the tissue culture plate (TCP) method. All of them (100%) produced biofilms, while 23 isolates (56.1%) were classified as strong producers, 11 (26.8%) as moderate producers, and 7 (17.1%) as weak producers. A correlation between the MDR and XDR phenotypes and weak or moderate biofilm production was identified. Moreover, the presence of four biofilm-associated genes bap, blaPER, ompA, and csuE was examined by PCR, and they were detected in 100%, 65.9%, 97.6%, and 95.1% of the strains respectively. All isolates carried at least two of the investigated genes, whereas most of the strong biofilm producers carried all four genes. In conclusion, the spread and persistence of biofilm-producing Ab strains in veterinary facilities is a matter of concern, since they are regularly obtained from infected animals, indicating their potential as challenging pathogens for veterinarians due to multidrug resistance and tolerance in conventional eradication measures. Furthermore, considering that companion animals can act as reservoirs of relevant strains, public health concerns emerge.
Collapse
Affiliation(s)
- Marios Lysitsas
- Faculty of Veterinary Science, University of Thessaly, 43100 Karditsa, Greece; (M.L.); (C.B.)
| | | | | | - Konstantina Antoniou
- Vet Analyseis, Private Diagnostic Laboratory, 41335 Larissa, Greece; (E.T.); (K.A.)
| | - Vassiliki Spyrou
- Department of Animal Science, University of Thessaly, 41334 Larissa, Greece;
| | - Charalambos Billinis
- Faculty of Veterinary Science, University of Thessaly, 43100 Karditsa, Greece; (M.L.); (C.B.)
| | - George Valiakos
- Faculty of Veterinary Science, University of Thessaly, 43100 Karditsa, Greece; (M.L.); (C.B.)
| |
Collapse
|
10
|
Seo S, Yang Q, Jeong S, Della Porta A, Kapoor H, Gibson DJ. A surfactant-based dressing can reduce the appearance of Pseudomonas aeruginosa pigments and uncover the dermal extracellular matrix in an ex vivo porcine skin wound model. Int Wound J 2024; 21:e14510. [PMID: 38148595 PMCID: PMC10958096 DOI: 10.1111/iwj.14510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 12/28/2023] Open
Abstract
From previous studies, we have shown that viable colony forming units of bacteria and bacterial biofilms are reduced after sequential treatment with a surfactant-based dressing. Here, we sought to test the impact on visible bacterial pigments and the ultrastructural impact following the sequential treatment of the same surfactant-based dressing. Mature Pseudomonas aeruginosa biofilms were grown on ex vivo porcine skin explants, and an imaging-based analysis was used to compare the skin with and without a concentrated surfactant. In explants naturally tinted by bacterial chromophores, wiping alone had no effect, while the use of a surfactant-based dressing reduced coloration. Similarly, daily wiping led to increased immunohistochemical staining for P. aeruginosa antigens, but not in the surfactant group. Confocal immunofluorescent imaging revealed limited bacterial penetration and coating of the dermis and loose pieces of sloughing material. Ultrastructural analysis confirmed that the biofilms were masking the extracellular matrix (ECM), but the surfactant could remove them, re-exposing the ECM. The masking of the ECM may provide another non-inflammatory explanation for delayed healing, as the ECM is no longer accessible for wound cell locomotion. The use of a poloxamer-based surfactant appears to be an effective way to remove bacterial chromophores and the biofilm coating the ECM fibres.
Collapse
Affiliation(s)
- Soojung Seo
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
- Department of Biological SciencesUniversity of AlabamaTuscaloosaAlabamaUSA
| | - Qingping Yang
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Sunyoung Jeong
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Alessandra Della Porta
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Harris Kapoor
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
| | - Daniel J. Gibson
- Department of OB/GYNInstitute for Wound Research, University of FloridaGainesvilleFloridaUSA
- Capstone College of NursingUniversity of AlabamaTuscaloosaAlabamaUSA
| |
Collapse
|
11
|
Saeed S, Martins-Green M. Assessing Animal Models to Study Impaired and Chronic Wounds. Int J Mol Sci 2024; 25:3837. [PMID: 38612647 PMCID: PMC11011258 DOI: 10.3390/ijms25073837] [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: 02/20/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Impaired healing wounds do not proceed through the normal healing processes in a timely and orderly manner, and while they do eventually heal, their healing is not optimal. Chronic wounds, on the other hand, remain unhealed for weeks or months. In the US alone, chronic wounds impact ~8.5 million people and cost ~USD 28-90 billion per year, not accounting for the psychological and physical pain and emotional suffering that patients endure. These numbers are only expected to rise in the future as the elderly populations and the incidence of comorbidities such as diabetes, hypertension, and obesity increase. Over the last few decades, scientists have used a variety of approaches to treat chronic wounds, but unfortunately, to date, there is no effective treatment. Indeed, while there are thousands of drugs to combat cancer, there is only one single drug approved for the treatment of chronic wounds. This is in part because wound healing is a very complex process involving many phases that must occur sequentially and in a timely manner. Furthermore, models that fully mimic human chronic wounds have not been developed. In this review, we assess various models currently being used to study the biology of impaired healing and chronic non-healing wounds. Among them, this paper also highlights one model which shows significant promise; this model uses aged and obese db/db-/- mice and the chronic wounds that develop show characteristics of human chronic wounds that include increased oxidative stress, chronic inflammation, damaged microvasculature, abnormal collagen matrix deposition, a lack of re-epithelialization, and the spontaneous development of multi-bacterial biofilm. We also discuss how important it is that we continue to develop chronic wound models that more closely mimic those of humans and that can be used to test potential treatments to heal chronic wounds.
Collapse
Affiliation(s)
| | - Manuela Martins-Green
- Department of Molecular, Cell, and Systems Biology, University of California, Riverside, CA 92521, USA;
| |
Collapse
|
12
|
Yang Y, Huang J, Zeng A, Long X, Yu N, Wang X. The role of the skin microbiome in wound healing. BURNS & TRAUMA 2024; 12:tkad059. [PMID: 38444635 PMCID: PMC10914219 DOI: 10.1093/burnst/tkad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/05/2023] [Accepted: 11/21/2023] [Indexed: 03/07/2024]
Abstract
The efficient management of skin wounds for rapid and scarless healing represents a major clinical unmet need. Nonhealing skin wounds and undesired scar formation impair quality of life and result in high healthcare expenditure worldwide. The skin-colonizing microbiota contributes to maintaining an intact skin barrier in homeostasis, but it also participates in the pathogenesis of many skin disorders, including aberrant wound healing, in many respects. This review focuses on the composition of the skin microbiome in cutaneous wounds of different types (i.e. acute and chronic) and with different outcomes (i.e. nonhealing and hypertrophic scarring), mainly based on next-generation sequencing analyses; furthermore, we discuss the mechanistic insights into host-microbe and microbe-microbe interactions during wound healing. Finally, we highlight potential therapeutic strategies that target the skin microbiome to improve healing outcomes.
Collapse
Affiliation(s)
- Yuyan Yang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Jiuzuo Huang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Ang Zeng
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Nanze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Xiaojun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| |
Collapse
|
13
|
Kumar M, Banerjee P, Das A, Singh K, Guith T, Kacar S, Gourishetti K, Sen CK, Roy S, Khanna S. Hydrolyzed Collagen Powder Dressing Improves Wound Inflammation, Perfusion, and Breaking Strength of Repaired Tissue. Adv Wound Care (New Rochelle) 2024; 13:70-82. [PMID: 37534840 DOI: 10.1089/wound.2023.0065] [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: 08/04/2023] Open
Abstract
Objective: Hydrolyzed collagen-based matrices are widely used as wound care dressings. Information on the mechanism of action of such dressings is scanty. The objective of this study was to test the effect of a specific hydrolyzed collagen powder (HCP), which is extensively used for wound care management in the United States. Approach: The effects of HCP on resolution of wound inflammation, perfusion, closure, and breaking strength of the repaired skin were studied in an experimental murine model. Results: In early (day 7) inflammatory phase of wound macrophages, HCP treatment boosted phagocytosis and efferocytosis of wound-site macrophages. In these cells, inducible reactive oxygen species were also higher on day (d) 7. HCP treatment potentiated the expression of anti-inflammatory interleukin (IL)-10 cytokine and proangiogenic vascular endothelial growth factor (VEGF) production. Excisional wounds dressed with HCP showed complete closure on day 21, while the control wounds remained open. HCP treatment also demonstrated improved quality of wound healing as marked by the improved breaking strength of the closed wound tissue/repaired skin. Innovation: These data represent first evidence on the mechanism of action of clinically used HCP. Conclusion: HCP dressing favorably influenced both wound inflammation and vascularization. Improved breaking strength of HCP-treated repaired skin lays the rationale for future studies testing the hypothesis that HCP-treated closed wounds would show fewer recurrences.
