1
|
Redeghieri P, Moray J, Kerff F, Gohy S, Leal T, Muyldermans S, Vanbever R, Morales‐Yánez FJ, Dumoulin M. Enzymatic, structural, and biophysical characterization of a single-domain antibody (VHH) selectively and tightly inhibiting neutrophil elastase and exhibiting favorable developability properties. Protein Sci 2024; 33:e5227. [PMID: 39604162 PMCID: PMC11602439 DOI: 10.1002/pro.5227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Human neutrophil elastase (hNE), a serine protease released by neutrophils during inflammation, plays a major role in the pathophysiology of several conditions especially in inflammatory lung diseases. Its inhibition constitutes, therefore, a promising therapeutic strategy to combat these diseases. In this work, we characterized the in vitro properties of a VHH (i.e., the antigen binding domain of camelid heavy chain-only antibodies), referred to as NbE201. This VHH is able to inhibit tightly, selectively and competitively both human and murine elastases with the inhibition constants (Ki) of 4.1 ± 0.9 nM and 36.8 ± 3.9 nM, respectively. The IC50 for the inhibition of the hydrolysis of elastin is in the same range to that of alpha-1 antitrypsin (i.e., the main endogenous inhibitor of hNE also used in the clinic) and 14 times better than that of Sivelestat (i.e., the 2nd clinically approved hNE inhibitor). The X-ray crystal structure of the NbE201-hNE complex reveals that the Complementarity Determining Regions CDR1 and CDR3 of the VHH bind into the substrate binding pocket of hNE and prevent the access to small or macromolecular substrates. They do not, however, bind deep enough into the pocket to be hydrolyzed. NbE201 is highly stable towards oxidation, deamidation, and chemical or thermal denaturation. NbE201 is therefore likely to tolerate manufacturing processes during drug development. These results highlight the high potential of NbE201 as a (pre)clinical tool to diagnose and treat diseases associated with excessive hNE activity, and for fundamental research to better understand the role of hNE in these conditions.
Collapse
Affiliation(s)
- Paola Redeghieri
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Joël Moray
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Frédéric Kerff
- Biological Macromolecule Crystallography, Centre for Protein Engineering, InBios, Department of Life SciencesUniversity of LiègeLiègeBelgium
| | - Sophie Gohy
- Department of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
- Cystic Fibrosis Reference CentreCliniques Universitaires Saint‐LucBrusselsBelgium
| | - Teresinha Leal
- Louvain Center for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular ImmunologyVrije Universiteit BrusselBrusselsBelgium
| | - Rita Vanbever
- Louvain Drug Research Institute (LDRI)Université Catholique de LouvainBrusselsBelgium
| | - Francisco Javier Morales‐Yánez
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Mireille Dumoulin
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| |
Collapse
|
2
|
Hartman E, Forsberg F, Kjellström S, Petrlova J, Luo C, Scott A, Puthia M, Malmström J, Schmidtchen A. Peptide clustering enhances large-scale analyses and reveals proteolytic signatures in mass spectrometry data. Nat Commun 2024; 15:7128. [PMID: 39164298 PMCID: PMC11336174 DOI: 10.1038/s41467-024-51589-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 08/13/2024] [Indexed: 08/22/2024] Open
Abstract
Recent advances in mass spectrometry-based peptidomics have catalyzed the identification and quantification of thousands of endogenous peptides across diverse biological systems. However, the vast peptidomic landscape generated by proteolytic processing poses several challenges for downstream analyses and limits the comparability of clinical samples. Here, we present an algorithm that aggregates peptides into peptide clusters, reducing the dimensionality of peptidomics data, improving the definition of protease cut sites, enhancing inter-sample comparability, and enabling the implementation of large-scale data analysis methods akin to those employed in other omics fields. We showcase the algorithm by performing large-scale quantitative analysis of wound fluid peptidomes of highly defined porcine wound infections and human clinical non-healing wounds. This revealed signature phenotype-specific peptide regions and proteolytic activity at the earliest stages of bacterial colonization. We validated the method on the urinary peptidome of type 1 diabetics which revealed potential subgroups and improved classification accuracy.
Collapse
Affiliation(s)
- Erik Hartman
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.
| | - Fredrik Forsberg
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Sven Kjellström
- Division of Mass Spectrometry, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jitka Petrlova
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Congyu Luo
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Aaron Scott
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Manoj Puthia
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Artur Schmidtchen
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
3
|
Kolahreez D, Ghasemi-Mobarakeh L, Quartinello F, Liebner FW, Guebitz GM, Ribitsch D. Multifunctional Casein-Based Wound Dressing Capable of Monitoring and Moderating the Proteolytic Activity of Chronic Wounds. Biomacromolecules 2024; 25:700-714. [PMID: 38295273 PMCID: PMC10865360 DOI: 10.1021/acs.biomac.3c00910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 02/02/2024]
Abstract
Every 1.2 s, a diabetic foot ulcer is developed, and every 20 s, one amputation is carried out in diabetic patients. Monitoring and controlling protease activity have been considered as a strategy for more efficient management of diabetic and other chronic wounds. This study aimed to develop a casein-based dressing that, by its disappearance, provides information about the activity of proteases and simultaneously harnesses proteolytic activity. Casein films were fabricated by using an aqueous solution, and heat treatment was successfully deployed as a green and clean approach to confer hydrolytic stability. Our results showed that casein-based films' mechanical characteristics, water absorption, and proteolytic stability could be controlled by the length of the heat treatment, which proved to be a useful tool. An increase in the treatment duration from 30 min to 3 h led to toleration of 2.4 times higher stress, 2 times lower water uptake, and 3.4 times higher proteolytic stability at examined conditions. Selected casein-based structures responded to Bacillus sp. bacteria's protease (BSP) and human neutrophil elastase (HNE) as representatives of bacterial and nonbacterial proteases found in the wounds at 10 and 200 ng mL-1 levels, respectively. The hydrolysis was accompanied by a 36% reduction in proteolytic activity measured by using a casein-based universal protease activity assay. The released casein fragments could scavenge 90% of the examined radicals. In-vitro cell culture studies showed that the hydrolysates were not cytotoxic, and the casein-based film had a favorable interaction with fibroblast cells, indicating its potential as a scaffold in the case that proteolytic activity would not be to the extent that causes its rapid disintegration. In general, these findings hold promise for applying the developed casein-based structure for detecting proteolytic activity without the need for any equipment, kits, or expertise and, more importantly, in a highly economical manner. In the case that the proteolytic activity would not be severe, it could also serve as a substrate for cell adhesion and growth; this would aid in the healing process.
