101
|
Staphylococcus aureus biofilms release leukocidins to elicit extracellular trap formation and evade neutrophil-mediated killing. Proc Natl Acad Sci U S A 2018; 115:7416-7421. [PMID: 29941565 DOI: 10.1073/pnas.1721949115] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bacterial biofilms efficiently evade immune defenses, greatly complicating the prognosis of chronic infections. How methicillin-resistant Staphylococcus aureus (MRSA) biofilms evade host immune defenses is largely unknown. This study describes some of the major mechanisms required for S. aureus biofilms to evade the innate immune response and provides evidence of key virulence factors required for survival and persistence of bacteria during chronic infections. Neutrophils are the most abundant white blood cells in circulation, playing crucial roles in the control and elimination of bacterial pathogens. Specifically, here we show that, unlike single-celled populations, S. aureus biofilms rapidly skew neutrophils toward neutrophil extracellular trap (NET) formation through the combined activity of leukocidins Panton-Valentine leukocidin and γ-hemolysin AB. By eliciting this response, S. aureus was able to persist, as the antimicrobial activity of released NETs was ineffective at clearing biofilm bacteria. Indeed, these studies suggest that NETs could inadvertently potentiate biofilm infections. Last, chronic infection in a porcine burn wound model clearly demonstrated that leukocidins are required for "NETosis" and facilitate bacterial survival in vivo.
Collapse
|
102
|
Alves DR, Booth SP, Scavone P, Schellenberger P, Salvage J, Dedi C, Thet NT, Jenkins ATA, Waters R, Ng KW, Overall ADJ, Metcalfe AD, Nzakizwanayo J, Jones BV. Development of a High-Throughput ex-Vivo Burn Wound Model Using Porcine Skin, and Its Application to Evaluate New Approaches to Control Wound Infection. Front Cell Infect Microbiol 2018; 8:196. [PMID: 29963501 PMCID: PMC6013584 DOI: 10.3389/fcimb.2018.00196] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/25/2018] [Indexed: 12/05/2022] Open
Abstract
Biofilm formation in wounds is considered a major barrier to successful treatment, and has been associated with the transition of wounds to a chronic non-healing state. Here, we present a novel laboratory model of wound biofilm formation using ex-vivo porcine skin and a custom burn wound array device. The model supports high-throughput studies of biofilm formation and is compatible with a range of established methods for monitoring bacterial growth, biofilm formation, and gene expression. We demonstrate the use of this model by evaluating the potential for bacteriophage to control biofilm formation by Staphylococcus aureus, and for population density dependant expression of S. aureus virulence factors (regulated by the Accessory Gene Regulator, agr) to signal clinically relevant wound infection. Enumeration of colony forming units and metabolic activity using the XTT assay, confirmed growth of bacteria in wounds and showed a significant reduction in viable cells after phage treatment. Confocal laser scanning microscopy confirmed the growth of biofilms in wounds, and showed phage treatment could significantly reduce the formation of these communities. Evaluation of agr activity by qRT-PCR showed an increase in activity during growth in wound models for most strains. Activation of a prototype infection-responsive dressing designed to provide a visual signal of wound infection, was related to increased agr activity. In all assays, excellent reproducibility was observed between replicates using this model.
Collapse
Affiliation(s)
- Diana R Alves
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom.,The Blond McIndoe Research Foundation, Queen Victoria Hospital, East Grinstead, United Kingdom.,The Queen Victoria Hospital NHS Foundation Trust, East Grinstead, United Kingdom
| | - Simon P Booth
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom.,The Queen Victoria Hospital NHS Foundation Trust, East Grinstead, United Kingdom
| | - Paola Scavone
- Department of Microbiology, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Pascale Schellenberger
- Electron Microscopy Imaging Centre, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Jonathan Salvage
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Cinzia Dedi
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Naing-Tun Thet
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - A Toby A Jenkins
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Ryan Waters
- The Pirbright Institute, Woking, United Kingdom
| | - Keng W Ng
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom.,School of Pharmacy, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Andrew D J Overall
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Anthony D Metcalfe
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom.,The Blond McIndoe Research Foundation, Queen Victoria Hospital, East Grinstead, United Kingdom.,School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Jonathan Nzakizwanayo
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom
| | - Brian V Jones
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton, United Kingdom.,The Queen Victoria Hospital NHS Foundation Trust, East Grinstead, United Kingdom.,Department of Biology and Biological Sciences, University of Bath, Bath, United Kingdom
| |
Collapse
|
103
|
Mori R, Tanaka K, Shimokawa I. Identification and functional analysis of inflammation-related miRNAs in skin wound repair. Dev Growth Differ 2018; 60:306-315. [PMID: 29873073 DOI: 10.1111/dgd.12542] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Inflammation at a wound site is essential for preventing infection. However, misregulated inflammation leads to pathologies of the healing process, including chronic non-healing wounds and scarring. MicroRNAs (miRNAs) are key regulators of the inflammatory response and tissue repair, acting by translational processing of target mRNAs. In the final step of miRNA processing, Argonaute 2 (Ago2)-bound mature miRNA complexes bind to target mRNAs and inhibit their translation. A variety of wound healing-related miRNAs have been identified and their misregulation likely contributes to wound pathologies, including scarring and chronic healing. Recently, we have developed an Ago2-bound mature miRNA purification system that uses Ago2 antibody to analyze the expression of miRNAs from wound tissues by microarray and next generation sequencing. We have identified several wound inflammation-related miRNAs via Ago2-target immunoprecipitation assays and next generation sequencing of wound tissues from wild-type and PU.1 knockout mice, which exhibit no inflammatory response because of a lack of immune cell lineages. We demonstrated that miR-142, an identified inflammation-related miRNA, is essential role for neutrophilic chemotaxis via inhibition of small GTPase translation; its misregulation leads to susceptibility to infection against Staphylococcus aureus at skin wound sites. In this review, we summarize recent advances of miRNA studies in skin wound healing, introduce our miRNA purification system using an immunoprecipitation assay method, and discuss the function of miR-142 in skin wound healing.
Collapse
Affiliation(s)
- Ryoichi Mori
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Katsuya Tanaka
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Plastic and Reconstructive Surgery, Ehime Prefectural Center Hospital, Matsuyama, Japan
| | - Isao Shimokawa
- Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
104
|
Li J, Ghatak S, El Masry MS, Das A, Liu Y, Roy S, Lee RJ, Sen CK. Topical Lyophilized Targeted Lipid Nanoparticles in the Restoration of Skin Barrier Function following Burn Wound. Mol Ther 2018; 26:2178-2188. [PMID: 29802017 DOI: 10.1016/j.ymthe.2018.04.021] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 12/25/2022] Open
Abstract
Lyophilized keratinocyte-targeted nanocarriers (TLNκ) loaded with locked nucleic acid (LNA) modified anti-miR were developed for topical application to full thickness burn injury. TLNκ were designed to selectively deliver LNA-anti-miR-107 to keratinocytes using the peptide sequence ASKAIQVFLLAG. TLNκ employed DOTAP/DODAP combination pH-responsive lipid components to improve endosomal escape. To minimize interference of clearance by non-targeted cells, especially immune cells in the acute wound microenvironment, surface charge was neutralized. Lyophilization was performed to extend the shelf life of the lipid nanoparticles (LNPs). Encapsulation efficiency of anti-miR in lyophilized TLNκ was estimated to be 96.54%. Cargo stability of lyophilized TLNκ was tested. After 9 days of loading with anti-miR-210, TLNκ was effective in lowering abundance of the hypoxamiR miR-210 in keratinocytes challenged with hypoxia. Keratinocyte uptake of DiD-labeled TLNκ was selective and exceeded 90% within 4 hr. Topical application of hydrogel-dispersed lyophilized TLNκ encapsulating LNA anti-miR-107 twice a week significantly accelerated wound closure and restoration of skin barrier function. TLNκ/anti-miR-107 application depleted miR-107 and upregulated dicer expression, which accelerated differentiation of keratinocytes. Expression of junctional proteins such as claudin-1, loricrin, filaggrin, ZO-1, and ZO-2 were significantly upregulated following TLNκ/anti-miR-107 treatment. These LNPs are promising as topical therapeutic agents in the management of burn injury.
Collapse
Affiliation(s)
- Jilong Li
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Subhadip Ghatak
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Mohamed S El Masry
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Department of General Surgery (Plastic Surgery Unit), Zagazig University, 44519, Egypt
| | - Amitava Das
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Yang Liu
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Sashwati Roy
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Robert J Lee
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA
| | - Chandan K Sen
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA; Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
105
|
Mihai MM, Preda M, Lungu I, Gestal MC, Popa MI, Holban AM. Nanocoatings for Chronic Wound Repair-Modulation of Microbial Colonization and Biofilm Formation. Int J Mol Sci 2018; 19:E1179. [PMID: 29649179 PMCID: PMC5979353 DOI: 10.3390/ijms19041179] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/16/2022] Open
Abstract
Wound healing involves a complex interaction between immunity and other natural host processes, and to succeed it requires a well-defined cascade of events. Chronic wound infections can be mono- or polymicrobial but their major characteristic is their ability to develop a biofilm. A biofilm reduces the effectiveness of treatment and increases resistance. A biofilm is an ecosystem on its own, enabling the bacteria and the host to establish different social interactions, such as competition or cooperation. With an increasing incidence of chronic wounds and, implicitly, of chronic biofilm infections, there is a need for alternative therapeutic agents. Nanotechnology shows promising openings, either by the intrinsic antimicrobial properties of nanoparticles or their function as drug carriers. Nanoparticles and nanostructured coatings can be active at low concentrations toward a large variety of infectious agents; thus, they are unlikely to elicit emergence of resistance. Nanoparticles might contribute to the modulation of microbial colonization and biofilm formation in wounds. This comprehensive review comprises the pathogenesis of chronic wounds, the role of chronic wound colonization and infection in the healing process, the conventional and alternative topical therapeutic approaches designed to combat infection and stimulate healing, as well as revolutionizing therapies such as nanotechnology-based wound healing approaches.