Collapse
Affiliation(s)
- Manishekhar Kumar
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Pradipta Banerjee
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Amitava Das
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kanhaiya Singh
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tanner Guith
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sedat Kacar
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Karthik Gourishetti
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chandan K Sen
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sashwati Roy
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Savita Khanna
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
14
|
Bhattacharya M, Horswill AR. The role of human extracellular matrix proteins in defining Staphylococcus aureus biofilm infections. FEMS Microbiol Rev 2024; 48:fuae002. [PMID: 38337187 PMCID: PMC10873506 DOI: 10.1093/femsre/fuae002] [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: 07/19/2023] [Revised: 01/26/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024] Open
Abstract
Twenty to forty one percent of the world's population is either transiently or permanently colonized by the Gram-positive bacterium, Staphylococcus aureus. In 2017, the CDC designated methicillin-resistant S. aureus (MRSA) as a serious threat, reporting ∼300 000 cases of MRSA-associated hospitalizations annually, resulting in over 19 000 deaths, surpassing that of HIV in the USA. S. aureus is a proficient biofilm-forming organism that rapidly acquires resistance to antibiotics, most commonly methicillin (MRSA). This review focuses on a large group of (>30) S. aureus adhesins, either surface-associated or secreted that are designed to specifically bind to 15 or more of the proteins that form key components of the human extracellular matrix (hECM). Importantly, this includes hECM proteins that are pivotal to the homeostasis of almost every tissue environment [collagen (skin), proteoglycans (lung), hemoglobin (blood), elastin, laminin, fibrinogen, fibronectin, and fibrin (multiple organs)]. These adhesins offer S. aureus the potential to establish an infection in every sterile tissue niche. These infections often endure repeated immune onslaught, developing into chronic, biofilm-associated conditions that are tolerant to ∼1000 times the clinically prescribed dose of antibiotics. Depending on the infection and the immune response, this allows S. aureus to seamlessly transition from colonizer to pathogen by subtly manipulating the host against itself while providing the time and stealth that it requires to establish and persist as a biofilm. This is a comprehensive discussion of the interaction between S. aureus biofilms and the hECM. We provide particular focus on the role of these interactions in pathogenesis and, consequently, the clinical implications for the prevention and treatment of S. aureus biofilm infections.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, CO 80045, United States
| |
Collapse
|
15
|
Liu Y, McQuillen EA, Rana PSJB, Gloag ES, Parsek MR, Wozniak DJ. A bacterial pigment provides cross-species protection from H 2O 2- and neutrophil-mediated killing. Proc Natl Acad Sci U S A 2024; 121:e2312334121. [PMID: 38170744 PMCID: PMC10786307 DOI: 10.1073/pnas.2312334121] [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: 07/19/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024] Open
Abstract
Bacterial infections are often polymicrobial. Pseudomonas aeruginosa and Staphylococcus aureus cause chronic co-infections, which are more problematic than mono-species infections. Understanding the mechanisms of their interactions is crucial for treating co-infections. Staphyloxanthin (STX), a yellow pigment synthesized by the S. aureus crt operon, promotes S. aureus resistance to oxidative stress and neutrophil-mediated killing. We found that STX production by S. aureus, either as surface-grown macrocolonies or planktonic cultures, was elevated when exposed to the P. aeruginosa exoproduct, 2-heptyl-4-hydroxyquinoline N-oxide (HQNO). This was observed with both mucoid and non-mucoid P. aeruginosa strains. The induction phenotype was found in a majority of P. aeruginosa and S. aureus clinical isolates examined. When subjected to hydrogen peroxide or human neutrophils, P. aeruginosa survival was significantly higher when mixed with wild-type (WT) S. aureus, compared to P. aeruginosa alone or with an S. aureus crt mutant deficient in STX production. In a murine wound model, co-infection with WT S. aureus, but not the STX-deficient mutant, enhanced P. aeruginosa burden and disease compared to mono-infection. In conclusion, we identified a role for P. aeruginosa HQNO mediating polymicrobial interactions with S. aureus by inducing STX production, which consequently promotes resistance to the innate immune effectors H2O2 and neutrophils. These results further our understanding of how different bacterial species cooperatively cause co-infections.
Collapse
Affiliation(s)
- Yiwei Liu
- Department of Microbiology, Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, Ohio State University College of Medicine, Columbus, OH43210
| | - Eleanor A. McQuillen
- Department of Health and Rehabilitation Sciences, Ohio State University College of Medicine, Columbus, OH43210
| | - Pranav S. J. B. Rana
- Department of Microbiology, Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, Ohio State University College of Medicine, Columbus, OH43210
| | - Erin S. Gloag
- Department of Microbial Infection and Immunity, Ohio State University College of Medicine, Columbus, OH43210
- Department of Biomedical Sciences and Pathobiology, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA24060
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington School of Medicine, Seattle, WA98195
| | - Daniel J. Wozniak
- Department of Microbiology, Ohio State University, Columbus, OH43210
- Department of Microbial Infection and Immunity, Ohio State University College of Medicine, Columbus, OH43210
| |
Collapse
|
16
|
Chan RK, Nuutila K, Mathew-Steiner SS, Diaz V, Anselmo K, Batchinsky M, Carlsson A, Ghosh N, Sen CK, Roy S. A Prospective, Randomized, Controlled Study to Evaluate the Effectiveness of a Fabric-Based Wireless Electroceutical Dressing Compared to Standard-of-Care Treatment Against Acute Trauma and Burn Wound Biofilm Infection. Adv Wound Care (New Rochelle) 2024; 13:1-13. [PMID: 36855334 PMCID: PMC10654645 DOI: 10.1089/wound.2023.0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Objective: Despite advances in the use of topical and parenteral antimicrobial therapy and the practice of early tangential burn wound excision to manage bacterial load, 60% of the mortality from burns is attributed to bacterial biofilm infection. A low electric field (∼1 V) generated by the novel FDA-cleared wireless electroceutical dressing (WED) was previously shown to significantly prevent and disrupt burn biofilm infection in preclinical studies. Based on this observation, the purpose of this clinical trial was to evaluate the efficacy of the WED dressing powered by a silver-zinc electrocouple in the prevention and disruption of biofilm infection. Approach: A prospective, randomized, controlled, single-center clinical trial was performed to evaluate the efficacy of the WED compared with standard-of-care (SoC) dressing to treat biofilms. Burn wounds were randomized to receive either SoC or WED. Biopsies were collected on days 0 and 7 for histology, scanning electron microscopy (SEM) examination of biofilm, and for quantitative bacteriological analyses. Results: In total, 38 subjects were enrolled in the study. In 52% of the WED-treated wounds, little to no biofilm could be detected by SEM. WED significantly lowered or prevented increase of biofilm in all wounds compared with the pair-matched SoC-treated wounds. Innovation: WED is a simple, easy, and rapid method to protect the wound while also inhibiting infection. It is activated by a moist environment and the electrical field induces transient and micromolar amounts of superoxide anion radicals that will prevent bacterial growth. Conclusion: WED decreased biofilm infection better compared with SoC. The study was registered in clinicaltrials.gov as NCT04079998.
Collapse
Affiliation(s)
- Rodney K. Chan
- United States Army Institute of Surgical Research, Ft. Sam Houston, Texas, USA
| | - Kristo Nuutila
- United States Army Institute of Surgical Research, Ft. Sam Houston, Texas, USA
| | | | | | | | - Maria Batchinsky
- United States Army Institute of Surgical Research, Ft. Sam Houston, Texas, USA
| | - Anders Carlsson
- United States Army Institute of Surgical Research, Ft. Sam Houston, Texas, USA
- Metis Foundation, San Antonio, Texas, USA
| | - Nandini Ghosh
- Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chandan K. Sen
- Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sashwati Roy
- Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
17
|
Khaledi M, Zandi B, Mohsenipour Z. The Effect of Mesenchymal Stem Cells on the Wound Infection. Curr Stem Cell Res Ther 2024; 19:1084-1092. [PMID: 37815189 DOI: 10.2174/011574888x252482230926104342] [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: 03/08/2023] [Revised: 07/10/2023] [Accepted: 08/17/2023] [Indexed: 10/11/2023]
Abstract
Wound infection often requires a long period of care and an onerous treatment process. Also, the rich environment makes the wound an ideal niche for microbial growth. Stable structures, like biofilm, and drug-resistant strains cause a delay in the healing process, which has become one of the important challenges in wound treatment. Many studies have focused on alternative methods to deal the wound infections. One of the novel and highly potential ways is mesenchymal stromal cells (MSCs). MSCs are mesoderm-derived pluripotent adult stem cells with the capacity for self-renewal, multidirectional differentiation, and immunological control. Also, MSCs have anti-inflammatory and antiapoptotic effects. MScs, as pluripotent stromal cells, differentiate into many mature cells. Also, MSCs produce antimicrobial compounds, such as antimicrobial peptides (AMP), as well as secrete immune modulators, which are two basic features considered in wound healing. Despite the advantages, preserving the structure and activity of MSCs is considered one of the most important points in the treatment. MSCs' antimicrobial effects on microorganisms involved in wound infection have been confirmed in various studies. In this review, we aimed to discuss the antimicrobial and therapeutic applications of MSCs in the infected wound healing processes.
Collapse
Affiliation(s)
- Mansoor Khaledi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Bita Zandi
- Department of Microbiology, Faculty of advanced science and technology, Tehran medical science, Islamic Azad University, Tehran, Iran
| | - Zeinab Mohsenipour
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Sen CK. Human Wound and Its Burden: Updated 2022 Compendium of Estimates. Adv Wound Care (New Rochelle) 2023; 12:657-670. [PMID: 37756368 PMCID: PMC10615092 DOI: 10.1089/wound.2023.0150] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 09/24/2023] [Indexed: 09/29/2023] Open
Abstract
Significance: Chronic wounds affect 10.5 million (up 2.3 million from the 2014 update) of U.S. Medicare beneficiaries. Chronic wounds impact the quality of life of nearly 2.5% of the total population of the United States. This fraction is larger in the elderly. These wounds can lead to a range of complications and health care costs. Given the aging population, the continued threat of diabetes and obesity worldwide, and the persistent problem of infection, it is expected that chronic wounds will continue to be a substantial clinical, social, and economic challenge. Disparities in the prevalence and management of chronic wounds exist, with underserved communities and marginalized populations often facing greater challenges in accessing quality wound care. These disparities exacerbate the public health burden. Recent Advances: U.S. Centers for Medicare and Medicaid Services had proposed revision of its local coverage determination limiting the use of skin substitute grafts/cellular and/or tissue-based products for the treatment of diabetic foot ulcers and venous leg ulcers in the U.S. Medicare population. In response to the comment phase, this proposal has been put on hold. The U.S. Food and Drug Administration (FDA) has renewed its focus on addressing nonhealing chronic wounds and has outlined efforts to address identified barriers to product development for nonhealing chronic wounds. The new approach places emphasis on engaging key wound healing stakeholders, including academia, professional associations, patient groups, reimbursement organizations, and industry. Finally, recent advances demonstrating that wounds closed by current FDA definition of wound closure may remain functionally open because of deficiencies in restoration of barrier function warrant revisiting the wound closure endpoint. Such "closed" wounds that are functionally open, also known as invisible wounds, are likely to be associated with high wound recurrence. Future Directions: Addressing the public health problem of chronic wounds will require a multifaceted approach that includes prevention, improved wound care management, and addressing the underlying risk factors.