Collapse
Affiliation(s)
- Davood Kolahreez
- Department
of Textile Engineering, Isfahan University
of Technology, Isfahan 84156-83111, Iran
- Institute
of Environmental Biotechnology, Department of Agrobiotechnology, IFA-Tulln, University of Natural Resources and Life Sciences, Vienna, Konrad-Lorenz-Strasse 20, 3430 Tulln an der Donau, Austria
- Institute
of Chemistry of Renewable Resources, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Konrad-Lorenz-Strasse 24, 3430 Tulln an der Donau, Austria
| | - Laleh Ghasemi-Mobarakeh
- Department
of Textile Engineering, Isfahan University
of Technology, Isfahan 84156-83111, Iran
| | - Felice Quartinello
- Institute
of Environmental Biotechnology, Department of Agrobiotechnology, IFA-Tulln, University of Natural Resources and Life Sciences, Vienna, Konrad-Lorenz-Strasse 20, 3430 Tulln an der Donau, Austria
| | - Falk W. Liebner
- Institute
of Chemistry of Renewable Resources, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Konrad-Lorenz-Strasse 24, 3430 Tulln an der Donau, Austria
| | - Georg M. Guebitz
- Institute
of Environmental Biotechnology, Department of Agrobiotechnology, IFA-Tulln, University of Natural Resources and Life Sciences, Vienna, Konrad-Lorenz-Strasse 20, 3430 Tulln an der Donau, Austria
- Austrian
Centre of Industrial Biotechnology (ACIB), Konrad-Lorenz-Strasse 20, 3430 Tulln an der Donau, Austria
| | - Doris Ribitsch
- Institute
of Environmental Biotechnology, Department of Agrobiotechnology, IFA-Tulln, University of Natural Resources and Life Sciences, Vienna, Konrad-Lorenz-Strasse 20, 3430 Tulln an der Donau, Austria
- Austrian
Centre of Industrial Biotechnology (ACIB), Konrad-Lorenz-Strasse 20, 3430 Tulln an der Donau, Austria
| |
Collapse
|
4
|
Khaledi M, Zandi B, Mohsenipour Z. The Effect of Mesenchymal Stem Cells on the Wound Infection. Curr Stem Cell Res Ther 2024; 19:1084-1092. [PMID: 37815189 DOI: 10.2174/011574888x252482230926104342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/10/2023] [Accepted: 08/17/2023] [Indexed: 10/11/2023]
Abstract
Wound infection often requires a long period of care and an onerous treatment process. Also, the rich environment makes the wound an ideal niche for microbial growth. Stable structures, like biofilm, and drug-resistant strains cause a delay in the healing process, which has become one of the important challenges in wound treatment. Many studies have focused on alternative methods to deal the wound infections. One of the novel and highly potential ways is mesenchymal stromal cells (MSCs). MSCs are mesoderm-derived pluripotent adult stem cells with the capacity for self-renewal, multidirectional differentiation, and immunological control. Also, MSCs have anti-inflammatory and antiapoptotic effects. MScs, as pluripotent stromal cells, differentiate into many mature cells. Also, MSCs produce antimicrobial compounds, such as antimicrobial peptides (AMP), as well as secrete immune modulators, which are two basic features considered in wound healing. Despite the advantages, preserving the structure and activity of MSCs is considered one of the most important points in the treatment. MSCs' antimicrobial effects on microorganisms involved in wound infection have been confirmed in various studies. In this review, we aimed to discuss the antimicrobial and therapeutic applications of MSCs in the infected wound healing processes.
Collapse
Affiliation(s)
- Mansoor Khaledi
- Department of Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Bita Zandi
- Department of Microbiology, Faculty of advanced science and technology, Tehran medical science, Islamic Azad University, Tehran, Iran
| | - Zeinab Mohsenipour
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Dabas M, Kreychman I, Katz T, Gefen A. Testing the effectiveness of a polymeric membrane dressing in modulating the inflammation of intact, non-injured, mechanically irritated skin. Int Wound J 2024; 21:e14347. [PMID: 37568272 PMCID: PMC10777768 DOI: 10.1111/iwj.14347] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
We investigated the inflammatory (IL-1 alpha) and thermal (infrared thermography) reactions of healthy sacral skin to sustained, irritating mechanical loading. We further acquired digital photographs of the irritated skin (at the visible light domain) to assess whether infrared imaging is advantageous. For clinical context, the skin status was monitored under a polymeric membrane dressing known to modulate the inflammatory skin response. The IL-1 alpha and infrared thermography measurements were consistent in representing the skin status after 40 min of continuous irritation. Infrared thermography overpowered conventional digital photography as a contactless optical method for image processing inputs, by revealing skin irritation trends that were undetectable through digital photography in the visual light, not even with the aid of advanced image processing. The polymeric membrane dressings were shown to offer prophylactic benefits over simple polyurethane foam in the aspects of inflammation reduction and microclimate management. We also concluded that infrared thermography is a feasible method for monitoring the skin health status and the risk for pressure ulcers, as it avoids the complexity of biological marker studies and empowers visual skin assessments or digital photography of skin, both of which were shown to be insufficient for detecting the inflammatory skin status.