Collapse
Affiliation(s)
- Mara Mădălina Mihai
- Department of Oncologic Dermatology and Allergology, "Carol Davila" University of Medicine and Pharmacy, 030167 Bucharest, Romania.
- Department of Dermatology, "Elias" University Emergency Hospital, 011461 Bucharest, Romania.
| | - Mădălina Preda
- Department of Microbiology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 030167 Bucharest, Romania.
- Cantacuzino National Medico-Military Research and Development Institute, 050096 Bucharest, Romania.
| | - Iulia Lungu
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, University Politehnica of Bucharest, 060042 București, Romania.
| | - Monica Cartelle Gestal
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, GA 30602, USA.
| | - Mircea Ioan Popa
- Department of Microbiology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 030167 Bucharest, Romania.
- Cantacuzino National Medico-Military Research and Development Institute, 050096 Bucharest, Romania.
| | - Alina Maria Holban
- Department of Microbiology, Faculty of Biology, University of Bucharest, 030018 București, Romania.
- Research Institute of the University of Bucharest (ICUB), 050107 Bucharest, Romania.
| |
Collapse
|
106
|
Rink C, Wernke MM, Powell HM, Gynawali S, Schroeder RM, Kim JY, Denune JA, Gordillo GM, Colvin JM, Sen CK. Elevated vacuum suspension preserves residual-limb skin health in people with lower-limb amputation: Randomized clinical trial. ACTA ACUST UNITED AC 2018; 53:1121-1132. [PMID: 28355039 DOI: 10.1682/jrrd.2015.07.0145] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 04/13/2016] [Indexed: 11/05/2022]
Abstract
A growing number of clinical trials and case reports support qualitative claims that use of an elevated vacuum suspension (EVS) prosthesis improves residual-limb health on the basis of self-reported questionnaires, clinical outcomes scales, and wound closure studies. Here, we report first efforts to quantitatively assess residual-limb circulation in response to EVS. Residual-limb skin health and perfusion of people with lower-limb amputation (N = 10) were assessed during a randomized crossover study comparing EVS with nonelevated vacuum suspension (control) over a 32 wk period using noninvasive probes (transepidermal water loss, laser speckle imaging, transcutaneous oxygen measurement) and functional hyperspectral imaging approaches. Regardless of the suspension system, prosthesis donning decreased perfusion in the residual limb under resting conditions. After 16 wk of use, EVS improved residual-limb oxygenation during treadmill walking. Likewise, prosthesis-induced reactive hyperemia was attenuated with EVS following 16 wk of use. Skin barrier function was preserved with EVS but disrupted after control socket use. Taken together, outcomes suggest chronic EVS use improves perfusion and preserves skin barrier function in people with lower-limb amputation. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; "Evaluation of limb health associated with a prosthetic vacuum socket system": NCT01839123; https://clinicaltrials.gov/ct2/show/NCT01839123?term=NCT01839123&rank=1.
Collapse
Affiliation(s)
- Cameron Rink
- Department of Surgery, Comprehensive Wound Center, and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Heather M Powell
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | - Surya Gynawali
- Department of Surgery, Comprehensive Wound Center, and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Jayne Y Kim
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | | | - Gayle M Gordillo
- Department of Surgery, Comprehensive Wound Center, and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH.,Department of Plastic Surgery, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Chandan K Sen
- Department of Surgery, Comprehensive Wound Center, and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
107
|
Khansa I, Barker JC, Ghatak PD, Sen CK, Gordillo GM. Use of antibiotic impregnated resorbable beads reduces pressure ulcer recurrence: A retrospective analysis. Wound Repair Regen 2018; 26:221-227. [PMID: 29770531 PMCID: PMC7173798 DOI: 10.1111/wrr.12638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 04/22/2018] [Indexed: 12/01/2022]
Abstract
Recurrence of pressure ulcers remains common. We have employed resorbable antibiotic beads as a therapeutic strategy to deliver high local antibiotic concentrations to the debridement site. Our objective was to determine whether the use of resorbable antibiotic- beads would reduce pressure ulcer recurrence. We reviewed all stage IV pressure ulcers treated with excision, partial ostectomy and flap coverage over 16 years. Baseline patient factors (location of ulcer, presence of osteomyelitis, preoperative prealbumin), surgical factors (type of flap, use of antibiotic beads, bone culture results) and postoperative outcomes (ulcer recurrence at 1 year, dehiscence, seroma, cellulitis) were collected. Outcomes of patients who received antibiotic-impregnated beads were compared to those who did not. Eighty-six patients with 120 stage IV pressure ulcers underwent excision and flap coverage. This included 16 ulcers where antibiotic beads were used and 104 where they were not. The overall ulcer recurrence rate at 12 months was 35.8%. The recurrence rate in the group treated with antibiotic beads was significantly lower than the group without beads (12.5% vs. 39.4%, p = 0.03). Overall, complication rates between the two groups were similar (43.8% vs. 51.9%, p = 0.54). No systemic or local toxicity from antibiotic beads occurred. Scanning electron microscopy images of sacral bone from one case showed bacterial biofilm even after debridement. Pressure ulcer recurrence at 1 year after excision and flap coverage decreased significantly with the use of resorbable antibiotic beads.
Collapse
Affiliation(s)
- Ibrahim Khansa
- Department of Plastic Surgery, The Ohio State University, Columbus, Ohio
| | - Jenny C Barker
- Department of Plastic Surgery, The Ohio State University, Columbus, Ohio
| | - Piya Das Ghatak
- Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Chandan K Sen
- Department of Surgery, The Ohio State University, Columbus, Ohio
| | - Gayle M Gordillo
- Department of Plastic Surgery, The Ohio State University, Columbus, Ohio
| |
Collapse
|
108
|
Pestrak MJ, Chaney SB, Eggleston HC, Dellos-Nolan S, Dixit S, Mathew-Steiner SS, Roy S, Parsek MR, Sen CK, Wozniak DJ. Pseudomonas aeruginosa rugose small-colony variants evade host clearance, are hyper-inflammatory, and persist in multiple host environments. PLoS Pathog 2018; 14:e1006842. [PMID: 29394295 PMCID: PMC5812653 DOI: 10.1371/journal.ppat.1006842] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/14/2018] [Accepted: 12/22/2017] [Indexed: 12/23/2022] Open
Abstract
Pseudomonas aeruginosa causes devastating infections in immunocompromised individuals. Once established, P. aeruginosa infections become incredibly difficult to treat due to the development of antibiotic tolerant, aggregated communities known as biofilms. A hyper-biofilm forming clinical variant of P. aeruginosa, known as a rugose small-colony variant (RSCV), is frequently isolated from chronic infections and is correlated with poor clinical outcome. The development of these mutants during infection suggests a selective advantage for this phenotype, but it remains unclear how this phenotype promotes persistence. While prior studies suggest RSCVs could survive by evading the host immune response, our study reveals infection with the RSCV, PAO1ΔwspF, stimulated an extensive inflammatory response that caused significant damage to the surrounding host tissue. In both a chronic wound model and acute pulmonary model of infection, we observed increased bacterial burden, host tissue damage, and a robust neutrophil response during RSCV infection. Given the essential role of neutrophils in P. aeruginosa-mediated disease, we investigated the impact of the RSCV phenotype on neutrophil function. The RSCV phenotype promoted phagocytic evasion and stimulated neutrophil reactive oxygen species (ROS) production. We also demonstrate that bacterial aggregation and TLR-mediated pro-inflammatory cytokine production contribute to the immune response to RSCVs. Additionally, RSCVs exhibited enhanced tolerance to neutrophil-produced antimicrobials including H2O2 and the antimicrobial peptide LL-37. Collectively, these data indicate RSCVs elicit a robust but ineffective neutrophil response that causes significant host tissue damage. This study provides new insight on RSCV persistence, and indicates this variant may have a critical role in the recurring tissue damage often associated with chronic infections.
Collapse
Affiliation(s)
- Matthew J. Pestrak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Sarah B. Chaney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Heather C. Eggleston
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Sheri Dellos-Nolan
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| | - Sriteja Dixit
- Department of Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | | | - Sashwati Roy
- Department of Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | - Matthew R. Parsek
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Chandan K. Sen
- Department of Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
109
|
Das Ghatak P, Mathew-Steiner SS, Pandey P, Roy S, Sen CK. A surfactant polymer dressing potentiates antimicrobial efficacy in biofilm disruption. Sci Rep 2018; 8:873. [PMID: 29343818 PMCID: PMC5772662 DOI: 10.1038/s41598-018-19175-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/13/2017] [Indexed: 12/26/2022] Open
Abstract
A 100% water-soluble surfactant polymer dressing (SPD) that is bio-compatible and non-ionic has been reported to improve wound closure in preliminary clinical studies. The mechanism of action of SPD in wound healing remains unclear. Biofilm infection is a significant problem that hinders proper wound closure. The objective of this study was to characterize the mechanism of action of SPD inhibition of bacterial biofilm development. Static biofilms (48 h) of the primary wound pathogens Pseudomonas aeruginosa (PA01), Staphylococcus aureus (USA300) were grown on polycarbonate membranes and treated with SPD with and without antibiotics for an additional 24 h. The standard antibiotics - tobramycin (10 μg/ml) for PA01 and rifampicin (10 μg/ml) for USA300, were used in these studies. Following 24 h treatment with and without antibiotics, the biofilms were characterized using scanning electron microscopy (SEM) structural imaging, in vitro imaging system (IVIS) proliferation imaging, colony forming units (CFU), viability assay, quantitative PCR (qPCR) for virulence gene expression. Because SPD is a surfactant based dressing, it potentially has a direct effect on Gram negative bacteria such as Pseudomonas primarily due to the lipid-based outer membrane of the bacteria. SPD is a surfactant based dressing that has potent anti-biofilm properties directly or in synergy with antibiotics.
Collapse
Affiliation(s)
- Piya Das Ghatak
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, 43210, United States of America
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, 43210, United States of America
| | - Shomita S Mathew-Steiner
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, 43210, United States of America
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, 43210, United States of America
| | - Priyanka Pandey
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, 43210, United States of America
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, 43210, United States of America
| | - Sashwati Roy
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, 43210, United States of America
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, 43210, United States of America
| | - Chandan K Sen
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, 43210, United States of America.