Collapse
Affiliation(s)
- Chandan K. Sen
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh Medical Center Health System Wound Care Service, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
19
|
Khan I, Minto RE, Kelley-Patteson C, Singh K, Timsina L, Suh LJ, Rinne E, Van Natta BW, Neumann CR, Mohan G, Lester M, VonDerHaar RJ, German R, Marino N, Hassanein AH, Gordillo GM, Kaplan MH, Sen CK, Kadin ME, Sinha M. Biofilm-derived oxylipin 10-HOME-mediated immune response in women with breast implants. J Clin Invest 2023; 134:e165644. [PMID: 38032740 PMCID: PMC10849761 DOI: 10.1172/jci165644] [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: 09/27/2022] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
This study investigates a mechanistic link of bacterial biofilm-mediated host-pathogen interaction leading to immunological complications associated with breast implant illness (BII). Over 10 million women worldwide have breast implants. In recent years, women have described a constellation of immunological symptoms believed to be related to their breast implants. We report that periprosthetic breast tissue of participants with symptoms associated with BII had increased abundance of biofilm and biofilm-derived oxylipin 10-HOME compared with participants with implants who are without symptoms (non-BII) and participants without implants. S. epidermidis biofilm was observed to be higher in the BII group compared with the non-BII group and the normal tissue group. Oxylipin 10-HOME was found to be immunogenically capable of polarizing naive CD4+ T cells with a resulting Th1 subtype in vitro and in vivo. Consistently, an abundance of CD4+Th1 subtype was observed in the periprosthetic breast tissue and blood of people in the BII group. Mice injected with 10-HOME also had increased Th1 subtype in their blood, akin to patients with BII, and demonstrated fatigue-like symptoms. The identification of an oxylipin-mediated mechanism of immune activation induced by local bacterial biofilm provides insight into the possible pathogenesis of the implant-associated immune symptoms of BII.
Collapse
Affiliation(s)
- Imran Khan
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert E. Minto
- Department of Chemistry and Chemical Biology, Indiana University–Purdue University Indianapolis, Indianapolis, Indiana, USA
| | | | - Kanhaiya Singh
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lava Timsina
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lily J. Suh
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ethan Rinne
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Colby R. Neumann
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ganesh Mohan
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mary Lester
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - R. Jason VonDerHaar
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rana German
- Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Department of Medicine, and
| | - Natascia Marino
- Susan G. Komen Tissue Bank at the IU Simon Comprehensive Cancer Center, Department of Medicine, and
- Division of Hematology & Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Aladdin H. Hassanein
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Gayle M. Gordillo
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- McGowan Institute for Regenerative Medicine, Department of Plastic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mark H. Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Chandan K. Sen
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Marshall E. Kadin
- Department of Dermatology, Roger Williams Medical Center, Boston University School of Medicine, Providence, Rhode Island, USA
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Mithun Sinha
- Division of Plastic Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
20
|
Campbell AE, McCready-Vangi AR, Uberoi A, Murga-Garrido SM, Lovins VM, White EK, Pan JTC, Knight SAB, Morgenstern AR, Bianco C, Planet PJ, Gardner SE, Grice EA. Variable staphyloxanthin production by Staphylococcus aureus drives strain-dependent effects on diabetic wound-healing outcomes. Cell Rep 2023; 42:113281. [PMID: 37858460 PMCID: PMC10680119 DOI: 10.1016/j.celrep.2023.113281] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 08/24/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023] Open
Abstract
Strain-level variation in Staphylococcus aureus is a factor that contributes to disease burden and clinical outcomes in skin disorders and chronic wounds. However, the microbial mechanisms that drive these variable host responses are poorly understood. To identify mechanisms underlying strain-specific outcomes, we perform high-throughput phenotyping screens on S. aureus isolates cultured from diabetic foot ulcers. Isolates from non-healing wounds produce more staphyloxanthin, a cell membrane pigment. In murine diabetic wounds, staphyloxanthin-producing isolates delay wound closure significantly compared with staphyloxanthin-deficient isolates. Staphyloxanthin promotes resistance to oxidative stress and enhances bacterial survival in neutrophils. Comparative genomic and transcriptomic analysis of genetically similar clinical isolates with disparate staphyloxanthin phenotypes reveals a mutation in the sigma B operon, resulting in marked differences in stress response gene expression. Our work illustrates a framework to identify traits that underlie strain-level variation in disease burden and suggests more precise targets for therapeutic intervention in S. aureus-positive wounds.
Collapse
Affiliation(s)
- Amy E Campbell
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amelia R McCready-Vangi
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aayushi Uberoi
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sofía M Murga-Garrido
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Victoria M Lovins
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ellen K White
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jamie Ting-Chun Pan
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon A B Knight
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexis R Morgenstern
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Colleen Bianco
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul J Planet
- Division of Infectious Disease, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Departments of Pediatrics and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sue E Gardner
- College of Nursing, University of Iowa, Iowa City, IA 52242, USA
| | - Elizabeth A Grice
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
21
|
Huang CX, Siwan E, Fox SL, Longfield M, Twigg SM, Min D. Comparison of digital and traditional skin wound closure assessment methods in mice. Lab Anim Res 2023; 39:25. [PMID: 37891640 PMCID: PMC10605778 DOI: 10.1186/s42826-023-00176-1] [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: 07/12/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Chronic skin wounds are a common complication of many diseases such as diabetes. Various traditional methods for assessing skin wound closure are used in animal studies, including wound tracing, calliper measurements and histological analysis. However, these methods have poorly defined wound closure or practical limitations. Digital image analysis of wounds is an increasingly popular, accessible alternative, but it is unclear whether digital assessment is consistent with traditional methods. This study aimed to optimise and compare digital wound closure assessment with traditional methods, using a diabetic mouse model. Diabetes was induced in male C57BL/6J mice by high-fat diet feeding combined with low dose (65 mg/kg of body weight) streptozotocin injections. Mice fed normal chow were included as controls. After 18 weeks, four circular full-thickness dorsal skin wounds of 4 mm diameter were created per mouse. The wounds were photographed and measured by callipers. Wound closure rate (WCR) was digitally assessed by two reporters using two methods: wound outline (WCR-O) and re-epithelialisation (WCR-E). Wounded skin tissues were collected at 10-days post-wounding and wound width was measured from haematoxylin and eosin-stained skin tissue. RESULTS Between reporters, WCR-O was more consistent than WCR-E, and WCR-O correlated with calliper measurements. Histological analysis supported digital assessments, especially WCR-E, when wounds were histologically closed. CONCLUSIONS WCR-O could replace calliper measurements to measure skin wound closure, but WCR-E assessment requires further refinement. Small animal studies of skin wound healing can greatly benefit from standardised definitions of wound closure and more consistent digital assessment protocols.
Collapse
Affiliation(s)
- Coco X Huang
- Greg Brown Diabetes and Endocrine Research Laboratory, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Elisha Siwan
- Greg Brown Diabetes and Endocrine Research Laboratory, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Sarah L Fox
- Greg Brown Diabetes and Endocrine Research Laboratory, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Matilda Longfield
- Greg Brown Diabetes and Endocrine Research Laboratory, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Stephen M Twigg
- Greg Brown Diabetes and Endocrine Research Laboratory, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Danqing Min
- Greg Brown Diabetes and Endocrine Research Laboratory, Sydney Medical School (Central), Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
22
|
Ghatak S, Hemann C, Boslett J, Singh K, Sharma A, El Masry MS, Abouhashem AS, Ghosh N, Mathew-Steiner SS, Roy S, Zweier JL, Sen CK. Bacterial Pyocyanin Inducible Keratin 6A Accelerates Closure of Epithelial Defect under Conditions of Mitochondrial Dysfunction. J Invest Dermatol 2023; 143:2052-2064.e5. [PMID: 37044260 PMCID: PMC10529774 DOI: 10.1016/j.jid.2023.03.1671] [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: 09/10/2021] [Revised: 03/13/2023] [Accepted: 03/19/2023] [Indexed: 04/14/2023]
Abstract
Repair of epithelial defect is complicated by infection and related metabolites. Pyocyanin (PYO) is one such metabolite that is secreted during Pseudomonas aeruginosa infection. Keratinocyte (KC) migration is required for the closure of skin epithelial defects. This work sought to understand PYO-KC interaction and its significance in tissue repair. Stable Isotope Labeling by Amino acids in Cell culture proteomics identified mitochondrial dysfunction as the top pathway responsive to PYO exposure in human KCs. Consistently, functional studies showed mitochondrial stress, depletion of reducing equivalents, and adenosine triphosphate. Strikingly, despite all stated earlier, PYO markedly accelerated KC migration. Investigation of underlying mechanisms revealed, to our knowledge, a previously unreported function of keratin 6A in KCs. Keratin 6A was PYO inducible and accelerated closure of epithelial defect. Acceleration of closure was associated with poor quality healing, including compromised expression of apical junction proteins. This work recognizes keratin 6A for its role in enhancing KC migration under conditions of threat posed by PYO. Qualitatively deficient junctional proteins under conditions of defensive acceleration of KC migration explain why an infected wound close with deficient skin barrier function as previously reported.
Collapse
Affiliation(s)
- Subhadip Ghatak
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Craig Hemann
- Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - James Boslett
- Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA; Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Anu Sharma
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Mohamed S El Masry
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Ahmed Safwat Abouhashem
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Nandini Ghosh
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Shomita S Mathew-Steiner
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Jay L Zweier
- Division of Cardiovascular Medicine, Department of Internal Medicine, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, School of Medicine, Indiana University, Indianapolis, Indiana, USA.
| |
Collapse
|
23
|
Kletzer J, Raval YS, Mohamed A, Mandrekar JN, Greenwood-Quaintance KE, Beyenal H, Patel R. In vitro activity of hypochlorous acid generating electrochemical bandage against monospecies and dual-species bacterial biofilms. J Appl Microbiol 2023; 134:lxad194. [PMID: 37667489 PMCID: PMC10508963 DOI: 10.1093/jambio/lxad194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/18/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023]
Abstract
AIMS As antimicrobial resistance is on the rise, treating chronic wound infections is becoming more complex. The presence of biofilms in wound beds contributes to this challenge. Here, the activity of a novel hypochlorous acid (HOCl) producing electrochemical bandage (e-bandage) against monospecies and dual-species bacterial biofilms formed by bacteria commonly found in wound infections was assessed. METHODS AND RESULTS The system was controlled by a wearable potentiostat powered by a 3V lithium-ion battery and maintaining a constant voltage of + 1.5V Ag/AgCl, allowing continuous generation of HOCl. A total of 19 monospecies and 10 dual-species bacterial biofilms grown on polycarbonate membranes placed on tryptic soy agar (TSA) plates were used as wound biofilm models, with HOCl producing e-bandages placed over the biofilms. Viable cell counts were quantified after e-bandages were continuously polarized for 2, 4, 6, and 12 hours. Time-dependent reductions in colony forming units (CFUs) were observed for all studied isolates. After 12 hours, average CFU reductions of 7.75 ± 1.37 and 7.74 ± 0.60 log10 CFU/cm2 were observed for monospecies and dual-species biofilms, respectively. CONCLUSIONS HOCl producing e-bandages reduce viable cell counts of in vitro monospecies and dual-species bacterial biofilms in a time-dependent manner in vitro. After 12 hours, >99.999% reduction in cell viability was observed for both monospecies and dual-species biofilms.