Collapse
Affiliation(s)
- Mai Dabas
- Department of Biomedical Engineering, Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
| | - Ida Kreychman
- Department of Biomedical Engineering, Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
| | - Tomer Katz
- Department of Biomedical Engineering, Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
| | - Amit Gefen
- Department of Biomedical Engineering, Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
6
|
Zheng H, Cheng X, Jin L, Shan S, Yang J, Zhou J. Recent advances in strategies to target the behavior of macrophages in wound healing. Biomed Pharmacother 2023; 165:115199. [PMID: 37517288 DOI: 10.1016/j.biopha.2023.115199] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 08/01/2023] Open
Abstract
Chronic wounds and scar formation are widespread due to limited suitable remedies. The macrophage is a crucial regulator in wound healing, controlling the onset and termination of inflammation and regulating other processes related to wound healing. The current breakthroughs in developing new medications and drug delivery methods have enabled the accurate targeting of macrophages in oncology and rheumatic disease therapies through clinical trials. These successes have cleared the way to utilize drugs targeting macrophages in various disorders. This review thus summarizes macrophage involvement in normal and pathologic wound healing. It further details the targets available for macrophage intervention and therapeutic strategies for targeting the behavior of macrophages in tissue repair and regeneration.
Collapse
Affiliation(s)
- Hongkun Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xinwei Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lu Jin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Resovi A, Persichitti P, Brunelli L, Minoli L, Borsotti P, Garattini G, Tironi M, Dugnani E, Redegalli M, De Simone G, Pastorelli R, Bani MR, Piemonti L, Mosher DF, Giavazzi R, Taraboletti G, Belotti D. Fibronectin fragments generated by pancreatic trypsin act as endogenous inhibitors of pancreatic tumor growth. J Exp Clin Cancer Res 2023; 42:201. [PMID: 37559126 PMCID: PMC10411016 DOI: 10.1186/s13046-023-02778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND The pancreatic microenvironment has a defensive role against cancer but it can acquire tumor-promoting properties triggered by multiple mechanisms including alterations in the equilibrium between proteases and their inhibitors. The identification of proteolytic events, targets and pathways would set the basis for the design of new therapeutic approaches. METHODS AND RESULTS Here we demonstrate that spheroids isolated from human and murine healthy pancreas and co-transplanted orthotopically with pancreatic ductal adenocarcinoma (PDAC) in mouse pancreas inhibited tumor growth. The effect was mediated by trypsin-generated fibronectin (FN) fragments released by pancreatic spheroids. Tumor inhibition was observed also in a model of acute pancreatitis associated with trypsin activation. Mass spectrometry proteomic analysis of fragments and mAb against different FN epitopes identified the FN type III domain as responsible for the activity. By inhibiting integrin α5β1, FAK and FGFR1 signaling, the fragments induced tumor cell detachment and reduced cell proliferation. Consistent with the mutual relationship between the two pathways, FGF2 restored both FGFR1 and FAK signaling and promoted PDAC cell adhesion and proliferation. FAK and FGFR inhibitors additively inhibited PDAC growth in vitro and in orthotopic in vivo models. CONCLUSIONS This study identifies a novel role for pancreatic trypsin and fibronectin cleavage as a mechanism of protection against cancer by the pancreatic microenvironment. The finding of a FAK-FGFR cross-talk in PDAC support the combination of FAK and FGFR inhibitors for PDAC treatment to emulate the protective effect of the normal pancreas against cancer.
Collapse
Affiliation(s)
- Andrea Resovi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Perla Persichitti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Laura Brunelli
- Department of Environmental Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Lucia Minoli
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Patrizia Borsotti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Giulia Garattini
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Matteo Tironi
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Erica Dugnani
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Miriam Redegalli
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Giulia De Simone
- Department of Environmental Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Roberta Pastorelli
- Department of Environmental Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Maria Rosa Bani
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Deane F Mosher
- Departments of Biomolecular Chemistry and Medicine, University of Wisconsin, Madison, WI, USA
| | - Raffaella Giavazzi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Giulia Taraboletti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy
| | - Dorina Belotti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo and Milan, Italy.
| |
Collapse
|
8
|
Cullen B, Gefen A. The biological and physiological impact of the performance of wound dressings. Int Wound J 2023; 20:1292-1303. [PMID: 36110054 PMCID: PMC10031231 DOI: 10.1111/iwj.13960] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 11/27/2022] Open
Abstract
Chronic wounds affect millions globally and are a huge financial burden. Whilst there are many wound dressings commercially available to manage these wounds, the complexity of the repair process makes it difficult to select the right dressing for the right wound at the right time. Thus, in this narrative review, we have examined reasons why wounds fail to heal, summarised the pathophysiology of the chronic wound environment and provided an evidence-based, clinically-relevant compilation of the published literature relevant to dressing design and evaluation. This has highlighted the need for a deeper understanding of wound exudates, how exudates change throughout the healing process, and how they are impacted by different dressing materials. Studies assessing biochemical and biophysical changes in exudates throughout the healing process are extremely valuable in this regard, enhancing both our understanding of the wound healing process and the ability to assess dressing performance. In addition, this knowledge allows us to replicate various wound conditions in the laboratory, and develop clinically-relevant models for testing current and new dressings, therefore providing a more comprehensive understanding of how and when they should be used. This approach makes the use of dressings more effective, thereby improving outcomes, and reducing the economic burden of chronic wounds.