- Center for Regenerative Medicine and Cell-Based Therapies, The Ohio State University, Columbus, 43210, United States of America.
| |
Collapse
|
110
|
Staphylococcus aureus Triggers Induction of miR-15B-5P to Diminish DNA Repair and Deregulate Inflammatory Response in Diabetic Foot Ulcers. J Invest Dermatol 2017; 138:1187-1196. [PMID: 29273315 DOI: 10.1016/j.jid.2017.11.038] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 11/25/2017] [Accepted: 11/27/2017] [Indexed: 01/13/2023]
Abstract
Diabetic foot ulcers (DFUs) are a debilitating complication of diabetes in which bacterial presence, including the frequent colonizer Staphylococcus aureus, contributes to inhibition of healing. MicroRNAs (miRs) play a role in healing and host response to bacterial pathogens. However, the mechanisms by which miR response to cutaneous S. aureus contributes to DFU pathophysiology are unknown. Here, we show that S. aureus inhibits wound closure and induces miR-15b-5p in acute human and porcine wound models and in chronic DFUs. Transcriptome analyses of DFU tissue showed induction of miR-15b-5p to be critical, regulating many cellular processes, including DNA repair and inflammatory response, by suppressing downstream targets IKBKB, WEE1, FGF2, RAD50, MSH2, and KIT. Using a human wound model, we confirmed that S. aureus-triggered miR-15b-5p induction results in suppression of the inflammatory- and DNA repair-related genes IKBKB and WEE1. Inhibition of DNA repair and accumulation of DNA breaks was functionally confirmed by the presence of the pH2AX within colonized DFUs. We conclude that S. aureus induces miR-15b-5p, subsequently repressing DNA repair and inflammatory response, showing a mechanism of inhibition of healing in DFUs previously unreported, to our knowledge. This underscores a previously unknown role of DNA damage repair in the pathophysiology of DFUs colonized with S. aureus.
Collapse
|
111
|
Ray VA, Hill PJ, Stover CK, Roy S, Sen CK, Yu L, Wozniak DJ, DiGiandomenico A. Anti-Psl Targeting of Pseudomonas aeruginosa Biofilms for Neutrophil-Mediated Disruption. Sci Rep 2017; 7:16065. [PMID: 29167572 PMCID: PMC5700069 DOI: 10.1038/s41598-017-16215-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/08/2017] [Indexed: 01/05/2023] Open
Abstract
Bacterial biofilms are recalcitrant to antibiotic therapy and a major cause of persistent and recurrent infections. New antibody-based therapies may offer potential to target biofilm specific components for host-cell mediated bacterial clearance. For Pseudomonas aeruginosa, human monoclonal antibodies (mAbs) targeting the Psl biofilm exopolysaccharide exhibit protective activity against planktonic bacteria in acute infection models. However, anti-Psl mAb activity against P. aeruginosa biofilms is unknown. Here, we demonstrate that anti-Psl mAbs targeting three distinct Psl epitopes exhibit stratified binding in mature in vitro biofilms and bind Psl within the context of a chronic biofilm infection. These mAbs also exhibit differential abilities to inhibit early biofilm events and reduce biomass from mature biofilms in the presence of neutrophils. Importantly, a mAb mixture with neutrophils exhibited the greatest biomass reduction, which was further enhanced when combined with meropenem, a common anti-Pseudomonal carbapenem antibiotic. Moreover, neutrophil-mediated killing of biofilm bacteria correlated with the evident mAb epitope stratification within the biofilm. Overall, our results suggest that anti-Psl mAbs might be promising candidates for adjunctive use with antibiotics to inhibit/disrupt P. aeruginosa biofilms as a result of chronic infection.
Collapse
Affiliation(s)
- Valerie A Ray
- Center for Microbial Interface Biology, Departments of Microbial Infection and Immunity, Microbiology, Ohio State University, Columbus, OH, 43210, USA
| | - Preston J Hill
- Center for Microbial Interface Biology, Departments of Microbial Infection and Immunity, Microbiology, Ohio State University, Columbus, OH, 43210, USA
| | - C Kendall Stover
- Department of Infectious Diseases, MedImmune, LLC, Gaithersburg, MD, 20878, USA
| | - Sashwati Roy
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Surgery, College of Veterinary Medicine, Ohio State University, Columbus, OH, 43210, USA
| | - Chandan K Sen
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Center for Regenerative Medicine and Cell Based Therapies, Ohio State University Medical Center, Columbus, OH, 43210, USA.,Department of Surgery, College of Veterinary Medicine, Ohio State University, Columbus, OH, 43210, USA
| | - Li Yu
- Translational Sciences, MedImmune, LLC, Gaithersburg, MD, 20878, USA
| | - Daniel J Wozniak
- Center for Microbial Interface Biology, Departments of Microbial Infection and Immunity, Microbiology, Ohio State University, Columbus, OH, 43210, USA
| | | |
Collapse
|
112
|
Molecular Determinants of the Thickened Matrix in a Dual-Species Pseudomonas aeruginosa and Enterococcus faecalis Biofilm. Appl Environ Microbiol 2017; 83:AEM.01182-17. [PMID: 28842537 DOI: 10.1128/aem.01182-17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/15/2017] [Indexed: 02/08/2023] Open
Abstract
Biofilms are microbial communities that inhabit various surfaces and are surrounded by extracellular matrices (ECMs). Clinical microbiologists have shown that the majority of chronic infections are caused by biofilms, following the introduction of the first biofilm infection model by J. W. Costerton and colleagues (J. Lam, R. Chan, K. Lam, and J. W. Costerton, Infect Immun 28:546-556, 1980). However, treatments for chronic biofilm infections are still limited to surgical removal of the infected sites. Pseudomonas aeruginosa and Enterococcus faecalis are two frequently identified bacterial species in biofilm infections; nevertheless, the interactions between these two species, especially during biofilm growth, are not clearly understood. In this study, we observed phenotypic changes in a dual-species biofilm of P. aeruginosa and E. faecalis, including a dramatic increase in biofilm matrix thickness. For clear elucidation of the spatial distribution of the dual-species biofilm, P. aeruginosa and E. faecalis were labeled with red and green fluorescence, respectively. E. faecalis was located at the lower part of the dual-species biofilm, while P. aeruginosa developed a structured biofilm on the upper part. Mutants with altered exopolysaccharide (EPS) productions were constructed in order to determine the molecular basis for the synergistic effect of the dual-species biofilm. Increased biofilm matrix thickness was associated with EPSs, not extracellular DNA. In particular, Pel and Psl contributed to interspecies and intraspecies interactions, respectively, in the dual-species P. aeruginosa and E. faecalis biofilm. Accordingly, targeting Pel and Psl might be an effective part of eradicating P. aeruginosa polymicrobial biofilms.IMPORTANCE Chronic infection is a serious problem in the medical field. Scientists have observed that chronic infections are closely associated with biofilms, and the vast majority of infection-causing biofilms are polymicrobial. Many studies have reported that microbes in polymicrobial biofilms interact with each other and that the bacterial interactions result in elevated virulence, in terms of factors, such as infectivity and antibiotic resistance. Pseudomonas aeruginosa and Enterococcus faecalis are frequently isolated pathogens in chronic biofilm infections. Nevertheless, while both bacteria are known to be agents of numerous nosocomial infections and can cause serious diseases, interactions between the bacteria in biofilms have rarely been examined. In this investigation, we aimed to characterize P. aeruginosa and E. faecalis dual-species biofilms and to determine the molecular factors that cause synergistic effects, especially on the matrix thickening of the biofilm. We suspect that our findings will contribute to the development of more efficient methods for eradicating polymicrobial biofilm infections.
Collapse
|
113
|
Singh K, Pal D, Sinha M, Ghatak S, Gnyawali SC, Khanna S, Roy S, Sen CK. Epigenetic Modification of MicroRNA-200b Contributes to Diabetic Vasculopathy. Mol Ther 2017; 25:2689-2704. [PMID: 29037594 DOI: 10.1016/j.ymthe.2017.09.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/01/2017] [Accepted: 09/07/2017] [Indexed: 12/26/2022] Open
Abstract
Hyperglycemia (HG) induces genome-wide cytosine demethylation. Our previous work recognized miR-200b as a critical angiomiR, which must be transiently downregulated to initiate wound angiogenesis. Under HG, miR-200b downregulation is not responsive to injury. Here, we demonstrate that HG may drive vasculopathy by epigenetic modification of a miR promoter. In human microvascular endothelial cells (HMECs), HG also lowered DNA methyltransferases (DNMT-1 and DNMT-3A) and compromised endothelial function as manifested by diminished endothelial nitric oxide (eNOS), lowered LDL uptake, impaired Matrigel tube formation, lower NO production, and compromised VE-cadherin expression. Bisulfite-sequencing documented HG-induced miR-200b promoter hypomethylation in HMECs and diabetic wound-site endothelial cells. In HMECs, HG compromised endothelial function. Methyl donor S-adenosyl-L-methionine (SAM) corrected miR-200b promoter hypomethylaton and rescued endothelial function. In vivo, wound-site administration of SAM to diabetic mice improved wound perfusion by limiting the pathogenic rise of miR-200b. Quantitative stable isotope labeling by amino acids in cell culture (SILAC) proteomics and ingenuity pathway analysis identified HG-induced proteins and principal clusters in HMECs sensitive to the genetic inhibition of miR-200b. This work presents the first evidence of the miR-200b promoter methylation as a critical determinant of diabetic wound angiogenesis.
Collapse
Affiliation(s)
- Kanhaiya Singh
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Durba Pal
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Mithun Sinha
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Subhadip Ghatak
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Surya C Gnyawali
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Savita Khanna
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Sashwati Roy
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Chandan K Sen
- Department of Surgery, Davis Heart and Lung Research Institute, Center for Regenerative Medicine & Cell-Based Therapies, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
114
|
Keck J, Gupta R, Christenson LK, Arulanandam BP. MicroRNA mediated regulation of immunity against gram-negative bacteria. Int Rev Immunol 2017; 36:287-299. [PMID: 28800263 PMCID: PMC6904929 DOI: 10.1080/08830185.2017.1347649] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Evidence over the last couple decades has comprehensively established that short, highly conserved, non-coding RNA species called microRNA (miRNA) exhibit the ability to regulate expression and function of host genes at the messenger RNA (mRNA) level. MicroRNAs play key regulatory roles in immune cell development, differentiation, and protective function. Intrinsic host immune response to invading pathogens rely on intricate orchestrated events in the development of innate and adaptive arms of immunity. We discuss the involvement of miRNAs in regulating these processes against gram negative pathogens in this review.