Collapse
Affiliation(s)
- Joseph Kletzer
- Paracelsus Medical University, Salzburg 5020, Austria
- Division of Clinical Microbiology, Mayo Clinic Rochester, Rochester, MN 55905, United States
| | - Yash S Raval
- Division of Clinical Microbiology, Mayo Clinic Rochester, Rochester, MN 55905, United States
| | - Abdelrhman Mohamed
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164, United States
| | - Jayawant N Mandrekar
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN 55905, United States
| | | | - Haluk Beyenal
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164, United States
| | - Robin Patel
- Division of Clinical Microbiology, Mayo Clinic Rochester, Rochester, MN 55905, United States
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
24
|
Wang C, Li C, Li X, Cai L, Han Z, Du R. RETRACTED ARTICLE: Burn Wounds: Proliferating Site for Biofilm Infection. Appl Biochem Biotechnol 2023; 195:5478. [PMID: 35604533 DOI: 10.1007/s12010-022-03964-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Chao Wang
- Second Department of Burn and Plastic Surgery, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Chungeng Li
- Department of proctology, Tangshan Hospital of Traditional Chinese Medicine, 063000, Tangshan, Hebei, China
| | - Xiaoying Li
- Department of Internal Emergency, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Lanfang Cai
- Second Department of Burn and Plastic Surgery, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Zhenning Han
- Second Department of Burn and Plastic Surgery, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China
| | - Rui Du
- Department one of Cardiology, Tangshan Worker's Hospital, 063000, Tangshan, Hebei, China.
| |
Collapse
|
25
|
El Masry M, Bhasme P, Mathew-Steiner SS, Smith J, Smeenge T, Roy S, Sen CK. Swine Model of Biofilm Infection and Invisible Wounds. J Vis Exp 2023:10.3791/65301. [PMID: 37395583 PMCID: PMC10655070 DOI: 10.3791/65301] [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] [Indexed: 07/04/2023] Open
Abstract
Biofilm infection is a major contributor to wound chronicity. The establishment of clinically relevant experimental wound biofilm infection requires the involvement of the host immune system. Iterative changes in the host and pathogen during the formation of such clinically relevant biofilm can only occur in vivo. The swine wound model is recognized for its advantages as a powerful pre-clinical model. There are several reported approaches for studying wound biofilms. In vitro and ex vivo systems are deficient in terms of the host immune response. Short-term in vivo studies involve acute responses and, thus, do not allow for biofilm maturation, as is known to occur clinically. The first long-term swine wound biofilm study was reported in 2014. The study recognized that biofilm-infected wounds may close as determined by planimetry, but the skin barrier function of the affected site may fail to be restored. Later, this observation was validated clinically. The concept of functional wound closure was thus born. Wounds closed but deficient in skin barrier function may be viewed as invisible wounds. In this work, we seek to report the methodological details necessary to reproduce the long-term swine model of biofilm-infected severe burn injury, which is clinically relevant and has translational value. This protocol provides detailed guidance on establishing an 8 week wound biofilm infection using P. aeruginosa (PA01). Eight full-thickness burn wounds were created symmetrically on the dorsum of domestic white pigs, which were inoculated with (PA01) at day 3 post-burn; subsequently, noninvasive assessments of the wound healing were conducted at different time points using laser speckle imaging (LSI), high-resolution ultrasound (HUSD), and transepidermal water loss (TEWL). The inoculated burn wounds were covered with a four-layer dressing. Biofilms, as established and confirmed structurally by SEM at day 7 post-inoculation, compromised the functional wound closure. Such an adverse outcome is subject to reversal in response to appropriate interventions.
Collapse
Affiliation(s)
- Mohamed El Masry
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine
| | - Pramod Bhasme
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine
| | - Shomita S Mathew-Steiner
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine
| | - Jessica Smith
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine
| | - Thomas Smeenge
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine;
| |
Collapse
|
26
|
Abstract
Significance: Healthy skin provides a barrier to contaminants. Breaches in skin integrity are often encountered in the patient health care journey, owing to intrinsic health issues or to various procedures and medical devices used. The time has come to move clinical practice beyond mere awareness of medical adhesive-related skin injury and toward improved care and outcomes. Recent Advances: Methods developed in research settings allow quantitative assessments of skin damage based on the measurement of baseline skin properties. These properties become altered by stress and over time. Assessment methods typically used by the cosmetic industry to compare product performance could offer new possibilities to improve clinical practice by providing better information on the status of patient skin. This review summarizes available skin assessment methods as well as specific patient risks for skin damage. Critical Issues: Patients in health care settings may be at risk for skin damage owing to predisposing medical conditions, health status, medications taken, and procedures or devices used in their treatment. Skin injuries come as an additional burden to these medical circumstances and could be prevented. Technology should be leveraged to improve care, help maintain patient skin health, and better characterize functional wound closure. Future Directions: Skin testing methods developed to evaluate cosmetic products or assess damage caused by occupational exposure can provide detailed, quantitative information on the integrity of skin. Such methods have the potential to guide prevention and treatment efforts to improve the care of patients suffering from skin integrity issues while in the health care system.
Collapse
Affiliation(s)
- Stéphanie F. Bernatchez
- 3M Health Care, St. Paul, Minnesota, USA.,Correspondence: 3M Health Care, St. Paul, MN 55144-1000, USA
| | | |
Collapse
|
27
|
Abstract
OBJECTIVE This work addressing complexities in wound infection, seeks to test the reliance of bacterial pathogen Pseudomonas aeruginosa (PA) on host skin lipids to form biofilm with pathological consequences. BACKGROUND PA biofilm causes wound chronicity. Both CDC as well as NIH recognizes biofilm infection as a threat leading to wound chronicity. Chronic wounds on lower extremities often lead to surgical limb amputation. METHODS An established preclinical porcine chronic wound biofilm model, infected with PA or Pseudomonas aeruginosa ceramidase mutant (PA ∆Cer ), was used. RESULTS We observed that bacteria drew resource from host lipids to induce PA ceramidase expression by three orders of magnitude. PA utilized product of host ceramide catabolism to augment transcription of PA ceramidase. Biofilm formation was more robust in PA compared to PA ∆Cer . Downstream products of such metabolism such as sphingosine and sphingosine-1-phosphate were both directly implicated in the induction of ceramidase and inhibition of peroxisome proliferator-activated receptor (PPAR)δ, respectively. PA biofilm, in a ceram-idastin-sensitive manner, also silenced PPARδ via induction of miR-106b. Low PPARδ limited ABCA12 expression resulting in disruption of skin lipid homeostasis. Barrier function of the wound-site was thus compromised. CONCLUSIONS This work demonstrates that microbial pathogens must co-opt host skin lipids to unleash biofilm pathogenicity. Anti-biofilm strategies must not necessarily always target the microbe and targeting host lipids at risk of infection could be productive. This work may be viewed as a first step, laying fundamental mechanistic groundwork, toward a paradigm change in biofilm management.
Collapse
|
28
|
Zhao F, Su Y, Wang J, Romanova S, DiMaio DJ, Xie J, Zhao S. A Highly Efficacious Electrical Biofilm Treatment System for Combating Chronic Wound Bacterial Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208069. [PMID: 36385439 PMCID: PMC9918715 DOI: 10.1002/adma.202208069] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/24/2022] [Indexed: 05/26/2023]
Abstract
Biofilm infection has a high prevalence in chronic wounds and can delay wound healing. Current treatment using debridement and antibiotic administration imposes a significant burden on patients and healthcare systems. To address their limitations, a highly efficacious electrical antibiofilm treatment system is described in this paper. This system uses high-intensity current (75 mA cm-2 ) to completely debride biofilm above the wound surface and enhance antibiotic delivery into biofilm-infected wounds simultaneously. Combining these two effects, this system uses short treatments (≤2 h) to reduce bacterial count of methicillin-resistant S. aureus (MRSA) biofilm-infected ex vivo skin wounds from 1010 to 105.2 colony-forming units (CFU) g-1 . Taking advantage of the hydrogel ionic circuit design, this system enhances the in vivo safety of high-intensity current application compared to conventional devices. The in vivo antibiofilm efficacy of the system is tested using a diabetic mouse-based wound infection model. MRSA biofilm bacterial count decreases from 109.0 to 104.6 CFU g-1 at 1 day post-treatment and to 103.3 CFU g-1 at 7 days post-treatment, both of which are below the clinical threshold for infection. Overall, this novel technology provides a quick, safe, yet highly efficacious treatment to chronic wound biofilm infections.
Collapse
Affiliation(s)
- Fan Zhao
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yajuan Su
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Junying Wang
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Svetlana Romanova
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Dominick J DiMaio
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jingwei Xie
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Siwei Zhao
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
29
|
Sen CK, Roy S, Khanna S. Diabetic Peripheral Neuropathy Associated with Foot Ulcer: One of a Kind. Antioxid Redox Signal 2023. [PMID: 35850520 DOI: 10.1089/ars.2022.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Significance: Diabetic peripheral neuropathy (DPN) associated with a diabetic foot ulcer (DFU) is likely to be complicated with critical factors such as biofilm infection and compromised skin barrier function of the diabetic skin. Repaired skin with a history of biofilm infection is known to be compromised in barrier function. Loss of barrier function is also observed in the oxidative stress affected diabetic and aged skin. Recent Advances: Loss of barrier function makes the skin prone to biofilm infection and cellulitis, which contributes to chronic inflammation and vasculopathy. Hyperglycemia favors biofilm formation as glucose lowering led to reduction in biofilm development. While vasculopathy limits oxygen supply, the O2 cost of inflammation is high increasing hypoxia severity. Critical Issues: The host nervous system can be inhabited by bacteria. Because electrical impulses are a part of microbial physiology, polymicrobial colonization of the host's neural circuit is likely to influence transmission of action potential. The identification of perineural apatite in diabetic patients with peripheral neuropathy suggests bacterial involvement. DPN starts in both feet at the same time. Future Directions: Pair-matched studies of DPN in the foot affected with DFU (i.e., DFU-DPN) compared with DPN in the without ulcer, and intact skin barrier function, are likely to provide critical insight that would help inform effective care strategies. This review characterizes DFU-DPN from a translational science point of view presenting a new paradigm that recognizes the current literature in the context of factors that are unique to DFU-DPN.