Collapse
Affiliation(s)
| | - Amit Gefen
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
9
|
Role of Innate Immune Cells in Chronic Diabetic Wounds. J Indian Inst Sci 2023. [DOI: 10.1007/s41745-022-00355-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
10
|
de Souza RB, Lemes RB, Foresto-Neto O, Cassiano LL, Reinhardt DP, Meek KM, Koh IHJ, Lewis PN, Pereira LV. Extracellular matrix and vascular dynamics in the kidney of a murine model for Marfan syndrome. PLoS One 2023; 18:e0285418. [PMID: 37159453 PMCID: PMC10168582 DOI: 10.1371/journal.pone.0285418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 04/24/2023] [Indexed: 05/11/2023] Open
Abstract
Fibrillin-1 is a pivotal structural component of the kidney's glomerulus and peritubular tissue. Mutations in the fibrillin-1 gene result in Marfan syndrome (MFS), an autosomal dominant disease of the connective tissue. Although the kidney is not considered a classically affected organ in MFS, several case reports describe glomerular disease in patients. Therefore, this study aimed to characterize the kidney in the mgΔlpn-mouse model of MFS. Affected animals presented a significant reduction of glomerulus, glomerulus-capillary, and urinary space, and a significant reduction of fibrillin-1 and fibronectin in the glomerulus. Transmission electron microscopy and 3D-ultrastructure analysis revealed decreased amounts of microfibrils which also appeared fragmented in the MFS mice. Increased collagen fibers types I and III, MMP-9, and α-actin were also observed in affected animals, suggesting a tissue-remodeling process in the kidney. Video microscopy analysis showed an increase of microvessel distribution coupled with reduction of blood-flow velocity, while ultrasound flow analysis revealed significantly lower blood flow in the kidney artery and vein of the MFS mice. The structural and hemodynamic changes of the kidney indicate the presence of kidney remodeling and vascular resistance in this MFS model. Both processes are associated with hypertension which is expected to worsen the cardiovascular phenotype in MFS.
Collapse
Affiliation(s)
| | - Renan Barbosa Lemes
- Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, SP, Brazil
| | - Orestes Foresto-Neto
- Faculty of Medicine, Department of Clinical Medicine, Renal Division, University of São Paulo, São Paulo, Brazil
| | | | - Dieter P Reinhardt
- Department of Anatomy and Cell Biology Dentistry and Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Keith M Meek
- Structural Biophysics Research Group, School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Ivan Hong Jun Koh
- Department of Surgery, Federal University of São Paulo, São Paulo, SP, Brazil
| | - Philip N Lewis
- Structural Biophysics Research Group, School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Lygia V Pereira
- Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
11
|
Zhang S, Lin A, Tao Z, Fu Y, Xiao L, Ruan G, Li Y. Microsphere‐containing hydrogel scaffolds for tissue engineering. Chem Asian J 2022; 17:e202200630. [PMID: 35909078 DOI: 10.1002/asia.202200630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/25/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Shihao Zhang
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Anqi Lin
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Ziwei Tao
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Yingying Fu
- East China University of Science and Technology Engineering Research Center for Biomaterials of Ministry of Education CHINA
| | - Lan Xiao
- Queensland University of Technology Centre for Biomedical Technologies AUSTRALIA
| | | | - Yulin Li
- East China University of Science and Technology Meilong Road 130 Shanghai CHINA
| |
Collapse
|
12
|
Harvey J, Mellody KT, Cullum N, Watson REB, Dumville J. Wound fluid sampling methods for proteomic studies: A scoping review. Wound Repair Regen 2022; 30:317-333. [PMID: 35381119 PMCID: PMC9322564 DOI: 10.1111/wrr.13009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/01/2022] [Accepted: 03/08/2022] [Indexed: 01/02/2023]
Abstract
Understanding why some wounds are hard to heal is important for improving care and developing more effective treatments. The method of sample collection used is an integral step in the research process and thus may affect the results obtained. The primary objective of this study was to summarise and map the methods currently used to sample wound fluid for protein profiling and analysis. Eligible studies were those that used a sampling method to collect wound fluid from any human wound for analysis of proteins. A search for eligible studies was performed using MEDLINE, Embase and CINAHL Plus in May 2020. All references were screened for eligibility by one reviewer, followed by discussion and consensus with a second reviewer. Quantitative data were mapped and visualised using appropriate software and summarised via a narrative summary. After screening, 280 studies were included in this review. The most commonly used group of wound fluid collection methods were vacuum, drainage or use of other external devices, with surgical wounds being the most common sample source. Other frequently used collection methods were extraction from absorbent materials, collection beneath an occlusive dressing and direct collection of wound fluid. This scoping review highlights the variety of methods used for wound fluid collection. Many studies had small sample sizes and short sample collection periods; these weaknesses have hampered the discovery and validation of novel biomarkers. Future research should aim to assess the reproducibility and feasibility of sampling and analytical methods for use in larger longitudinal studies.