Collapse
Affiliation(s)
- Jonathon Keck
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| | - Lane K. Christenson
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249
| |
Collapse
|
115
|
Barker JC, Khansa I, Gordillo GM. A Formidable Foe Is Sabotaging Your Results: What You Should Know about Biofilms and Wound Healing. Plast Reconstr Surg 2017; 139:1184e-1194e. [PMID: 28445380 DOI: 10.1097/prs.0000000000003325] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
LEARNING OBJECTIVES After reading this article, the participant should be able to: 1. Describe biofilm pathogenesis as it relates to problem wounds. 2. Understand the preclinical and clinical evidence implicating biofilm in problem wounds. 3. Explain the diagnostic and treatment challenges that biofilms create for problem wounds. 4. Demonstrate a basic understanding of emerging strategies aimed at counteracting these processes. SUMMARY Biofilm represents a protected mode of growth for bacteria, allowing them to evade standard diagnostic techniques and avoid eradication by standard therapies. Although only recently discovered, biofilm has existed for millennia and complicates nearly every aspect of medicine. Biofilm impacts wound healing by allowing bacteria to evade immune responses, prolonging inflammation and disabling skin barrier function. It is important to understand why problem wounds persist despite state-of-the-art treatment, why they are difficult to accurately diagnose, and why they recur. The aim of this article is to focus on current gaps in knowledge related to problem wounds, specifically, biofilm infection.
Collapse
Affiliation(s)
- Jenny C Barker
- Columbus, Ohio.,From the Department of Plastic Surgery, Comprehensive Wound Center, The Ohio State University
| | - Ibrahim Khansa
- Columbus, Ohio.,From the Department of Plastic Surgery, Comprehensive Wound Center, The Ohio State University
| | - Gayle M Gordillo
- Columbus, Ohio.,From the Department of Plastic Surgery, Comprehensive Wound Center, The Ohio State University
| |
Collapse
|
116
|
Chaney SB, Ganesh K, Mathew-Steiner S, Stromberg P, Roy S, Sen CK, Wozniak DJ. Histopathological comparisons of Staphylococcus aureus and Pseudomonas aeruginosa experimental infected porcine burn wounds. Wound Repair Regen 2017; 25:541-549. [PMID: 28466497 DOI: 10.1111/wrr.12527] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/21/2017] [Indexed: 12/17/2022]
Abstract
Chronic skin wounds are a significant human health concern and are often complicated by infection with Pseudomonas aeruginosa and Staphylococcus aureus, particularly methicillin resistant S. aureus (MRSA). Translating the knowledge gained from extensive study of virulence mechanisms and pathogenesis of these bacterial species to new treatment modalities has been lacking in part due to a paucity of animal models able to recapitulate human disease. Our groups recently described a novel porcine chronic burn wound model for the study of bacterial infection; however, the histopathology of infection has yet to be described. The objective of this study is to define the histopathology of this model using important human chronic wound bacterial isolates. Porcine full-thickness burn wounds topically inoculated with P. aeruginosa strain PAO1, MRSA S. aureus strain USA300 or both bacteria were used to define and quantify histopathologic lesions. The development of a systemic, well-defined rubric for analysis allowed for evaluation of differences between infection groups. These differences, which included epithelial migration and proliferation, stromal necrosis, fluid accumulation and intensity and character of the innate and adaptive inflammatory cell responses, were identified temporally between infection groups. Mono-species infected wounds developed a hyper-proliferative wound edge. Coinfected wounds at day 35 had the largest wound sizes, increased amounts of neutrophilic inflammation, immaturity of the wound bed, and retention of necrotic tissue. Infection, regardless of species, inhibited wound contracture at all time points evaluated. Most importantly, this model recapitulated key features of chronic human wounds. Thus, this model will allow researchers to study novel treatment modalities in a biologically relevant animal model while monitoring both host and bacterial responses.
Collapse
Affiliation(s)
- Sarah B Chaney
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio
| | - Kasturi Ganesh
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Shomita Mathew-Steiner
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Paul Stromberg
- Department of Veterinary Bioscience, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Sashwati Roy
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Chandan K Sen
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio
| |
Collapse
|
117
|
Komodo dragon-inspired synthetic peptide DRGN-1 promotes wound-healing of a mixed-biofilm infected wound. NPJ Biofilms Microbiomes 2017. [PMID: 28649410 PMCID: PMC5445593 DOI: 10.1038/s41522-017-0017-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cationic antimicrobial peptides are multifunctional molecules that have a high potential as therapeutic agents. We have identified a histone H1-derived peptide from the Komodo dragon (Varanus komodoensis), called VK25. Using this peptide as inspiration, we designed a synthetic peptide called DRGN-1. We evaluated the antimicrobial and anti-biofilm activity of both peptides against Pseudomonas aeruginosa and Staphylococcus aureus. DRGN-1, more than VK25, exhibited potent antimicrobial and anti-biofilm activity, and permeabilized bacterial membranes. Wound healing was significantly enhanced by DRGN-1 in both uninfected and mixed biofilm (Pseudomonas aeruginosa and Staphylococcus aureus)-infected murine wounds. In a scratch wound closure assay used to elucidate the wound healing mechanism, the peptide promoted the migration of HEKa keratinocyte cells, which was inhibited by mitomycin C (proliferation inhibitor) and AG1478 (epidermal growth factor receptor inhibitor). DRGN-1 also activated the EGFR-STAT1/3 pathway. Thus, DRGN-1 is a candidate for use as a topical wound treatment. Wound infections are a major concern; made increasingly complicated by the emerging, rapid spread of bacterial resistance. The novel synthetic peptide DRGN-1 (inspired by a peptide identified from Komodo dragon) exhibits pathogen-directed and host-directed activities in promoting the clearance and healing of polymicrobial (Pseudomonas aeruginosa & Staphylococcus aureus) biofilm infected wounds. The effectiveness of this peptide cannot be attributed solely to its ability to act upon the bacteria and disrupt the biofilm, but also reflects the peptide’s ability to promsote keratinocyte migration. When applied in a murine model, infected wounds treated with DRGN-1 healed significantly faster than did untreated wounds, or wounds treated with other peptides. The host-directed mechanism of action was determined to be via the EGFR-STAT1/3 pathway. The pathogen-directed mechanism of action was determined to be via anti-biofilm activity and antibacterial activity through membrane permeabilization. This novel peptide may have potential as a future therapeutic for treating infected wounds. A synthetic peptide based on a natural molecule found in the Komodo dragon promotes healing of biofilm-infected wounds. Peptides are small protein-like molecules. Monique van Hoek, Barney Bishop and colleagues at George Mason University in Virginia, USA, isolated a natural peptide with some antimicrobial properties from Komodo dragon plasma. They designed a modified synthetic version with rearranged amino acids, named DRGN-1 in recognition of the “Komodo dragon” peptide that inspired it. In preliminary trials, DRGN-1 enhanced the healing of biofilm-infected wounds in mice, and was more effective than the natural peptide. This may be due to both bacterial- and host-directed effects. DRGN-1 reduced biofilm and bacterial number while increasing wound closure. The authors suggest DRGN-1 could be developed into a therapeutic agent that may treat the biofilm-infected wounds that are increasingly resistant to conventional antibiotics.
Collapse
|
118
|
Percival SL. Importance of biofilm formation in surgical infection. Br J Surg 2017; 104:e85-e94. [PMID: 28121033 DOI: 10.1002/bjs.10433] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 10/26/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND Biofilms are ubiquitous, and have been observed in both acute and chronic wounds. Their role in wound healing and infection, however, remains controversial. The aim of this review was to provide an overview of the role and relevance of biofilms to surgical wounds. METHODS A search of PubMed, Science Direct and Web of Science databases was performed to identify studies related to biofilms. Specifically, studies were sought in acute and chronic wounds, and the management and treatment of non-healing and infected skin and wounds. RESULTS Biofilms may develop in all open wounds. In chronic wounds, biofilms may play a role in prolonging and preventing healing, causing chronic inflammation and increasing the risk of infection. Controversies exist regarding the methods presently employed for biofilm detection and management and few data exist to underpin these decisions. CONCLUSION Biofilms in acute surgical and chronic wounds appear to cause a delay in healing and potentially increase the risk of infection. Biofilms can be prevented and once developed can be controlled using wound desloughing and debridement.
Collapse
Affiliation(s)
- S L Percival
- 5D Health Protection Group Ltd, Liverpool Bio-Innovation Hub, William Duncan Building, West Derby Street, Liverpool L7 8TX, UK
| |
Collapse
|
119
|
Herter EK, Xu Landén N. Non-Coding RNAs: New Players in Skin Wound Healing. Adv Wound Care (New Rochelle) 2017; 6:93-107. [PMID: 28289554 PMCID: PMC5346954 DOI: 10.1089/wound.2016.0711] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 11/26/2016] [Indexed: 12/22/2022] Open
Abstract
Significance: Wound healing is a basic physiological process that is utilized to keep the integrity of the skin. Impaired wound repair, such as chronic wounds and pathological scars, presents a major health and economic burden worldwide. To date, efficient targeted treatment for these wound disorders is still lacking, which is largely due to our limited understanding of the biological mechanisms underlying these diseases. Research driven around discovering new therapies for these complications is, therefore, an urgent need. Recent Advances: The vast majority of the human genome is transcribed to RNAs that lack protein-coding capacity. Intensive research in the recent decade has revealed that these non-coding RNAs (ncRNAs) function as important regulators of cellular physiology and pathology, which makes them promising therapeutic and diagnostic entities. Critical Issues: A class of short ncRNAs, microRNAs, has been found to be indispensable for all the phases of skin wound healing and plays important roles in the pathogenesis of wound complications. The role of long ncRNAs (lncRNA) in skin wound healing remains largely unexplored. Recent studies revealed the essential role of lncRNAs in epidermal differentiation and stress response, indicating their potential importance for skin wound healing, which warrants future research. Future Directions: An investigation of ncRNAs will add new layers of complexity to our understanding of normal skin wound healing as well as to the pathogenesis of wound disorders. Development of ncRNA-based biomarkers and treatments is an interesting and important avenue for future research on wound healing.