Collapse
Affiliation(s)
- Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Savita Khanna
- Indiana Center for Regenerative Medicine & Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
30
|
Liu Y, McQuillen EA, Rana PSJB, Gloag ES, Wozniak DJ. Cross-Species Protection to Innate Immunity Mediated by A Bacterial Pigment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.15.524085. [PMID: 36711503 PMCID: PMC9882196 DOI: 10.1101/2023.01.15.524085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Bacterial infections are often polymicrobial. Pseudomonas aeruginosa and Staphylococcus aureus cause chronic co-infections, which are more problematic than mono-species infections. We found that the production of S. aureus membrane-bound pigment staphyloxanthin (STX), was induced by the P. aeruginosa exoproduct, 2-heptyl-4-hydroxyquinoline N-oxide (HQNO). The induction phenotype was conserved in P. aeruginosa and S. aureus clinical isolates examined. When subjected to hydrogen peroxide or human neutrophils, P. aeruginosa survival was significantly higher when mixed with wild-type (WT) S. aureus , compared to a mutant deficient in STX production or P. aeruginosa alone. In a murine wound model, co-infection with WT S. aureus , but not the STX-deficient mutant, enhanced P. aeruginosa burden and disease compared to mono-infection. In conclusion, we discovered a novel role for P. aeruginosa HQNO mediating polymicrobial interactions with S. aureus by inducing STX production, which consequently promotes resistance of both pathogens to innate immune effectors. These results further our understanding of how different bacterial species cooperatively cause co-infections.
Collapse
|
31
|
Jones TL, Holmes CM, Katona A, Martin CL, Niewczas MA, Pop-Busui R, Schmidt BM, Sen CK, Tomic-Canic M, Veves A. The NIDDK Diabetic Foot Consortium. J Diabetes Sci Technol 2023; 17:7-14. [PMID: 36059271 PMCID: PMC9846389 DOI: 10.1177/19322968221121152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Diabetic Foot Consortium (DFC) was established in September 2018 by the NIDDK to build an organization to facilitate the highest quality of clinical research on diabetic foot ulcers (DFUs) that will answer clinically significant questions to improve DFU healing and prevent amputations. The initial focus of the DFC is to develop and validate biomarkers for DFUs that can be used in clinical care and research. The DFC consists of a data coordinating center (DCC) for operational oversight and statistical analysis, clinical sites for participant recruitment and evaluation, and biomarker analysis units (BAUs). The DFC is currently studying biomarkers to predict wound healing and recurrence and is collecting biosamples for future studies through a biorepository. The DFC plans to address the challenges of recruitment and eligibility criteria for DFU clinical trials by taking an approach of "No DFU Patient Goes Unstudied." In this platform approach, clinical history, DFU outcome, wound imaging, and biologic measurements from a large number of patients will be captured and the in-depth longitudinal data set will be analyzed to develop a computational-based DFU risk factor profile to facilitate scientifically sound clinical trial design. The DFC will expand its platform to include studies of the role of social determinants of health, such as food insecurity, housing instability, limited health literacy, and poor social support. The DFC is starting partnerships with the broad group of stakeholders in the wound care community.
Collapse
Affiliation(s)
- Teresa L.Z. Jones
- National Institute of Diabetes
and Digestive and Kidney Diseases (NIDDK), Bethesda, MD, USA
| | | | - Aimee Katona
- University of Michigan Medical
School, Ann Arbor, MI, USA
| | | | - Monika A. Niewczas
- Section on Genetics and
Epidemiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA,
USA
| | | | | | - Chandan K. Sen
- Indiana University School of
Medicine and Indiana University Health Comprehensive Wound Center,
Indianapolis, IN, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative
Medicine Research Program, University of Miami Miller School of Medicine,
Miami, FL, USA
| | - Aristidis Veves
- The Rongxiang Xu, MD, Center for
Regenerative Therapeutics, Joslin-Beth Israel Deaconess Foot Center, Beth
Israel Deaconess Medical Center, Harvard Medical School, Boston, MA,
USA
| |
Collapse
|
32
|
Manero A, Crawford KE, Prock‐Gibbs H, Shah N, Gandhi D, Coathup MJ. Improving disease prevention, diagnosis, and treatment using novel bionic technologies. Bioeng Transl Med 2023; 8:e10359. [PMID: 36684104 PMCID: PMC9842045 DOI: 10.1002/btm2.10359] [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: 02/23/2022] [Revised: 05/09/2022] [Accepted: 05/30/2022] [Indexed: 01/25/2023] Open
Abstract
Increased human life expectancy, due in part to improvements in infant and childhood survival, more active lifestyles, in combination with higher patient expectations for better health outcomes, is leading to an extensive change in the number, type and manner in which health conditions are treated. Over the next decades as the global population rapidly progresses toward a super-aging society, meeting the long-term quality of care needs is forecast to present a major healthcare challenge. The goal is to ensure longer periods of good health, a sustained sense of well-being, with extended periods of activity, social engagement, and productivity. To accomplish these goals, multifunctionalized interfaces are an indispensable component of next generation medical technologies. The development of more sophisticated materials and devices as well as an improved understanding of human disease is forecast to revolutionize the diagnosis and treatment of conditions ranging from osteoarthritis to Alzheimer's disease and will impact disease prevention. This review examines emerging cutting-edge bionic materials, devices and technologies developed to advance disease prevention, and medical care and treatment in our elderly population including developments in smart bandages, cochlear implants, and the increasing role of artificial intelligence and nanorobotics in medicine.
Collapse
Affiliation(s)
- Albert Manero
- Limbitless SolutionsUniversity of Central FloridaOrlandoFloridaUSA
- Biionix ClusterUniversity of Central FloridaOrlandoFloridaUSA
| | - Kaitlyn E. Crawford
- Biionix ClusterUniversity of Central FloridaOrlandoFloridaUSA
- Department of Materials Science and EngineeringUniversity of Central FloridaOrlandoFloridaUSA
| | | | - Neel Shah
- College of MedicineUniversity of Central FloridaOrlandoFloridaUSA
| | - Deep Gandhi
- College of MedicineUniversity of Central FloridaOrlandoFloridaUSA
| | | |
Collapse
|
33
|
Dreifus JE, O’Neal L, Jacobs HM, Subramanian AS, Howell PL, Wozniak DJ, Parsek MR. The Sia System and c-di-GMP Play a Crucial Role in Controlling Cell-Association of Psl in Planktonic P. aeruginosa. J Bacteriol 2022; 204:e0033522. [PMID: 36448788 PMCID: PMC9794950 DOI: 10.1128/jb.00335-22] [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: 09/07/2022] [Accepted: 11/01/2022] [Indexed: 12/02/2022] Open
Abstract
Many bacterial species use the secondary messenger, c-di-GMP, to promote the production of biofilm matrix components. In Pseudomonas aeruginosa, c-di-GMP production is stimulated upon initial surface contact and generally remains high throughout biofilm growth. Transcription of several gene clusters, including the Sia signal transduction system, are induced in response to high cellular levels of c-di-GMP. The output of this system is SiaD, a diguanylate cyclase whose activity is induced in the presence of the detergent SDS. Previous studies demonstrated that Sia-mediated cellular aggregation is a key feature of P. aeruginosa growth in the presence of SDS. Here, we show that the Sia system is important for producing low levels of c-di-GMP when P. aeruginosa is growing planktonically. In addition, we show that Sia activity is important for maintaining cell-associated Psl in planktonic populations. We also demonstrate that Sia mutant strains have reduced cell-associated Psl and a surface attachment-deficient phenotype. The Sia system also appears to posttranslationally impact cell-associated Psl levels. Collectively, our findings suggest a novel role for the Sia system and c-di-GMP in planktonic populations by regulating levels of cell-associated Psl.
Collapse
Affiliation(s)
- Julia E. Dreifus
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Lindsey O’Neal
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Holly M. Jacobs
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Adithya S. Subramanian
- Program in Molecular Medicine, Research Institute the Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - P. Lynne Howell
- Program in Molecular Medicine, Research Institute the Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Daniel J. Wozniak
- Department of Microbial Infections and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
34
|
Fig latex inhibits the growth of pathogenic bacteria invading human diabetic wounds and accelerates wound closure in diabetic mice. Sci Rep 2022; 12:21852. [PMID: 36528674 PMCID: PMC9759588 DOI: 10.1038/s41598-022-26338-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Impaired wound healing is one of the most critical complications associated with diabetes mellitus. Infections and foot ulcers are major causes of morbidity for diabetic patients. The current treatment of diabetic foot ulcers, commonly used antibiotics, is associated with the development of bacterial resistance. Hence, novel and more effective natural therapeutic antibacterial agents are urgently needed and should be developed against the pathogenic bacteria inhabiting diabetic wounds. Therefore, the current study aimed to investigate the impact of fig latex on pathogenic bacteria and its ability to promote the healing process of diabetic wounds. The pathogenic bacteria were isolated from patients with diabetic foot ulcers admitted to Assiut University Hospital. Fig latex was collected from trees in the Assiut region, and its chemical composition was analyzed using GC‒MS. The antibacterial efficacy of fig latex was assessed on the isolated bacteria. An in vivo study to investigate the effect of fig latex on diabetic wound healing was performed using three mouse groups: nondiabetic control mice, diabetic mice and diabetic mice treated with fig latex. The influence of fig latex on the expression levels of β-defensin-1, PECAM-1, CCL2 and ZO-1 and collagen formation was investigated. The GC‒MS analysis demonstrated the presence of triterpenoids, comprising more than 90% of the total latex content. Furthermore, using a streptozotocin-induced diabetic mouse model, topical treatment of diabetic wound tissues with fig latex was shown to accelerate and improve wound closure by increasing the expression levels of β-defensin-1, collagen, and PECAM-1 compared to untreated diabetic wounds. Additionally, fig latex decreased the expression levels of ZO-1 and CCL2.
Collapse
|
35
|
Development of an implantable three-dimensional model of a functional pathogenic multispecies biofilm to study infected wounds. Sci Rep 2022; 12:21846. [PMID: 36528648 PMCID: PMC9759537 DOI: 10.1038/s41598-022-25569-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Chronic wounds cannot heal due to impairment of regeneration, mainly caused by the persistent infection of multispecies biofilms. Still, the effects of biofilm wound infection and its interaction with the host are not fully described. We aimed to study functional biofilms in physiological conditions in vitro, and their potential effects in health and regeneration in vivo. Therefore, Pseudomonas aeruginosa, Staphylococcus aureus and Enterococcus faecalis were seeded in collagen-based scaffolds for dermal regeneration. After 24 h, scaffolds had bacterial loads depending on the initial inoculum, containing viable biofilms with antibiotic tolerance. Afterwards, scaffolds were implanted onto full skin wounds in mice, together with daily supervision and antibiotic treatment. Although all mice survived their health was affected, displaying fever and weight loss. After ten days, histomorphology of scaffolds showed high heterogeneity in samples and within groups. Wounds were strongly, mildly, or not infected according to colony forming units, and P. aeruginosa had higher identification frequency. Biofilm infection induced leucocyte infiltration and elevated interferon-γ and interleukin-10 in scaffolds, increase of size and weight of spleen and high systemic pro-calcitonin concentrations. This functional and implantable 3D biofilm model allows to study host response during infection, providing a useful tool for infected wounds therapy development.