Collapse
Affiliation(s)
- Joe Harvey
- Centre for Dermatology Research, School of Biological SciencesThe University of Manchester & Salford Royal NHS Foundation Trust, Manchester Academic Health Science CentreUK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
| | - Kieran T. Mellody
- Centre for Dermatology Research, School of Biological SciencesThe University of Manchester & Salford Royal NHS Foundation Trust, Manchester Academic Health Science CentreUK
| | - Nicky Cullum
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
- Division of Nursing, Midwifery & Social WorkSchool of Health Sciences, The University of ManchesterManchesterUK
| | - Rachel E. B. Watson
- Centre for Dermatology Research, School of Biological SciencesThe University of Manchester & Salford Royal NHS Foundation Trust, Manchester Academic Health Science CentreUK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
- Manchester Institute for Collaborative Research on AgeingThe University of ManchesterManchesterUK
| | - Jo Dumville
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science CentreManchesterUK
- Division of Nursing, Midwifery & Social WorkSchool of Health Sciences, The University of ManchesterManchesterUK
| |
Collapse
|
13
|
A Review of Alpha-1 Antitrypsin Binding Partners for Immune Regulation and Potential Therapeutic Application. Int J Mol Sci 2022; 23:ijms23052441. [PMID: 35269582 PMCID: PMC8910375 DOI: 10.3390/ijms23052441] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Alpha-1 antitrypsin (AAT) is the canonical serine protease inhibitor of neutrophil-derived proteases and can modulate innate immune mechanisms through its anti-inflammatory activities mediated by a broad spectrum of protein, cytokine, and cell surface interactions. AAT contains a reactive methionine residue that is critical for its protease-specific binding capacity, whereby AAT entraps the protease on cleavage of its reactive centre loop, neutralises its activity by key changes in its tertiary structure, and permits removal of the AAT-protease complex from the circulation. Recently, however, the immunomodulatory role of AAT has come increasingly to the fore with several prominent studies focused on lipid or protein-protein interactions that are predominantly mediated through electrostatic, glycan, or hydrophobic potential binding sites. The aim of this review was to investigate the spectrum of AAT molecular interactions, with newer studies supporting a potential therapeutic paradigm for AAT augmentation therapy in disorders in which a chronic immune response is strongly linked.
Collapse
|
14
|
Issa R, Thompson KL, Price BL. CONTROL OF STAPHYLOCOCCAL-MEDIATED ENDOGENOUS PROTEASE ACTIVITY ALTERS WOUND CLOSURE TIME IN A COMPLEX WOUND MODEL. J Dermatol Sci 2022; 105:105-112. [DOI: 10.1016/j.jdermsci.2022.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 10/19/2022]
|
15
|
Liu Q, Zhang J, Han X, Chen J, Zhai Y, Lin Y, Ma H, Feng F, He X, Li P. Huiyang Shengji decoction promotes wound healing in diabetic mice by activating the EGFR/PI3K/ATK pathway. Chin Med 2021; 16:111. [PMID: 34727961 PMCID: PMC8565039 DOI: 10.1186/s13020-021-00497-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/27/2021] [Indexed: 01/13/2023] Open
Abstract
Background Common chronic wounds include diabetic ulcers, venous ulcers, and pressure ulcers. The traditional Chinese medicine Huiyang Shengji decoction (HYSJD) has been shown to promote the healing of diabetic chronic wounds, however, its pharmacological mechanism is still unclear. Purpose This study aimed to determine the mechanism of HYSJD in promoting the healing of diabetic chronic skin ulcers. Methods Ultra-performance liquid chromatography was combined with tandem mass spectrometry (UPLC-MS/MS) to analyze the main components of HYSJD and the absorbed components in mouse serum at 30 min after oral administration of HYSJD. db/db mouse models for chronic skin ulcers were constructed by full-thickness skin resection. Wound tissues at day 7 post wound formation were used to perform microarray analysis of growth factors and chemokine expression. GO and KEGG enrichment analysis was performed on differentially expressed proteins. ELISA assays were used to measure differential expressed cytokines in the serum and Western blot analysis was used to determine the expression levels of related pathway proteins in the skin wounds. Results UPLC-MS/MS analysis showed that the main chemical components of HYSJD were flavonoids, terpenes, alkaloids, phenylpropanoids, and carbohydrates. At 30 min after oral administration of HYSJD, five absorbed components were detected in the serum, these included formononetin, calycosin, hypaconitine, calycosin-7-glucoside, and sinapic acid. HYSJD was found to increase the wound healing rate in chronic skin ulcers in db/db mice at days 3, 7, and 14 post wound formation, and promote the proliferation of epidermal cells. Two proteins that were differentially expressed between the different groups, i.e., IGF-1 and EGFR, were further validated. Serum ELISA assays showed that serum EGFR in the HYSJD treatment group was significantly increased. KEGG pathway analysis suggested that the PI3K/AKT pathway involved in HYSJD promoting the proliferation of epidermal cells in chronic wounds in db/db mice. Experimental verification showed that HYSJD activated the PI3K/AKT signaling pathway in mouse wound skin. Conclusion HYSJD promotes the proliferation of epidermal cells in chronic diabetic wounds by increasing EGFR expression in the wounds and activating the PI3K/AKT signaling pathway. Our study provides an experimental basis for the pharmacological mechanism of HYSJD. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-021-00497-0.