Collapse
Affiliation(s)
- Eva K. Herter
- Unit of Dermatology and Venereology, Molecular Dermatology Research Group, Department of Medicine, Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| | - Ning Xu Landén
- Unit of Dermatology and Venereology, Molecular Dermatology Research Group, Department of Medicine, Center for Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
120
|
Landis RF, Gupta A, Lee YW, Wang LS, Golba B, Couillaud B, Ridolfo R, Das R, Rotello VM. Cross-Linked Polymer-Stabilized Nanocomposites for the Treatment of Bacterial Biofilms. ACS NANO 2017; 11:946-952. [PMID: 28005325 PMCID: PMC5848076 DOI: 10.1021/acsnano.6b07537] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Infections caused by bacterial biofilms are an emerging threat to human health. Conventional antibiotic therapies are ineffective against biofilms due to poor penetration of the extracellular polymeric substance secreted by colonized bacteria coupled with the rapidly growing number of antibiotic-resistant strains. Essential oils are promising natural antimicrobial agents; however, poor solubility in biological conditions limits their applications against bacteria in both dispersed (planktonic) and biofilm settings. We report here an oil-in-water cross-linked polymeric nanocomposite (∼250 nm) incorporating carvacrol oil that penetrates and eradicates multidrug-resistant (MDR) biofilms. The therapeutic potential of these materials against challenging wound biofilm infections was demonstrated through specific killing of bacteria in a mammalian cell-biofilm coculture wound model.
Collapse
Affiliation(s)
- Ryan F. Landis
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Akash Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Li-Sheng Wang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Bianka Golba
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- Department of Chemistry, Boğaziçi University, Bebek, Istanbul, Turkey, 34342
| | - Brice Couillaud
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- Department of Chemistry, University of Pierre and Marie Curie Paris 6, 4 Place Jussieu, Paris, France, 75005
| | - Roxane Ridolfo
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
- Department of Polymer Science, Colloids and Formulation, ENSCBP – Bordeaux INP, 16 Avenue Pey Berland, Pessac, France 33607
| | - Riddha Das
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| |
Collapse
|
121
|
Malone M, Bjarnsholt T, McBain A, James G, Stoodley P, Leaper D, Tachi M, Schultz G, Swanson T, Wolcott R. The prevalence of biofilms in chronic wounds: a systematic review and meta-analysis of published data. J Wound Care 2017; 26:20-25. [DOI: 10.12968/jowc.2017.26.1.20] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- M. Malone
- Head of Department, Podiatric Medicine, Global Wound Biofilm Expert Panel; Liverpool Hospital, South West Sydney LHD, Australia; and Ingham Institute of Applied Medical Research, Sydney, Australia
| | - T. Bjarnsholt
- University of Copenhagen, Costerton Biofilm Center, Denmark; and Department of Clinical Microbiology, Rigshospitalet, Denmark
| | - A.J. McBain
- Faculty of Biology, Medicine and Health, University of Manchester
| | - G.A. James
- Center for Biofilm Engineering, Montana State University, US
| | - P. Stoodley
- Center for Microbial Interface Biology and Department of Microbial infection, Immunity and Orthopaedics, Ohio State University, US
| | - D. Leaper
- Institute of Skin Integrity and Infection Prevention, University of Huddersfield, UK; and Imperial College, London, UK
| | - M. Tachi
- Tohoku University Graduate School of Medicine, Sendai, Japan
| | - G. Schultz
- Institute of Wound Research, Department of Obstetrics and Gynecology, College of Medicine, University of Florida
| | - T. Swanson
- South West Healthcare, Warrnambool, Victoria, Australia
| | - R.D. Wolcott
- Southwest Regional Wound Care Centre, Lubbock Texas, US
| |
Collapse
|
122
|
Sen CK, Ghatak S, Gnyawali SC, Roy S, Gordillo GM. Cutaneous Imaging Technologies in Acute Burn and Chronic Wound Care. Plast Reconstr Surg 2016; 138:119S-128S. [PMID: 27556752 PMCID: PMC5207795 DOI: 10.1097/prs.0000000000002654] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Wound assessment relies on visual evaluation by physicians. Such assessment is largely subjective and presents the opportunity to explore the use of emergent technologies. METHODS Emergent and powerful noninvasive imaging technologies applicable to assess burn and chronic wounds are reviewed. RESULTS The need to estimate wound depth is critical in both chronic wound and burn injury settings. Harmonic ultrasound technology is powerful to study wound depth. It addresses the limitations of optical imaging with limited depth of penetration. What if a wound appears epithelialized by visual inspection, which shows no discharge yet is covered by repaired skin that lacks barrier function? In this case although the wound is closed as defined by current standards, it remains functionally open, presenting the risk of infection and other postclosure complications. Thus, assessment of skin barrier function is valuable in the context of assessing wound closure. Options for the study of tissue vascularization are many. If noncontact and noninvasive criteria are of importance, laser speckle imaging is powerful. Fluorescence imaging is standard in several clinical settings and is likely to serve the wound clinics well as long as indocyanine green injection is not of concern. A major advantage of harmonic ultrasound imaging of wound depth is that the same system is capable of providing information on blood flow dynamics in arterial perforators. CONCLUSION With many productive imaging platforms to choose from, wound care is about to be transformed by technology that would help assess wound severity.
Collapse
Affiliation(s)
- Chandan K Sen
- Columbus, Ohio
- From the Center for Regenerative Medicine & Cell-Based Therapies, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Subhadip Ghatak
- Columbus, Ohio
- From the Center for Regenerative Medicine & Cell-Based Therapies, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Surya C Gnyawali
- Columbus, Ohio
- From the Center for Regenerative Medicine & Cell-Based Therapies, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Sashwati Roy
- Columbus, Ohio
- From the Center for Regenerative Medicine & Cell-Based Therapies, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| | - Gayle M Gordillo
- Columbus, Ohio
- From the Center for Regenerative Medicine & Cell-Based Therapies, Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center
| |
Collapse
|
123
|
Anghel EL, DeFazio MV, Barker JC, Janis JE, Attinger CE. Current Concepts in Debridement. Plast Reconstr Surg 2016; 138:82S-93S. [DOI: 10.1097/prs.0000000000002651] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
124
|
Polymicrobial–Host Interactions during Infection. J Mol Biol 2016; 428:3355-71. [DOI: 10.1016/j.jmb.2016.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/02/2016] [Accepted: 05/04/2016] [Indexed: 02/08/2023]
|
125
|
Watters C, Fleming D, Bishop D, Rumbaugh KP. Host Responses to Biofilm. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 142:193-239. [PMID: 27571696 DOI: 10.1016/bs.pmbts.2016.05.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
From birth to death the human host immune system interacts with bacterial cells. Biofilms are communities of microbes embedded in matrices composed of extracellular polymeric substance (EPS), and have been implicated in both the healthy microbiome and disease states. The immune system recognizes many different bacterial patterns, molecules, and antigens, but these components can be camouflaged in the biofilm mode of growth. Instead, immune cells come into contact with components of the EPS matrix, a diverse, hydrated mixture of extracellular DNA (bacterial and host), proteins, polysaccharides, and lipids. As bacterial cells transition from planktonic to biofilm-associated they produce small molecules, which can increase inflammation, induce cell death, and even cause necrosis. To survive, invading bacteria must overcome the epithelial barrier, host microbiome, complement, and a variety of leukocytes. If bacteria can evade these initial cell populations they have an increased chance at surviving and causing ongoing disease in the host. Planktonic cells are readily cleared, but biofilms reduce the effectiveness of both polymorphonuclear neutrophils and macrophages. In addition, in the presence of these cells, biofilm formation is actively enhanced, and components of host immune cells are assimilated into the EPS matrix. While pathogenic biofilms contribute to states of chronic inflammation, probiotic Lactobacillus biofilms cause a negligible immune response and, in states of inflammation, exhibit robust antiinflammatory properties. These probiotic biofilms colonize and protect the gut and vagina, and have been implicated in improved healing of damaged skin. Overall, biofilms stimulate a unique immune response that we are only beginning to understand.
Collapse
Affiliation(s)
- C Watters
- Wound Infections Department, Naval Medical Research Center, Silver Spring, MD, United States
| | - D Fleming
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - D Bishop
- Wound Infections Department, Naval Medical Research Center, Silver Spring, MD, United States
| | - K P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States.
| |
Collapse
|
126
|
Roy DC, Tomblyn S, Isaac KM, Kowalczewski CJ, Burmeister DM, Burnett LR, Christy RJ. Ciprofloxacin-loaded keratin hydrogels reduce infection and support healing in a porcine partial-thickness thermal burn. Wound Repair Regen 2016; 24:657-68. [PMID: 27238250 DOI: 10.1111/wrr.12449] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 05/12/2016] [Indexed: 12/31/2022]
Abstract
Infection is a leading cause of morbidity and mortality in burn patients. Current therapies include silver-based creams and dressings, which display limited antimicrobial effectiveness and impair healing. The need exists for a topical, point-of-injury antibiotic treatment that provides sustained antimicrobial activity without impeding wound repair. Fitting this description are keratin-based hydrogels, which are fully biocompatible and support the slow-release of antibiotics. Here we develop a porcine model of an infected partial-thickness burn to test the effects of ciprofloxacin-loaded keratin hydrogels on infection and wound healing. Partial-thickness burns were inoculated with either Pseudomonas aeruginosa or Methicillin-resistant Staphylococcus aureus, resulting in infections that persisted for >2 weeks that exceeded 10(5) and 10(6) cfu per gram of tissue, respectively. Compared to silver sulfadiazine, ciprofloxacin-loaded keratin hydrogel treatment significantly reduced the amount of P. aeruginosa and S. aureus in the burn by >99% on days 4, 7, 11, and 15 postinjury. Further, burns treated with ciprofloxacin-loaded keratin hydrogels exhibited similar healing patterns as uninfected burns with regards to reepithelialization, macrophage recruitment, and collagen deposition and remodeling. The ability of keratin hydrogels to deliver antibiotics to fight infection and support healing of partial-thickness burns make them a strong candidate as a first-line burn therapy.