Collapse
|
36
|
Anju VT, Busi S, Imchen M, Kumavath R, Mohan MS, Salim SA, Subhaswaraj P, Dyavaiah M. Polymicrobial Infections and Biofilms: Clinical Significance and Eradication Strategies. Antibiotics (Basel) 2022; 11:antibiotics11121731. [PMID: 36551388 PMCID: PMC9774821 DOI: 10.3390/antibiotics11121731] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Biofilms are population of cells growing in a coordinated manner and exhibiting resistance towards hostile environments. The infections associated with biofilms are difficult to control owing to the chronicity of infections and the emergence of antibiotic resistance. Most microbial infections are contributed by polymicrobial or mixed species interactions, such as those observed in chronic wound infections, otitis media, dental caries, and cystic fibrosis. This review focuses on the polymicrobial interactions among bacterial-bacterial, bacterial-fungal, and fungal-fungal aggregations based on in vitro and in vivo models and different therapeutic interventions available for polymicrobial biofilms. Deciphering the mechanisms of polymicrobial interactions and microbial diversity in chronic infections is very helpful in anti-microbial research. Together, we have discussed the role of metagenomic approaches in studying polymicrobial biofilms. The outstanding progress made in polymicrobial research, especially the model systems and application of metagenomics for detecting, preventing, and controlling infections, are reviewed.
Collapse
Affiliation(s)
- V T Anju
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
- Correspondence:
| | - Madangchanok Imchen
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kerala 671316, India
- Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Mahima S. Mohan
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Simi Asma Salim
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Pattnaik Subhaswaraj
- Department of Biotechnology and Bioinformatics, Sambalpur University, Burla, Sambalpur 768019, India
| | - Madhu Dyavaiah
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| |
Collapse
|
37
|
Cheng Y, De Bank PA, Bolhuis A. An in vitro and ex vivo wound infection model to test topical and systemic treatment with antibiotics. J Appl Microbiol 2022; 133:2993-3006. [PMID: 35916629 PMCID: PMC9804477 DOI: 10.1111/jam.15756] [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: 04/29/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 01/05/2023]
Abstract
AIMS This study aimed to develop a wound infection model that could be used to test antibiotic-loaded electrospun matrices for the topical treatment of infected skin and compare the effectiveness of this treatment to systemically applied antibiotics. METHODS AND RESULTS 3D-printed flow chambers were made in which Staphylococcus aureus biofilms were grown either on a polycarbonate membrane or explanted porcine skin. The biofilms were then treated either topically, by placing antibiotic-loaded electrospun matrices on top of the biofilms, or systemically by the addition of antibiotics in the growth medium that flowed underneath the membrane or skin. The medium that was used was either a rich medium or an artificial wound fluid. The results showed that microbial viability in the biofilms was reduced to a greater extent with the topical electrospun matrices when compared to systemic treatment. CONCLUSIONS An ex vivo infection model was developed that is flexible and can be used to test both topical and systemic treatment of wound infections. It represents a significant improvement over previous in vitro models that we have used to test electrospun membranes. SIGNIFICANCE AND IMPACT OF THE STUDY The availability of a relatively simple wound infection model in which different delivery methods and dosage regimes can be tested is beneficial for the development of improved treatments for wound infections.
Collapse
Affiliation(s)
- Yanyan Cheng
- Department of Pharmacy and Pharmacology and the Centre for Therapeutic InnovationUniversity of BathBathUK
| | - Paul A. De Bank
- Department of Pharmacy and Pharmacology and the Centre for Therapeutic InnovationUniversity of BathBathUK
| | - Albert Bolhuis
- Department of Pharmacy and Pharmacology and the Centre for Therapeutic InnovationUniversity of BathBathUK
| |
Collapse
|
38
|
Rizzetto G, Molinelli E, Radi G, Cirioni O, Brescini L, Giacometti A, Offidani A, Simonetti O. MRSA and Skin Infections in Psoriatic Patients: Therapeutic Options and New Perspectives. Antibiotics (Basel) 2022; 11:1504. [PMID: 36358159 PMCID: PMC9686594 DOI: 10.3390/antibiotics11111504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 09/29/2023] Open
Abstract
Psoriatic patients present various infectious risk factors, but there are few studies in the literature evaluating the actual impact of psoriasis in severe staphylococcal skin infections. Our narrative review of the literature suggests that psoriatic patients are at increased risk of both colonization and severe infection, during hospitalization, by S. aureus. The latter also appears to play a role in the pathogenesis of psoriasis through the production of exotoxins. Hospitalized psoriatic patients are also at increased risk of MRSA skin infections. For this reason, new molecules are needed that could both overcome bacterial resistance and inhibit exotoxin production. In our opinion, in the near future, topical quorum sensing inhibitors in combination with current anti-MRSA therapies will be able to overcome the increasing resistance and block exotoxin production. Supplementation with Vitamin E (VE) or derivatives could also enhance the effect of anti-MRSA antibiotics, considering that psoriatic patients with metabolic comorbidities show a low intake of VE and low serum levels, making VE supplementation an interesting new perspective.
Collapse
Affiliation(s)
- Giulio Rizzetto
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Elisa Molinelli
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Giulia Radi
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oscar Cirioni
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Lucia Brescini
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Andrea Giacometti
- Clinic of Infectious Diseases, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Annamaria Offidani
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Oriana Simonetti
- Clinic of Dermatology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| |
Collapse
|
39
|
Powell LC, Cullen JK, Boyle GM, De Ridder T, Yap PY, Xue W, Pierce CJ, Pritchard MF, Menzies GE, Abdulkarim M, Adams JYM, Stokniene J, Francis LW, Gumbleton M, Johns J, Hill KE, Jones AV, Parsons PG, Reddell P, Thomas DW. Topical, immunomodulatory epoxy-tiglianes induce biofilm disruption and healing in acute and chronic skin wounds. Sci Transl Med 2022; 14:eabn3758. [DOI: 10.1126/scitranslmed.abn3758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The management of antibiotic-resistant, bacterial biofilm infections in chronic skin wounds is an increasing clinical challenge. Despite advances in diagnosis, many patients do not derive benefit from current anti-infective/antibiotic therapies. Here, we report a novel class of naturally occurring and semisynthetic epoxy-tiglianes, derived from the Queensland blushwood tree (
Fontainea picrosperma)
, and demonstrate their antimicrobial activity (modifying bacterial growth and inducing biofilm disruption), with structure/activity relationships established against important human pathogens. In vitro, the lead candidate EBC-1013 stimulated protein kinase C (PKC)–dependent neutrophil reactive oxygen species (ROS) induction and NETosis and increased expression of wound healing–associated cytokines, chemokines, and antimicrobial peptides in keratinocytes and fibroblasts. In vivo, topical EBC-1013 induced rapid resolution of infection with increased matrix remodeling in acute thermal injuries in calves. In chronically infected diabetic mouse wounds, treatment induced cytokine/chemokine production, inflammatory cell recruitment, and complete healing (in six of seven wounds) with ordered keratinocyte differentiation. These results highlight a nonantibiotic approach involving contrasting, orthogonal mechanisms of action combining targeted biofilm disruption and innate immune induction in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Lydia C. Powell
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
- Centre for Nanohealth, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Jason K. Cullen
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Glen M. Boyle
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Tom De Ridder
- QBiotics Group Limited Yungaburra, Queensland 4884, Australia
| | - Pei-Yi Yap
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Wenya Xue
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Carly J. Pierce
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | | | - Muthanna Abdulkarim
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Joana Stokniene
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Lewis W. Francis
- Centre for Nanohealth, Swansea University Medical School, Swansea University, Swansea SA2 8PP, UK
| | - Mark Gumbleton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff CF10 3NB, UK
| | - Jenny Johns
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Katja E. Hill
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| | - Adam V. Jones
- Oral Pathology, Cardiff and Vale University Health Board , Cardiff CF14 4XY, UK
| | - Peter G. Parsons
- Drug Discovery Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
| | - Paul Reddell
- QBiotics Group Limited Yungaburra, Queensland 4884, Australia
| | - David W. Thomas
- Advanced Therapies Group, Cardiff University School of Dentistry, Cardiff CF14 4XY, UK
| |
Collapse
|
40
|
Wong JJ, Ho FK, Choo PY, Chong KKL, Ho CMB, Neelakandan R, Keogh D, Barkham T, Chen J, Liu CF, Kline KA. Escherichia coli BarA-UvrY regulates the pks island and kills Staphylococci via the genotoxin colibactin during interspecies competition. PLoS Pathog 2022; 18:e1010766. [PMID: 36067266 PMCID: PMC9481169 DOI: 10.1371/journal.ppat.1010766] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/16/2022] [Accepted: 07/25/2022] [Indexed: 11/19/2022] Open
Abstract
Wound infections are often polymicrobial in nature, biofilm associated and therefore tolerant to antibiotic therapy, and associated with delayed healing. Escherichia coli and Staphylococcus aureus are among the most frequently cultured pathogens from wound infections. However, little is known about the frequency or consequence of E. coli and S. aureus polymicrobial interactions during wound infections. Here we show that E. coli kills Staphylococci, including S. aureus, both in vitro and in a mouse excisional wound model via the genotoxin, colibactin. Colibactin biosynthesis is encoded by the pks locus, which we identified in nearly 30% of human E. coli wound infection isolates. While it is not clear how colibactin is released from E. coli or how it penetrates target cells, we found that the colibactin intermediate N-myristoyl-D-Asn (NMDA) disrupts the S. aureus membrane. We also show that the BarA-UvrY two component system (TCS) senses the environment created during E. coli and S. aureus mixed species interaction, leading to upregulation of pks island genes. Further, we show that BarA-UvrY acts via the carbon storage global regulatory (Csr) system to control pks expression. Together, our data demonstrate the role of colibactin in interspecies competition and show that it is regulated by BarA-UvrY TCS during interspecies competition. Wound infections are often polymicrobial in nature and are associated with poor disease prognoses. Escherichia coli and Staphylococcus aureus are among the top five most cultured pathogens from wound infections. However, little is known about the polymicrobial interactions between E. coli and S. aureus during wound infections. In this study, we show that E. coli kills S. aureus both in vitro and in a mouse excisional wound model via the genotoxin, colibactin. We also show that the BarA-UvrY two component system (TCS) regulates the pks island during this mixed species interaction, acting through the carbon storage global regulatory (Csr) system to control colibactin production. Together, our data demonstrate the role of colibactin in interspecies competition and show that it is regulated by BarA-UvrY TCS during interspecies competition.