Collapse
Affiliation(s)
- Qingwu Liu
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Department of Dermatology, China-Japan Friendship Hospital, Beijing, China
| | - Jinchao Zhang
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuyang Han
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jia Chen
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yating Zhai
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yan Lin
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Huike Ma
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Fang Feng
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiujuan He
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Ping Li
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China. .,School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
16
|
Niemann S, Nguyen MT, Eble JA, Chasan AI, Mrakovcic M, Böttcher RT, Preissner KT, Roßlenbroich S, Peters G, Herrmann M. More Is Not Always Better-the Double-Headed Role of Fibronectin in Staphylococcus aureus Host Cell Invasion. mBio 2021; 12:e0106221. [PMID: 34663090 PMCID: PMC8524341 DOI: 10.1128/mbio.01062-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/17/2021] [Indexed: 12/30/2022] Open
Abstract
While Staphylococcus aureus has classically been considered an extracellular pathogen, these bacteria are also capable of being taken up by host cells, including nonprofessional phagocytes such as endothelial cells, epithelial cells, or osteoblasts. The intracellular S. aureus lifestyle contributes to infection development. The predominant recognition and internalization pathway appears to be the binding of the bacteria via a fibronectin bridge to the α5β1-integrin on the host cell membrane, followed by phagocytosis. Although osteoblasts showed high expression of α5β1-integrin and fibronectin, and bacteria adhered to osteoblasts to a high proportion, here we demonstrate by internalization assays and immunofluorescence microscopy that S. aureus was less engulfed in osteoblasts than in epithelial cells. The addition of exogenous fibronectin during the infection of cells with S. aureus resulted in an increased uptake by epithelial cells but not by osteoblasts. This contrasts with the previous conception of the uptake mechanism, where high expression of integrin and fibronectin would promote the bacterial uptake into host cells. Extracellular fibronectin surrounding osteoblasts, but not epithelial cells, is organized in a fibrillary network. The inhibition of fibril formation, the short interfering RNA-mediated reduction of fibronectin expression, and the disruption of the fibronectin-fibril meshwork all resulted in a significant increase in S. aureus uptake by osteoblasts. Thus, the network of fibronectin fibrils appears to strongly reduce the uptake of S. aureus into a given host cell, indicating that the supramolecular structure of fibronectin determines the capacity of particular host cells to internalize the pathogen. IMPORTANCE Traditionally, Staphylococcus aureus has been considered an extracellular pathogen. However, among other factors, the frequent failure of antimicrobial therapy and the ability of the pathogen to cause recurrent disease have established the concept of eukaryotic invasion of the pathogen, thereby evading the host's immune system. In the current model of host cell invasion, bacteria initially bind to α5β1 integrin on the host cell side via a fibronectin bridge, which eventually leads to phagocytosis of S. aureus by host cells. However, in this study, we demonstrate that not the crude amount but the supramolecular structure of fibronectin molecules deposited on the eukaryotic cell surface plays an essential role in bacterial uptake by host cells. Our findings explain the large differences of S. aureus uptake efficacy in different host cell types as well as in vivo differences between courses of bacterial infections and the localization of bacteria in different clinical settings.
Collapse
Affiliation(s)
- Silke Niemann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Minh-Thu Nguyen
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Achmet I. Chasan
- Institute of Immunology, University of Münster, Münster, Germany
| | - Maria Mrakovcic
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
- Institute of Infectiology, University of Münster, Münster, Germany
| | - Ralph T. Böttcher
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, Martinsried, Germany
| | - Klaus T. Preissner
- Kerckhoff-Herzforschungsinstitut, Department of Cardiology, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Steffen Roßlenbroich
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Münster, Münster, Germany
| | - Georg Peters
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
17
|
van der Plas MJA, Cai J, Petrlova J, Saleh K, Kjellström S, Schmidtchen A. Method development and characterisation of the low-molecular-weight peptidome of human wound fluids. eLife 2021; 10:e66876. [PMID: 34227939 PMCID: PMC8260221 DOI: 10.7554/elife.66876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/03/2021] [Indexed: 01/13/2023] Open
Abstract
The normal wound healing process is characterised by proteolytic events, whereas infection results in dysfunctional activations by endogenous and bacterial proteases. Peptides, downstream reporters of these proteolytic actions, could therefore serve as a promising tool for diagnosis of wounds. Using mass-spectrometry analyses, we here for the first time characterise the peptidome of human wound fluids. Sterile post-surgical wound fluids were found to contain a high degree of peptides in comparison to human plasma. Analyses of the peptidome from uninfected healing wounds and Staphylococcus aureus -infected wounds identify unique peptide patterns of various proteins, including coagulation and complement factors, proteases, and antiproteinases. Together, the work defines a workflow for analysis of peptides derived from wound fluids and demonstrates a proof-of-concept that such fluids can be used for analysis of qualitative differences of peptide patterns from larger patient cohorts, providing potential biomarkers for wound healing and infection.