Collapse
Affiliation(s)
- Daniel C Roy
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas.,KeraNetics, LLC, Winston-Salem, North Carolina
| | | | - Kameel M Isaac
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | - Christine J Kowalczewski
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas.,KeraNetics, LLC, Winston-Salem, North Carolina
| | - David M Burmeister
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| | | | - Robert J Christy
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas
| |
Collapse
|
127
|
Hou Q, Li M, Lu YH, Liu DH, Li CC. Burn wound healing properties of asiaticoside and madecassoside. Exp Ther Med 2016; 12:1269-1274. [PMID: 27588048 PMCID: PMC4997909 DOI: 10.3892/etm.2016.3459] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 03/17/2016] [Indexed: 02/07/2023] Open
Abstract
The healing of burn wounds has been widely characterized to be highly intricate, involving processes such as neo-vascularization, granulation, re-epithelialization, inflammation and wound contraction. Various therapies are available for the management of burn wounds; however, a truly effective therapeutic strategy has yet to be identified due to safety issues. The aim of the present study was to assess and confirm the burn wound healing properties of the compounds asiaticoside (AE) and madecassoside (MA), which are found in the herb Centella asiatica. The cytotoxic nature of the AE and MA were inspected and were confirmed to be non-toxic up to 500 ppm. The compounds AE and MA increased monocyte chemoattractant protein-1 production, but caused no significant effect on vascular endothelial growth factor production. In addition, an in vivo animal burn model was employed to represent the features of burn wound healing. Hence, the present results warrant the further investigation of C. asiatica extracts for use in burn healing.
Collapse
Affiliation(s)
- Qiang Hou
- Department of Burns, Affiliated Hospital of Taishan Medical College, Tai'an, Shandong 271000, P.R. China
| | - Ming Li
- Department of Burns, Affiliated Hospital of Taishan Medical College, Tai'an, Shandong 271000, P.R. China
| | - Yan-Hua Lu
- Department of Burns, Affiliated Hospital of Taishan Medical College, Tai'an, Shandong 271000, P.R. China
| | - Dong-Hong Liu
- Department of Burns, Affiliated Hospital of Taishan Medical College, Tai'an, Shandong 271000, P.R. China
| | - Cheng-Cun Li
- Department of Burns, Affiliated Hospital of Taishan Medical College, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
128
|
Francis NC, Yao W, Grundfest WS, Taylor ZD. Laser-Generated Shockwaves as a Treatment to Reduce Bacterial Load and Disrupt Biofilm. IEEE Trans Biomed Eng 2016; 64:882-889. [PMID: 27323358 DOI: 10.1109/tbme.2016.2581778] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The goal of this paper is to demonstrate and evaluate the potential efficacy of laser-generated shockwave (LGS) therapy on biofilm infected tissue. METHODS To demonstrate proof of concept, Staphylococcus epidermidis was allowed to proliferate on ex vivo pigskin, until mature biofilm formation was achieved, and then subjected to LGS. Bacterial load between control and treated samples was compared using the swab technique and colony counting. Scanning electron microscopy (SEM) was then used to visualize the biofilm growth and resulting reduction in biofilm coverage from treatment. Images were false colored to improve contrast of biofilm, and percent biofilm coverage was computed, along with biofilm cluster size. RESULTS LGS reduced bacterial load by 69% (p = 0.008). Imaging showed biofilm coverage reduced by 52% and significantly reduced average cluster size (p 0.001). CONCLUSION LGS therapy reduced the burden of bacterial biofilm on ex vivo pigskin and can be visualized using SEM imaging. SIGNIFICANCE LGS therapy is a new treatment for infected wounds, allowing rapid disruption of biofilm to 1) remove bacteria and 2) increase susceptibility of remaining biofilm to topical antibiotics. This can lead to improved wound healing times, reduced patient morbidity, and decreased healthcare costs.
Collapse
|
129
|
Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci 2016; 73:3861-85. [PMID: 27180275 PMCID: PMC5021733 DOI: 10.1007/s00018-016-2268-0] [Citation(s) in RCA: 1010] [Impact Index Per Article: 112.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/22/2016] [Accepted: 05/06/2016] [Indexed: 02/08/2023]
Abstract
The ability to rapidly restore the integrity of a broken skin barrier is critical and is the ultimate goal of therapies for hard-to-heal-ulcers. Unfortunately effective treatments to enhance healing and reduce scarring are still lacking. A deeper understanding of the physiology of normal repair and of the pathology of delayed healing is a prerequisite for the development of more effective therapeutic interventions. Transition from the inflammatory to the proliferative phase is a key step during healing and accumulating evidence associates a compromised transition with wound healing disorders. Thus, targeting factors that impact this phase transition may offer a rationale for therapeutic development. This review summarizes mechanisms regulating the inflammation-proliferation transition at cellular and molecular levels. We propose that identification of such mechanisms will reveal promising targets for development of more effective therapies.
Collapse
|
130
|
Gunn JS, Bakaletz LO, Wozniak DJ. What's on the Outside Matters: The Role of the Extracellular Polymeric Substance of Gram-negative Biofilms in Evading Host Immunity and as a Target for Therapeutic Intervention. J Biol Chem 2016; 291:12538-12546. [PMID: 27129225 DOI: 10.1074/jbc.r115.707547] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Biofilms are organized multicellular communities encased in an extracellular polymeric substance (EPS). Biofilm-resident bacteria resist immunity and antimicrobials. The EPS provides structural stability and presents a barrier; however, a complete understanding of how EPS structure relates to biological function is lacking. This review focuses on the EPS of three Gram-negative pathogens: Pseudomonas aeruginosa, nontypeable Haemophilus influenzae, and Salmonella enterica serovar Typhi/Typhimurium. Although EPS proteins and polysaccharides are diverse, common constituents include extracellular DNA, DNABII (DNA binding and bending) proteins, pili, flagella, and outer membrane vesicles. The EPS biochemistry promotes recalcitrance and informs the design of therapies to reduce or eliminate biofilm burden.
Collapse
Affiliation(s)
- John S Gunn
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, Ohio 43210; Center for Microbial Interface Biology, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Lauren O Bakaletz
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, Ohio 43210; Center for Microbial Interface Biology, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205; Departments of Pediatrics and Otolaryngology, The Research Institute at Nationwide Children's Hospital and Ohio State University, Columbus, Ohio 43210
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, Ohio State University, Columbus, Ohio 43210; Center for Microbial Interface Biology, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205; Department of Microbiology, Ohio State University, Columbus, Ohio 43210.
| |
Collapse
|
131
|
Taylor B, Marsh L, Nosworthy J, Williams D. A novel approach to antibiofilm susceptibility testing using a thermo-reversible matrix. J Wound Care 2016; 25:62, 64-7. [DOI: 10.12968/jowc.2016.25.2.62] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- B.J. Taylor
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff
| | - L.L. Marsh
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff
| | - J.O. Nosworthy
- Advanced Medical Solutions Ltd, 33 Premier Park, Winsford, Cheshire
| | - D.W. Williams
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff
| |
Collapse
|
132
|
Novel burn device for rapid, reproducible burn wound generation. Burns 2016; 42:384-91. [PMID: 26803369 DOI: 10.1016/j.burns.2015.08.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/13/2015] [Accepted: 08/18/2015] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Scarring following full thickness burns leads to significant reductions in range of motion and quality of life for burn patients. To effectively study scar development and the efficacy of anti-scarring treatments in a large animal model (female red Duroc pigs), reproducible, uniform, full-thickness, burn wounds are needed to reduce variability in observed results that occur with burn depth. Prior studies have proposed that initial temperature of the burner, contact time with skin, thermal capacity of burner material, and the amount of pressure applied to the skin need to be strictly controlled to ensure reproducibility. The purpose of this study was to develop a new burner that enables temperature and pressure to be digitally controlled and monitored in real-time throughout burn wound creation and compare it to a standard burn device. METHODS A custom burn device was manufactured with an electrically heated burn stylus and a temperature control feedback loop via an electronic microstat. Pressure monitoring was controlled by incorporation of a digital scale into the device, which measured downward force. The standard device was comprised of a heat resistant handle with a long rod connected to the burn stylus, which was heated using a hot plate. To quantify skin surface temperature and internal stylus temperature as a function of contact time, the burners were heated to the target temperature (200±5°C) and pressed into the skin for 40s to create the thermal injuries. Time to reach target temperature and elapsed time between burns were recorded. In addition, each unit was evaluated for reproducibility within and across three independent users by generating burn wounds at contact times spanning from 5 to 40s at a constant pressure and at pressures of 1 or 3lbs with a constant contact time of 40s. Biopsies were collected for histological analysis and burn depth quantification using digital image analysis (ImageJ). RESULTS The custom burn device maintained both its internal temperature and the skin surface temperature near target temperature throughout contact time. In contrast, the standard burner required more than 20s of contact time to raise the skin surface temperature to target due to its quickly decreasing internal temperature. The custom burner was able to create four consecutive burns in less than half the time of the standard burner. Average burn depth scaled positively with time and pressure in both burn units. However, the distribution of burn depth within each time-pressure combination in the custom device was significantly smaller than with the standard device and independent of user. CONCLUSIONS The custom burn device's ability to continually heat the burn stylus and actively control pressure and temperature allowed for more rapid and reproducible burn wounds. Burns of tailored and repeatable depths, independent of user, provide a platform for the study of anti-scar and other wound healing therapies without the added variable of non-uniform starting injury.