Collapse
Affiliation(s)
- Jun Jie Wong
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- Singapore Centre for Environmental Life Sciences Engineering, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore, Singapore
| | - Foo Kiong Ho
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Pei Yi Choo
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Kelvin K. L. Chong
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- Nanyang Technological University Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, Singapore, Singapore
| | - Chee Meng Benjamin Ho
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Ramesh Neelakandan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Damien Keogh
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Timothy Barkham
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Laboratory Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - John Chen
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chuan Fa Liu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Kimberly A. Kline
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
41
|
Microbial Interplay in Skin and Chronic Wounds. CURRENT CLINICAL MICROBIOLOGY REPORTS 2022. [DOI: 10.1007/s40588-022-00180-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract
Purpose of Review
Microbial infections in chronic wounds can often lead to lower-limb amputation, decrease in quality of life, and increase in mortality rate, and there is an unmet need to distinguish between pathogens and colonisers in these chronic wounds. Hence, identifying the composition of healthy skin microbiota, microbes associated with chronic wound and healing processes, and microbial interactions and host response in healing wounds vs. non-healing wounds can help us in formulating innovative individual-centric treatment protocols.
Recent Findings
This review highlights various metabolites and biomarkers produced by microbes that have been identified to modulate these interactions, particularly those involved in host–microbe and microbe–microbe communication. Further, considering that many skin commensals demonstrate contextual pathogenicity, we provide insights into promising initiatives in the wound microbiome research.
Summary
The skin microbiome is highly diverse and variable, and considering its importance remains to be a hotspot of medical investigations and research to enable us to prevent and treat skin disorders and chronic wound infections. This is especially relevant now considering that non-healing and chronic wounds are highly prevalent, generally affecting lower extremities as seen in diabetic foot ulcers, venous leg ulcers, and pressure ulcers. Pathogenic bacteria are purported to have a key role in deferring healing of wounds. However, the role of skin microflora in wound progression has been a subject of debate. In this review, we discuss biomarkers associated with chronic wound microenvironment along with the relevance of skin microflora and their metabolites in determining the chronicity of wounds.
Collapse
|
42
|
Singh K, Rustagi Y, Abouhashem AS, Tabasum S, Verma P, Hernandez E, Pal D, Khona DK, Mohanty SK, Kumar M, Srivastava R, Guda PR, Verma SS, Mahajan S, Killian JA, Walker LA, Ghatak S, Mathew-Steiner SS, Wanczyk K, Liu S, Wan J, Yan P, Bundschuh R, Khanna S, Gordillo GM, Murphy MP, Roy S, Sen CK. Genome-wide DNA hypermethylation opposes healing in chronic wound patients by impairing epithelial-to-mesenchymal transition. J Clin Invest 2022; 132:157279. [PMID: 35819852 PMCID: PMC9433101 DOI: 10.1172/jci157279] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 07/07/2022] [Indexed: 12/15/2022] Open
Abstract
An extreme chronic wound tissue microenvironment causes epigenetic gene silencing. An unbiased whole-genome methylome was studied in the wound-edge tissue of patients with chronic wounds. A total of 4,689 differentially methylated regions (DMRs) were identified in chronic wound-edge skin compared with unwounded human skin. Hypermethylation was more frequently observed (3,661 DMRs) in the chronic wound-edge tissue compared with hypomethylation (1,028 DMRs). Twenty-six hypermethylated DMRs were involved in epithelial-mesenchymal transition (EMT). Bisulfite sequencing validated hypermethylation of a predicted specific upstream regulator TP53. RNA-Seq analysis was performed to qualify findings from methylome analysis. Analysis of the downregulated genes identified the TP53 signaling pathway as being significantly silenced. Direct comparison of hypermethylation and downregulated genes identified 4 genes, ADAM17, NOTCH, TWIST1, and SMURF1, that functionally represent the EMT pathway. Single-cell RNA-Seq studies revealed that these effects on gene expression were limited to the keratinocyte cell compartment. Experimental murine studies established that tissue ischemia potently induces wound-edge gene methylation and that 5′-azacytidine, inhibitor of methylation, improved wound closure. To specifically address the significance of TP53 methylation, keratinocyte-specific editing of TP53 methylation at the wound edge was achieved by a tissue nanotransfection-based CRISPR/dCas9 approach. This work identified that reversal of methylation-dependent keratinocyte gene silencing represents a productive therapeutic strategy to improve wound closure.
Collapse
Affiliation(s)
- Kanhaiya Singh
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Yashika Rustagi
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Ahmed S Abouhashem
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Saba Tabasum
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Priyanka Verma
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Edward Hernandez
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Ropar, India
| | - Dolly K Khona
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sujit K Mohanty
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Manishekhar Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Rajneesh Srivastava
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Poornachander R Guda
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sumit S Verma
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sanskruti Mahajan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Jackson A Killian
- Department of Physics, Ohio State University, Columbus, United States of America
| | - Logan A Walker
- Department of Physics, Ohio State University, Columbus, United States of America
| | - Subhadip Ghatak
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Shomita S Mathew-Steiner
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Kristen Wanczyk
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sheng Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, United States of America
| | - Jun Wan
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, United States of America
| | - Pearlly Yan
- Comprehensive Cancer Center, Ohio State University, Columbus, United States of America
| | - Ralf Bundschuh
- Department of Physics, Ohio State University, Columbus, United States of America
| | - Savita Khanna
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Gayle M Gordillo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Michael P Murphy
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Sashwati Roy
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| | - Chandan K Sen
- Department of Surgery, Indiana University School of Medicine, Indianapolis, United States of America
| |
Collapse
|
43
|
Bach MS, de Vries CR, Khosravi A, Sweere JM, Popescu MC, Chen Q, Demirdjian S, Hargil A, Van Belleghem JD, Kaber G, Hajfathalian M, Burgener EB, Liu D, Tran QL, Dharmaraj T, Birukova M, Sunkari V, Balaji S, Ghosh N, Mathew-Steiner SS, El Masry MS, Keswani SG, Banaei N, Nedelec L, Sen CK, Chandra V, Secor PR, Suh GA, Bollyky PL. Filamentous bacteriophage delays healing of Pseudomonas-infected wounds. Cell Rep Med 2022; 3:100656. [PMID: 35732145 PMCID: PMC9244996 DOI: 10.1016/j.xcrm.2022.100656] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/29/2022] [Accepted: 05/17/2022] [Indexed: 01/09/2023]
Abstract
Chronic wounds infected by Pseudomonas aeruginosa (Pa) are characterized by disease progression and increased mortality. We reveal Pf, a bacteriophage produced by Pa that delays healing of chronically infected wounds in human subjects and animal models of disease. Interestingly, impairment of wound closure by Pf is independent of its effects on Pa pathogenesis. Rather, Pf impedes keratinocyte migration, which is essential for wound healing, through direct inhibition of CXCL1 signaling. In support of these findings, a prospective cohort study of 36 human patients with chronic Pa wound infections reveals that wounds infected with Pf-positive strains of Pa are more likely to progress in size compared with wounds infected with Pf-negative strains. Together, these data implicate Pf phage in the delayed wound healing associated with Pa infection through direct manipulation of mammalian cells. These findings suggest Pf may have potential as a biomarker and therapeutic target in chronic wounds.
Collapse
Affiliation(s)
- Michelle S Bach
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Immunology, Stanford University, Stanford, CA 94305, USA
| | - Christiaan R de Vries
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Arya Khosravi
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Johanna M Sweere
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Immunology, Stanford University, Stanford, CA 94305, USA
| | - Medeea C Popescu
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Immunology, Stanford University, Stanford, CA 94305, USA
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sally Demirdjian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Aviv Hargil
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jonas D Van Belleghem
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Maryam Hajfathalian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth B Burgener
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Dan Liu
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Quynh-Lam Tran
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Tejas Dharmaraj
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Immunology, Stanford University, Stanford, CA 94305, USA
| | - Maria Birukova
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Immunology, Stanford University, Stanford, CA 94305, USA
| | - Vivekananda Sunkari
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Immunology, Stanford University, Stanford, CA 94305, USA
| | - Swathi Balaji
- Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nandini Ghosh
- Department of Surgery, Indiana University, Indianapolis, IN 46202, USA
| | | | | | - Sundeep G Keswani
- Division of Pediatric Surgery, Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Niaz Banaei
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Division of Pathology, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Laurence Nedelec
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Chandan K Sen
- Department of Surgery, Indiana University, Indianapolis, IN 46202, USA
| | - Venita Chandra
- Department of Surgery, Division of Vascular Surgery, Stanford University, Stanford, CA 94305, USA
| | - Patrick R Secor
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Gina A Suh
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55902, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
44
|
Resistance Is Not Futile: The Role of Quorum Sensing Plasticity in Pseudomonas aeruginosa Infections and Its Link to Intrinsic Mechanisms of Antibiotic Resistance. Microorganisms 2022; 10:microorganisms10061247. [PMID: 35744765 PMCID: PMC9228389 DOI: 10.3390/microorganisms10061247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 01/01/2023] Open
Abstract
Bacteria use a cell-cell communication process called quorum sensing (QS) to orchestrate collective behaviors. QS relies on the group-wide detection of extracellular signal molecules called autoinducers (AI). Quorum sensing is required for virulence and biofilm formation in the human pathogen Pseudomonas aeruginosa. In P. aeruginosa, LasR and RhlR are homologous LuxR-type soluble transcription factor receptors that bind their cognate AIs and activate the expression of genes encoding functions required for virulence and biofilm formation. While some bacterial signal transduction pathways follow a linear circuit, as phosphoryl groups are passed from one carrier protein to another ultimately resulting in up- or down-regulation of target genes, the QS system in P. aeruginosa is a dense network of receptors and regulators with interconnecting regulatory systems and outputs. Once activated, it is not understood how LasR and RhlR establish their signaling hierarchy, nor is it clear how these pathway connections are regulated, resulting in chronic infection. Here, we reviewed the mechanisms of QS progression as it relates to bacterial pathogenesis and antimicrobial resistance and tolerance.