Collapse
Affiliation(s)
- Mariena JA van der Plas
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of CopenhagenCopenhagenDenmark
| | - Jun Cai
- LEO Foundation Center for Cutaneous Drug Delivery, Department of Pharmacy, University of CopenhagenCopenhagenDenmark
| | - Jitka Petrlova
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
| | - Karim Saleh
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
- Dermatology, Skane University HospitalLundSweden
| | - Sven Kjellström
- Division of Mass Spectrometry, Department of Clinical Sciences, Lund UniversityLundSweden
| | - Artur Schmidtchen
- Division of Dermatology and Venereology, Department of Clinical Sciences, Lund UniversityLundSweden
- Dermatology, Skane University HospitalLundSweden
- Copenhagen Wound Healing Center, Bispebjerg Hospital, Department of Biomedical Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
18
|
Strömdahl AC, Ignatowicz L, Petruk G, Butrym M, Wasserstrom S, Schmidtchen A, Puthia M. Peptide-coated polyurethane material reduces wound infection and inflammation. Acta Biomater 2021; 128:314-331. [PMID: 33951491 DOI: 10.1016/j.actbio.2021.04.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 01/24/2023]
Abstract
There is an urgent need for treatments that not only reduce bacterial infection that occurs during wounding but that also target the accompanying excessive inflammatory response. TCP-25, a thrombin-derived antibacterial peptide, scavenges toll-like receptor agonists such as endotoxins and lipoteichoic acid and prevents toll-like receptor-4 dimerization to reduce infection-related inflammation in vivo. Using a combination of biophysical, cellular, and microbiological assays followed by experimental studies in mouse and pig models, we show that TCP-25, when delivered from a polyurethane (PU) material, exerts anti-infective and anti-inflammatory effects in vitro and in vivo. Specifically, TCP-25 killed the common wound pathogens, Pseudomonas aeruginosa and Staphylococcus aureus, in both in vitro and in vivo assays. Furthermore, after its release from the PU material, the peptide retained its capacity to induce its helical conformation upon endotoxin interaction, yielding reduced activation of NF-κB in THP-1 reporter cells, and diminished accumulation of inflammatory cells and subsequent release of IL-6 and TNF-α in subcutaneous implant models in vivo. Moreover, in a porcine partial thickness wound infection model, TCP-25 treated infection with S. aureus, and reduced the concomitant inflammatory response. Taken together, these findings demonstrate a combined antibacterial and anti-inflammatory effect of TCP-25 delivered from PU in vitro, and in mouse and porcine in vivo models of localized infection-inflammation. STATEMENT OF SIGNIFICANCE: Local wound infections may result in systemic complications and can be difficult to treat due to increasing antimicrobial resistance. Surgical site infections and biomaterial-related infections present a major challenge for hospitals. In recent years, various antimicrobial coatings have been developed for infection prevention and current concepts focus on various matrices with added anti-infective components, including various antibiotics and antiseptics. We have developed a dual action wound dressing concept where the host defense peptide TCP-25, when delivered from a PU material, targets both bacterial infection and the accompanying inflammation. TCP-25 PU showed efficacy in in vitro and experimental wound models in mouse and minipigs.
Collapse
|
19
|
Blum C, Taskin MB, Shan J, Schilling T, Schlegelmilch K, Teßmar J, Groll J. Appreciating the First Line of the Human Innate Immune Defense: A Strategy to Model and Alleviate the Neutrophil Elastase-Mediated Attack toward Bioactivated Biomaterials. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007551. [PMID: 33690981 DOI: 10.1002/smll.202007551] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/10/2021] [Indexed: 06/12/2023]
Abstract
Biointerface engineering is a wide-spread strategy to improve the healing process and subsequent tissue integration of biomaterials. Especially the integration of specific peptides is one promising strategy to promote the regenerative capacity of implants and 3D scaffolds. In vivo, these tailored interfaces are, however, first confronted with the innate immune response. Neutrophils are cells with pronounced proteolytic potential and the first recruited immune cells at the implant site; nonetheless, they have so far been underappreciated in the design of biomaterial interfaces. Herein, an in vitro approach is introduced to model and analyze the neutrophil interaction with bioactivated materials at the example of nano-bioinspired electrospun surfaces that reveals the vulnerability of a given biointerface design to the contact with neutrophils. A sacrificial, transient hydrogel coating that demonstrates optimal protection for peptide-modified surfaces and thus alleviates the immediate cleavage by neutrophil elastase is further introduced.
Collapse
Affiliation(s)
- Carina Blum
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Pleicherwall 2, Würzburg, 97070, Germany
| | - Mehmet Berat Taskin
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Pleicherwall 2, Würzburg, 97070, Germany
| | - Junwen Shan
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Pleicherwall 2, Würzburg, 97070, Germany
| | - Tatjana Schilling
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Pleicherwall 2, Würzburg, 97070, Germany
| | - Katrin Schlegelmilch
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Pleicherwall 2, Würzburg, 97070, Germany
| | - Jörg Teßmar
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Pleicherwall 2, Würzburg, 97070, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Pleicherwall 2, Würzburg, 97070, Germany
| |
Collapse
|
20
|
Bergant Suhodolčan A, Luzar B, Kecelj Leskovec N. Matrix metalloproteinase (MMP)-1 and MMP-2, but not COX-2 serve as additional predictors for chronic venous ulcer healing. Wound Repair Regen 2021; 29:725-731. [PMID: 33768649 DOI: 10.1111/wrr.12915] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/03/2021] [Accepted: 03/18/2021] [Indexed: 02/01/2023]
Abstract
Chronic venous ulcers affect 1% of the adult population and are associated with a marked reduction in quality of life, especially if healing is prolonged. Several matrix metalloproteinases (MMPs) appear to be involved in the pathophysiology of chronic venous ulcer healing, but their exact role is still unclear. Cyclooxygenase-2 (COX-2) is an important enzyme in prostanoid synthesis, induced during inflammation in chronic venous ulcer. The first aim of our study was to compare the expression of MMP-1, MMP-2, and COX-2 in wound tissue to that in normal skin. The second aim was to observe the expression of the above factors in 29 chronic venous ulcers in 22 patients at the beginning and 4 weeks later in relation to healing rates and final healing outcome after 24 weeks. The enrolled population was divided into two groups, healed and non-healed wounds after 24 weeks. The intensity of expression of MMP-1, MMP-2 and COX-2 was assessed for each ulcer in paired wound biopsy samples and wound size measurements using laser triangulation at the beginning and after 4 weeks of observation. Initial healing rates in the first 4 weeks were calculated and proved to be an important predictive factor of healing in 24 weeks. Decreases in MMP-1 and MMP-2 after 4 weeks of observation were distinct, positive predictors for ulcer healing. Healing odds were 3.7 times higher for a decrease in MMP-1 and 2.1 times higher for a decrease in MMP-2 compared to the healing odds for a non-decrease in MMP-1 and MMP-2. In conclusion, a decrease in MMP-1 and MMP-2, but not COX-2, in wound biopsy samples after 4 weeks of observation can predict better healing of chronic venous ulcer.