Collapse
|
133
|
Abstract
Interactions between microbes are complex and play an important role in the pathogenesis of infections. These interactions can range from fierce competition for nutrients and niches to highly evolved cooperative mechanisms between different species that support their mutual growth. An increasing appreciation for these interactions, and desire to uncover the mechanisms that govern them, has resulted in a shift from monomicrobial to polymicrobial biofilm studies in different disease models. Here we provide an overview of biofilm models used to study select polymicrobial infections and highlight the impact that the interactions between microbes within these biofilms have on disease progression. Notable recent advances in the development of polymicrobial biofilm-associated infection models and challenges facing the study of polymicrobial biofilms are addressed.
Collapse
Affiliation(s)
- Rebecca A Gabrilska
- Departments of Surgery & Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Kendra P Rumbaugh
- Departments of Surgery & Immunology & Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
134
|
Abstract
BACKGROUND The current standard of care for the prevention and treatment of scarring after burn injury is pressure garment therapy. Although this therapy has been used clinically for many years, controversy remains regarding its efficacy. The authors evaluated the efficacy of pressure garment therapy in a female red Duroc pig burn model in which wound depth could be tightly controlled. METHODS Full-thickness burn wounds were generated on female red Duroc pigs. At day 28 after burn, pressure garment therapy was applied to half the wounds (10 mmHg), with control wounds covered with garments that exerted no compression. Scar area, perfusion, hardness, and elasticity were quantified at days 0, 28, 42, 56, and 72 using computerized planimetry, laser Doppler, and torsional ballistometry. Scar morphology was assessed at days 28, 56, and 76 using histology, immunohistochemistry, and transmission electron microscopy. RESULTS Pressure garment therapy significantly hindered scar contraction, with control scars contracting to 64.6 percent + 13.9 percent original area at day 72, whereas pressure garment therapy scars contracted to 82.7 percent + 17.9 percent original area. Pressure garments significantly reduced skin hardness and increased skin strength by 1.3 times. No difference in perfusion or blood vessel density was observed. The average collagen fiber diameter was greater in control burns than in pressure garment therapy. CONCLUSIONS Pressure garment therapy was effective at reducing scar contraction and improving biomechanics compared with control scars. These results confirm the efficacy of pressure garments and highlight the need to further investigate the role of pressure magnitude and the time of therapy application to enhance efficacy for optimal biomechanics and patient mobility.
Collapse
|
135
|
Datta S, Malhotra L, Dickerson R, Chaffee S, Sen CK, Roy S. Laser capture microdissection: Big data from small samples. Histol Histopathol 2015; 30:1255-69. [PMID: 25892148 PMCID: PMC4665617 DOI: 10.14670/hh-11-622] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Any tissue is made up of a heterogeneous mix of spatially distributed cell types. In response to any (patho) physiological cue, responses of each cell type in any given tissue may be unique and cannot be homogenized across cell-types and spatial co-ordinates. For example, in response to myocardial infarction, on one hand myocytes and fibroblasts of the heart tissue respond differently. On the other hand, myocytes in the infarct core respond differently compared to those in the peri-infarct zone. Therefore, isolation of pure targeted cells is an important and essential step for the molecular analysis of cells involved in the progression of disease. Laser capture microdissection (LCM) is powerful to obtain a pure targeted cell subgroup, or even a single cell, quickly and precisely under the microscope, successfully tackling the problem of tissue heterogeneity in molecular analysis. This review presents an overview of LCM technology, the principles, advantages and limitations and its down-stream applications in the fields of proteomics, genomics and transcriptomics. With powerful technologies and appropriate applications, this technique provides unprecedented insights into cell biology from cells grown in their natural tissue habitat as opposed to those cultured in artificial petri dish conditions.
Collapse
Affiliation(s)
- Soma Datta
- Department of Surgery, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, Laser Capture Molecular Core, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Lavina Malhotra
- Department of Surgery, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, Laser Capture Molecular Core, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ryan Dickerson
- Department of Surgery, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, Laser Capture Molecular Core, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Scott Chaffee
- Department of Surgery, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, Laser Capture Molecular Core, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Chandan K Sen
- Department of Surgery, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, Laser Capture Molecular Core, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Sashwati Roy
- Department of Surgery, Center for Regenerative Medicine and Cell Based Therapies and Comprehensive Wound Center, Laser Capture Molecular Core, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
| |
Collapse
|
136
|
Shaik MM, Kowshik M. Novel melt-down neutralization method for synthesis of chitosan–silver scaffolds for tissue engineering applications. Polym Bull (Berl) 2015. [DOI: 10.1007/s00289-015-1522-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
137
|
Duncan B, Li X, Landis RF, Kim ST, Gupta A, Wang LS, Ramanathan R, Tang R, Boerth JA, Rotello VM. Nanoparticle-Stabilized Capsules for the Treatment of Bacterial Biofilms. ACS NANO 2015; 9:7775-82. [PMID: 26083534 PMCID: PMC5047390 DOI: 10.1021/acsnano.5b01696] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Bacterial biofilms are widely associated with persistent infections. High resistance to conventional antibiotics and prevalent virulence makes eliminating these bacterial communities challenging therapeutic targets. We describe here the fabrication of a nanoparticle-stabilized capsule with a multicomponent core for the treatment of biofilms. The peppermint oil and cinnamaldehyde combination that comprises the core of the capsules act as potent antimicrobial agents. An in situ reaction at the oil/water interface between the nanoparticles and cinnamaldehyde structurally augments the capsules to efficiently deliver the essential oil payloads, effectively eradicating biofilms of clinically isolated pathogenic bacteria strains. In contrast to their antimicrobial action, the capsules selectively promoted fibroblast proliferation in a mixed bacteria/mammalian cell system making them promising for wound healing applications.
Collapse
Affiliation(s)
- Bradley Duncan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Xiaoning Li
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Ryan F. Landis
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Sung Tae Kim
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Akash Gupta
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Li-Sheng Wang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Rajesh Ramanathan
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
- Ian Potter NanoBioSensing Facility and NanoBiotechnology Research Laboratory, School of Applied Sciences, RMIT University, GPO Box 2476 V, Melbourne, VIC 3001, Australia
| | - Rui Tang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Jeffrey A. Boerth
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
138
|
Ganesh K, Sinha M, Mathew-Steiner SS, Das A, Roy S, Sen CK. Chronic Wound Biofilm Model. Adv Wound Care (New Rochelle) 2015; 4:382-388. [PMID: 26155380 DOI: 10.1089/wound.2014.0587] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/05/2014] [Indexed: 12/27/2022] Open
Abstract
Significance: Multispecies microbial biofilms may contribute to wound chronicity by derailing the inherent reparative process of the host tissue. In the biofilm form, bacteria are encased within an extracellular polymeric substance and become recalcitrant to antimicrobials and host defenses. For biofilms of relevance to human health, there are two primary contributing factors: the microbial species involved and host response which, in turn, shapes microbial processes over time. This progressive interaction between microbial species and the host is an iterative process that helps evolve an acute-phase infection to a pathogenic chronic biofilm. Thus, long-term wound infection studies are needed to understand the longitudinal cascade of events that culminate into a pathogenic wound biofilm. Recent Advances: Our laboratory has recently published the first long-term (2 month) study of polymicrobial wound biofilm infection in a translationally valuable porcine wound model. Critical Issues: It is widely recognized that the porcine system represents the most translationally valuable approach to experimentally model human skin wounds. A meaningful experimental biofilm model must be in vivo, include mixed species of clinically relevant microbes, and be studied longitudinally long term. Cross-validation of such experimental findings with findings from biofilm-infected patient wounds is critically important. Future Directions: Additional value may be added to the experimental system described above by studying pigs with underlying health complications (e.g., metabolic syndrome), as is typically seen in patient populations.
Collapse
Affiliation(s)
- Kasturi Ganesh
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Mithun Sinha
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Shomita S. Mathew-Steiner
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Amitava Das
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Sashwati Roy
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Chandan K. Sen
- Department of Surgery, Comprehensive Wound Center, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| |
Collapse
|
139
|
Nunan R, Harding KG, Martin P. Clinical challenges of chronic wounds: searching for an optimal animal model to recapitulate their complexity. Dis Model Mech 2015; 7:1205-13. [PMID: 25359790 PMCID: PMC4213725 DOI: 10.1242/dmm.016782] [Citation(s) in RCA: 312] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The efficient healing of a skin wound is something that most of us take for granted but is essential for surviving day-to-day knocks and cuts, and is absolutely relied on clinically whenever a patient receives surgical intervention. However, the management of a chronic wound – defined as a barrier defect that has not healed in 3 months – has become a major therapeutic challenge throughout the Western world, and it is a problem that will only escalate with the increasing incidence of conditions that impede wound healing, such as diabetes, obesity and vascular disorders. Despite being clinically and molecularly heterogeneous, all chronic wounds are generally assigned to one of three major clinical categories: leg ulcers, diabetic foot ulcers or pressure ulcers. Although we have gleaned much knowledge about the fundamental cellular and molecular mechanisms that underpin healthy, acute wound healing from various animal models, we have learned much less about chronic wound repair pathology from these models. This might largely be because the animal models being used in this field of research have failed to recapitulate the clinical features of chronic wounds. In this Clinical Puzzle article, we discuss the clinical complexity of chronic wounds and describe the best currently available models for investigating chronic wound pathology. We also assess how such models could be optimised to become more useful tools for uncovering pathological mechanisms and potential therapeutic treatments.