Collapse
|
45
|
Suh LJ, Khan I, Kelley-Patteson C, Mohan G, Hassanein AH, Sinha M. Breast Implant-Associated Immunological Disorders. J Immunol Res 2022; 2022:8536149. [PMID: 35571560 PMCID: PMC9095406 DOI: 10.1155/2022/8536149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/05/2022] [Accepted: 04/16/2022] [Indexed: 11/18/2022] Open
Abstract
Background Breast implants are commonly placed postbreast cancer reconstruction, cosmetic augmentation, and gender-affirming surgery. Breast implant illness (BII) is a systemic complication associated with breast implants. Patients with BII may experience autoimmune symptoms including fatigue, difficulty concentrating, hair loss, weight change, and depression. BII is poorly understood, and the etiology is unknown. The purpose of this literature review is to characterize BII autoimmune disorders and determine possible causes for its etiology. Methods The PubMed, Google Scholar, Embase, Web of Science, and OVID databases were interrogated from 2010 to 2020 using a query strategy including search term combinations of "implants," "breast implant illness," "autoimmune," and "systemic illness." Results BII includes a spectrum of autoimmune symptoms such as fatigue, myalgias/arthralgias, dry eyes/mouth, and rash. A review of epidemiological studies in the past ten years exhibited evidence affirming an association between breast implants and autoimmune diseases. The most commonly recognized were Sjogren's syndrome, rheumatoid arthritis, systemic sclerosis, chronic fatigue syndrome, and Raynaud's syndrome. Explantation resulted in alleviation of symptoms in over 50% of patients, strengthening the hypothesis linking breast implants to BII. Studies have shown that silicone is a biologically inert material and unlikely to be the cause of these symptoms. This is supported by the fact that increased risk of autoimmune disease was also reported in patients with other implantable biomaterials such as orthopedic implants. Recent studies shed light on a possible role of bacterial biofilm and subsequent host-pathogen interactions as a confounding factor to this problem. Conclusion BII could be dependent on biofilm infection and the microenvironment around the implants. The true pathophysiology behind these complaints must be further investigated so that alternative treatment regimens other than explantation can be developed. Translational significance of these studies is not limited to breast implants but extends to other implants as well.
Collapse
Affiliation(s)
- Lily J. Suh
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Imran Khan
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | - Ganesh Mohan
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Aladdin H. Hassanein
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mithun Sinha
- Department of Surgery, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
46
|
Chen V, Burgess JL, Verpile R, Tomic-Canic M, Pastar I. Novel Diagnostic Technologies and Therapeutic Approaches Targeting Chronic Wound Biofilms and Microbiota. CURRENT DERMATOLOGY REPORTS 2022; 11:60-72. [PMID: 37007641 PMCID: PMC10065746 DOI: 10.1007/s13671-022-00354-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Purpose of Review To provide an up-to-date overview of recent developments in diagnostic methods and therapeutic approaches for chronic wound biofilms and pathogenic microbiota. Recent Findings Biofilm infections are one of the major contributors to impaired wound healing in chronic wounds, including diabetic foot ulcers, venous leg ulcers, pressure ulcers, and nonhealing surgical wounds. As an organized microenvironment commonly including multiple microbial species, biofilms develop and persist through methods that allow evasion from host immune response and antimicrobial treatments. Suppression and reduction of biofilm infection have been demonstrated to improve wound healing outcomes. However, chronic wound biofilms are a challenge to treat due to limited methods for accurate, accessible clinical identification and the biofilm's protective properties against therapeutic agents. Here we review recent approaches towards visual markers for less invasive, enhanced biofilm detection in the clinical setting. We outline progress in wound care treatments including investigation of their antibiofilm effects, such as with hydrosurgical and ultrasound debridement, negative pressure wound therapy with instillation, antimicrobial peptides, nanoparticles and nanocarriers, electroceutical dressings, and phage therapy. Summary Current evidence for biofilm-targeted treatments has been primarily conducted in preclinical studies, with limited clinical investigation for many therapies. Improved identification, monitoring, and treatment of biofilms require expansion of point-of-care visualization methods and increased evaluation of antibiofilm therapies in robust clinical trials.
Collapse
|
47
|
Li D, Niu G, Landén NX. Beyond the Code: Noncoding RNAs in Skin Wound Healing. Cold Spring Harb Perspect Biol 2022; 14:a041230. [PMID: 35197246 PMCID: PMC9438779 DOI: 10.1101/cshperspect.a041230] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
An increasing number of noncoding RNAs (ncRNAs) have been found to regulate gene expression and protein functions, playing important roles in diverse biological processes and diseases. Their crucial functions have been reported in almost every cell type and all stages of skin wound healing. Evidence of their pathogenetic roles in common wound complications, such as chronic nonhealing wounds and excessive scarring, is also accumulating. Given their unique expression and functional properties, ncRNAs are promising therapeutic and diagnostic entities. In this review, we discuss current knowledge about the functional roles of noncoding elements, such as microRNAs, long ncRNAs, and circular RNAs, in skin wound healing, focusing on in vivo evidence from studies of human wound samples and animal wound models. Finally, we provide a perspective on the outlook of ncRNA-based therapeutics in wound care.
Collapse
Affiliation(s)
- Dongqing Li
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Guanglin Niu
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Ning Xu Landén
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 17176 Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institute, 17177 Stockholm, Sweden
| |
Collapse
|
48
|
Aswathanarayan JB, Rao P, HM S, GS S, Rai RV. Biofilm-Associated Infections in Chronic Wounds and Their Management. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022. [DOI: 10.1007/5584_2022_738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
49
|
Li X, Ponandai‐Srinivasan S, Nandakumar KS, Fabre S, Xu Landén N, Mavon A, Khmaladze I. Targeting microRNA for improved skin health. Health Sci Rep 2021; 4:e374. [PMID: 34667882 PMCID: PMC8506131 DOI: 10.1002/hsr2.374] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND In human skin, miRNAs have important regulatory roles and are involved in the development, morphogenesis, and maintenance by influencing cell proliferation, differentiation, immune regulation, and wound healing. MiRNAs have been investigated for many years in various skin disorders such as atopic dermatitis, psoriasis, as well as malignant tumors. Only during recent times, cosmeceutical use of molecules/natural active ingredients to regulate miRNA expression for significant advances in skin health/care product development was recognized. AIM To review miRNAs with the potential to maintain and boost skin health and avoid premature aging by improving barrier function, preventing photoaging, hyperpigmentation, and chronological aging/senescence. METHODS Most of the cited articles were found through literature search on PubMed. The main search criteria was a keyword "skin" in combination with the following words: miRNA, photoaging, UV, barrier, aging, exposome, acne, wound healing, pigmentation, pollution, and senescence. Most of the articles reviewed for relevancy were published during the past 10 years. RESULTS All results are summarized in Figure 1, and they are based on cited references. CONCLUSIONS Thus, regulating miRNAs expression is a promising approach for novel therapy not only for targeting skin diseases but also for cosmeceutical interventions aiming to boost skin health.
Collapse
Affiliation(s)
- Xi Li
- Oriflame Cosmetics AB; Skin Research InstituteStockholmSweden
| | - Sakthi Ponandai‐Srinivasan
- Division of Obstetrics and Gynecology, Department of Women's and Children's HealthKarolinska Institute, and Karolinska University HospitalStockholmSweden
| | - Kutty Selva Nandakumar
- Southern Medical University, School of Pharmaceutical SciencesGuangzhouChina
- Medical Inflammation Research, Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Susanne Fabre
- Oriflame Cosmetics AB; Skin Research InstituteStockholmSweden
| | - Ning Xu Landén
- Department of Medicine, Solna, Dermatology and Venereology, Centre of Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Alain Mavon
- Oriflame Cosmetics AB; Skin Research InstituteStockholmSweden
| | - Ia Khmaladze
- Oriflame Cosmetics AB; Skin Research InstituteStockholmSweden
| |
Collapse
|
50
|
Raval YS, Mohamed A, Flurin L, Mandrekar JN, Greenwood Quaintance KE, Beyenal H, Patel R. Hydrogen-peroxide generating electrochemical bandage is active in vitro against mono- and dual-species biofilms. Biofilm 2021; 3:100055. [PMID: 34585138 PMCID: PMC8455977 DOI: 10.1016/j.bioflm.2021.100055] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/04/2021] [Accepted: 08/07/2021] [Indexed: 12/20/2022] Open
Abstract
Biofilms formed by antibiotic-resistant bacteria in wound beds present unique challenges in terms of treating chronic wound infections; biofilms formed by one or more than one bacterial species are often involved. In this work, the in vitro anti-biofilm activity of a novel electrochemical bandage (e-bandage) composed of carbon fabric and controlled by a wearable potentiostat, designed to continuously deliver low amounts of hydrogen peroxide (H2O2) was evaluated against 34 mono-species and 12 dual-species membrane bacterial biofilms formed by Staphylococcus aureus, S. epidermidis, Enterococcus faecium, E. faecalis, Streptococcus mutans, Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Klebsiella pneumoniae, Cutibacterium acnes, and Bacteroides fragilis. Biofilms were grown on polycarbonate membranes placed atop agar plates. An e-bandage, which electrochemically reduces dissolved oxygen to H2O2 when polarized at -0.6 VAg/AgCl, was then placed atop each membrane biofilm and polarized continuously for 12, 24, and 48 h using a wearable potentiostat. Time-dependent decreases in viable CFU counts of all mono- and dual-species biofilms were observed after e-bandage treatment. 48 h of e-bandage treatment resulted in an average reduction of 8.17 ± 0.40 and 7.99 ± 0.32 log10 CFU/cm2 for mono- and dual-species biofilms, respectively. Results suggest that the described H2O2 producing e-bandage can reduce in vitro viable cell counts of biofilms grown either in mono- or dual-species forms, and should be further developed as a potential antibiotic-free treatment strategy for treating chronic wound infections.
Collapse
Affiliation(s)
- Yash S. Raval
- Division of Clinical Microbiology, Mayo Clinic, Rochester, MN, USA
| | - Abdelrhman Mohamed
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, USA
| | - Laure Flurin
- Division of Clinical Microbiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Haluk Beyenal
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, USA
| | - Robin Patel
- Division of Clinical Microbiology, Mayo Clinic, Rochester, MN, USA
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|