Collapse
Affiliation(s)
- Robert Nunan
- Schools of Biochemistry and Physiology and Pharmacology, University of Bristol, Bristol, BS8 1TD, UK.
| | - Keith G Harding
- School of Medicine, University of Cardiff, Cardiff, CF14 4XN, UK
| | - Paul Martin
- Schools of Biochemistry and Physiology and Pharmacology, University of Bristol, Bristol, BS8 1TD, UK. School of Medicine, University of Cardiff, Cardiff, CF14 4XN, UK
| |
Collapse
|
140
|
Sinha M, Ghatak S, Roy S, Sen CK. microRNA-200b as a Switch for Inducible Adult Angiogenesis. Antioxid Redox Signal 2015; 22:1257-72. [PMID: 25761972 PMCID: PMC4410303 DOI: 10.1089/ars.2014.6065] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Revised: 02/26/2015] [Accepted: 03/07/2015] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Angiogenesis is the process by which new blood vessels develop from a pre-existing vascular system. It is required for physiological processes such as developmental biology and wound healing. Angiogenesis also plays a crucial role in pathological conditions such as tumor progression. The underlying importance of angiogenesis necessitates a highly regulated process. RECENT ADVANCES Recent works have demonstrated that the process of angiogenesis is regulated by small noncoding RNA molecules called microRNAs (miRs). These miRs, collectively referred to as angiomiRs, have been reported to have a profound effect on the process of angiogenesis by acting as either pro-angiogenic or anti-angiogenic regulators. CRITICAL ISSUES In this review, we will discuss the role of miR-200b as a regulator of angiogenesis. Once the process of angiogenesis is complete, anti-angiogenic miR-200b has been reported to provide necessary braking. Downregulation of miR-200b has been reported across various tumor types, as deregulated angiogenesis is necessary for tumor development. Transient downregulation of miR-200b in wounds drives wound angiogenesis. FUTURE DIRECTIONS New insights and understanding of the molecular mechanism of regulation of angiogenesis by miR-200b has opened new avenues of possible therapeutic interventions to treat angiogenesis-related patho-physiological conditions. Antioxid. Redox Signal. 22, 1257-1272.
Collapse
Affiliation(s)
- Mithun Sinha
- Center for Regenerative Medicine and Cell Based Therapies, Davis Heart and Lung Research Institute, Ohio State University , Columbus, Ohio
| | | | | | | |
Collapse
|
141
|
Silver oxynitrate, an unexplored silver compound with antimicrobial and antibiofilm activity. Antimicrob Agents Chemother 2015; 59:4031-9. [PMID: 25918137 DOI: 10.1128/aac.05177-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/18/2015] [Indexed: 01/22/2023] Open
Abstract
Historically it has been accepted, and recent research has established, that silver (Ag) is an efficacious antimicrobial agent. A dwindling pipeline of new antibiotics, combined with an increase in the number of antibiotic-resistant infections, is bringing Ag to the fore as a therapeutic compound to treat infectious diseases. Currently, many formulations of Ag are being deployed for commercial and medical purposes, with various degrees of effectiveness at killing microbial cells. Here, we evaluated the antimicrobial and antibiofilm capacity of our lead compound, silver oxynitrate [Ag(Ag3O4)2NO3 or Ag7NO11], against other metal compounds with documented antimicrobial activity, including Ag2SO4, AgNO3, silver sulfadiazine (AgSD), AgO, Ag2O, and CuSO4. Our findings reveal that Ag7NO11 eradicates biofilm and planktonic populations of Pseudomonas aeruginosa, Escherichia coli, Staphylococcus aureus, uropathogenic Escherichia coli (UPEC), fluoroquinolone-resistant Pseudomonas aeruginosa (FQRP), and methicillin-resistant Staphylococcus aureus (MRSA) at lower concentrations than those of the other tested metal salts. Altogether, our results demonstrate that Ag7NO11 has an enhanced efficacy for the treatment of biofilm-forming pathogens.
Collapse
|
142
|
Barrier Function of the Repaired Skin Is Disrupted Following Arrest of Dicer in Keratinocytes. Mol Ther 2015; 23:1201-1210. [PMID: 25896246 DOI: 10.1038/mt.2015.65] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 04/07/2015] [Indexed: 02/07/2023] Open
Abstract
Tissue injury transiently silences miRNA-dependent posttranscriptional gene silencing in its effort to unleash adult tissue repair. Once the wound is closed, miRNA biogenesis is induced averting neoplasia. In this work, we report that Dicer plays an important role in reestablishing the barrier function of the skin post-wounding via a miRNA-dependent mechanism. MicroRNA expression profiling of skin and wound-edge tissue revealed global upregulation of miRNAs following wound closure at day 14 post-wounding with significant induction of Dicer expression. Barrier function of the skin, as measured by trans-epidermal water loss, was compromised in keratinocyte-specific conditional (K14/Lox-Cre) Dicer-ablated mice because of malformed cornified epithelium lacking loricrin expression. Studies on human keratinocytes recognized that loricrin expression was inversely related to the expression of the cyclin-dependent kinase inhibitor p21(Waf1/Cip1). Compared to healthy epidermis, wound-edge keratinocytes from Dicer-ablated skin epidermis revealed elevated p21(Waf1/Cip1) expression. Adenoviral and pharmacological suppression of p21(Waf1/Cip1) in keratinocyte-specific conditional Dicer-ablated mice improved wound healing indicating a role of Dicer in the suppression of p21(Waf1/Cip1). This work upholds p21(Waf1/Cip1) as a druggable target to restore barrier function of skin suffering from loss of Dicer function as would be expected in diabetes and other forms of oxidant insult.
Collapse
|
143
|
Antibiofilm peptides increase the susceptibility of carbapenemase-producing Klebsiella pneumoniae clinical isolates to β-lactam antibiotics. Antimicrob Agents Chemother 2015; 59:3906-12. [PMID: 25896694 DOI: 10.1128/aac.00092-15] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/12/2015] [Indexed: 11/20/2022] Open
Abstract
Multidrug-resistant carbapenemase-producing Klebsiella pneumoniae (KpC) strains are becoming a common cause of infections in health care centers. Furthermore, Klebsiella can develop multicellular biofilms, which lead to elevated adaptive antibiotic resistance. Here, we describe the antimicrobial and antibiofilm activities of synthetic peptides DJK-5, DJK-6, and 1018 against five KpC isolates. Using static microplate assays, it was observed that the concentration required to prevent biofilm formation by these clinical isolates was below the MIC for planktonic cells. More-sophisticated flow cell experiments confirmed the antibiofilm activity of the peptides against 2-day-old biofilms of different KpC isolates, and in some cases, the peptides induced significant biofilm cell death. Clinically relevant combinations of DJK-6 and β-lactam antibiotics, including the carbapenem meropenem, also prevented planktonic growth and biofilm formation of KpC strain1825971. Interestingly, peptide DJK-6 was able to enhance, at least 16-fold, the ability of meropenem to eradicate preformed biofilms formed by this strain. Using peptide DJK-6 to potentiate the activity of β-lactams, including meropenem, represents a promising strategy to treat infections caused by KpC isolates.
Collapse
|
144
|
Silver-zinc redox-coupled electroceutical wound dressing disrupts bacterial biofilm. PLoS One 2015; 10:e0119531. [PMID: 25803639 PMCID: PMC4372374 DOI: 10.1371/journal.pone.0119531] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/20/2015] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa biofilm is commonly associated with chronic wound infection. A FDA approved wireless electroceutical dressing (WED), which in the presence of conductive wound exudate gets activated to generate electric field (0.3–0.9V), was investigated for its anti-biofilm properties. Growth of pathogenic P. aeruginosa strain PAO1 in LB media was markedly arrested in the presence of the WED. Scanning electron microscopy demonstrated that WED markedly disrupted biofilm integrity in a setting where silver dressing was ineffective. Biofilm thickness and number of live bacterial cells were decreased in the presence of WED. Quorum sensing genes lasR and rhlR and activity of electric field sensitive enzyme, glycerol-3-phosphate dehydrogenase was also repressed by WED. This work provides first electron paramagnetic resonance spectroscopy evidence demonstrating that WED serves as a spontaneous source of reactive oxygen species. Redox-sensitive multidrug efflux systems mexAB and mexEF were repressed by WED. Taken together, these observations provide first evidence supporting the anti-biofilm properties of WED.
Collapse
|
145
|
Gnyawali SC, Barki KG, Mathew-Steiner SS, Dixith S, Vanzant D, Kim J, Dickerson JL, Datta S, Powell H, Roy S, Bergdall V, Sen CK. High-resolution harmonics ultrasound imaging for non-invasive characterization of wound healing in a pre-clinical swine model. PLoS One 2015; 10:e0122327. [PMID: 25799513 PMCID: PMC4370665 DOI: 10.1371/journal.pone.0122327] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 02/10/2015] [Indexed: 11/18/2022] Open
Abstract
This work represents the first study employing non-invasive high-resolution harmonic ultrasound imaging to longitudinally characterize skin wound healing. Burn wounds (day 0-42), on the dorsum of a domestic Yorkshire white pig were studied non-invasively using tandem digital planimetry, laser speckle imaging and dual mode (B and Doppler) ultrasound imaging. Wound depth, as measured by B-mode imaging, progressively increased until day 21 and decreased thereafter. Initially, blood flow at the wound edge increased up to day 14 and subsequently regressed to baseline levels by day 21, when the wound was more than 90% closed. Coinciding with regression of blood flow at the wound edge, there was an increase in blood flow in the wound bed. This was observed to regress by day 42. Such changes in wound angiogenesis were corroborated histologically. Gated Doppler imaging quantitated the pulse pressure of the primary feeder artery supplying the wound site. This pulse pressure markedly increased with a bimodal pattern following wounding connecting it to the induction of wound angiogenesis. Finally, ultrasound elastography measured tissue stiffness and visualized growth of new tissue over time. These studies have elegantly captured the physiological sequence of events during the process of wound healing, much of which is anticipated based on certain dynamics in play, to provide the framework for future studies on molecular mechanisms driving these processes. We conclude that the tandem use of non-invasive imaging technologies has the power to provide unprecedented insight into the dynamics of the healing skin tissue.
Collapse
Affiliation(s)
- Surya C. Gnyawali
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Kasturi G. Barki
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Shomita S. Mathew-Steiner
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Sriteja Dixith
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Daniel Vanzant
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Jayne Kim
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Jennifer L. Dickerson
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Soma Datta
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Heather Powell
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States of America
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Sashwati Roy
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
| | - Valerie Bergdall
- Veterinary Preventive Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Chandan K. Sen
- Comprehensive Wound Center, Davis Heart and Lung Research Institute, Centers for Regenerative Medicine and Cell Based Therapies, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
146
|
Brackman G, Coenye T. In Vitro and In Vivo Biofilm Wound Models and Their Application. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 897:15-32. [DOI: 10.1007/5584_2015_5002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|