Collapse
Affiliation(s)
- Aleksandra Bergant Suhodolčan
- Department of Dermatovenereology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Boštjan Luzar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | | |
Collapse
|
21
|
Mehrabi T, Mesgar AS, Mohammadi Z. Bioactive Glasses: A Promising Therapeutic Ion Release Strategy for Enhancing Wound Healing. ACS Biomater Sci Eng 2020; 6:5399-5430. [PMID: 33320556 DOI: 10.1021/acsbiomaterials.0c00528] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The morbidity, mortality, and burden of burn victims and patients with severe diabetic wounds are still high, which leads to an extensively growing demand for novel treatments with high clinical efficacy. Biomaterial-based wound treatment approaches have progressed over time from simple cotton wool dressings to advanced skin substitutes containing cells and growth factors; however, no wound care approach is yet completely satisfying. Bioactive glasses are materials with potential in many areas that exhibit unique features in biomedical applications. Today, bioactive glasses are not only amorphous solid structures that can be used as a substitute in hard tissue but also are promising materials for soft tissue regeneration and wound healing applications. Biologically active elements such as Ag, B, Ca, Ce, Co, Cu, Ga, Mg, Se, Sr, and Zn can be incorporated in glass networks; hence, the superiority of these multifunctional materials over current materials results from their ability to release multiple therapeutic ions in the wound environment, which target different stages of the wound healing process. Bioactive glasses and their dissolution products have high potency for inducing angiogenesis and exerting several biological impacts on cell functions, which are involved in wound healing and some other features that are valuable in wound healing applications, namely hemostatic and antibacterial properties. In this review, we focus on skin structure, the dynamic process of wound healing in injured skin, and existing wound care approaches. The basic concepts of bioactive glasses are reviewed to better understand the relationship between glass structure and its properties. We illustrate the active role of bioactive glasses in wound repair and regeneration. Finally, research studies that have used bioactive glasses in wound healing applications are summarized and the future trends in this field are elaborated.
Collapse
Affiliation(s)
- Tina Mehrabi
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Abdorreza S Mesgar
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| | - Zahra Mohammadi
- Biomaterials Laboratory, Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran 1439957131, Iran
| |
Collapse
|
22
|
Abstract
Fibronectin (FN) circulating in the blood and produced by cells provides the basis of the extracellular matrix (ECM) formed in healing acute wounds. The time-dependent deposition of FN by macrophages, its synthesis by fibroblasts and myofibroblasts, and later degradation in the remodeled granulation tissue are a prerequisite for successful healing of wounds. However, the pattern of FN expression and deposition in skin lesions is disturbed. The degradation of the ECM components including FN in varicose veins prevails over ECM synthesis and deposition. FN is inconspicuous in the fibrotic lesions in lipodermatosclerosis, while tenascin-C containing FN-like peptide sequences are prominent. FN is produced in large amounts by fibroblasts at the edge of venous ulcers but FN deposition at the wound bed is impaired. Both the proteolytic environment in the wounds and the changed function of the ulcer fibroblasts may be responsible for the poor healing of venous ulcers. The aim of this review is to describe the current knowledge of FN pathophysiology in chronic venous diseases. In view of the fact that FN plays a crucial role in organizing the ECM, further research focused on FN metabolism in venous diseases may bring results applicable to the treatment of the diseases.
Collapse
Affiliation(s)
- Jiri Kanta
- Faculty of Medicine, Charles University, Hradec Kralove, Czech Republic
| | - Anna Zavadakova
- Biomedical Center, Faculty of Medicine, Charles University, Pilsen, Czech Republic
| |
Collapse
|
23
|
Yang T, Pan SC, Cheng CM. Paper-based human neutrophil elastase detection device for clinical wound monitoring. LAB ON A CHIP 2020; 20:2709-2716. [PMID: 32573571 DOI: 10.1039/d0lc00062k] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Paper-based diagnostic devices have been widely applied to assess the presence and status of a variety of clinical diseases by analyzing samples such as urine or blood. Due to their low cost, user-friendliness, and convenience, they have been used as point-of-care (POC) devices in countries lacking resources or energy. Despite wide-ranging research and implementation, paper-based devices have not previously been developed for wound analysis. Here, we discuss the successful development of such a tool to facilitate simple and rapid wound status assessment. The purpose of this study was to develop a paper-based elastase detection device (PEDD) for clinical wound assessment that specifically examines human neutrophil elastase (HNE), one of the most abundant serine proteases found in chronic wounds. The first step in this study was an examination of different paper substrate types (i.e., chromatography paper and filter paper) to determine which provided the best protease immobilization and colorimetric response. We then used a wax printing approach to create hydrophobic and hydrophilic regions and designated test zones created on both chromatography and filter papers. This allowed us to physically immobilize both substrate and protease within the desired test zone regions. This PEDD which demonstrated good sensitivity (0.631 μg mL-1, in a wound fluid system) can be used to monitor protease activity expressed in wounds. After developing this device, we examined samples from 9 patients presenting a total of 7 acute and 4 chronic wounds to determine wound HNE concentration. We believe that this study may be widely applicable in both academic and commercial sciences, including the development of practical POC detection devices.
Collapse
Affiliation(s)
- Ting Yang
- Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| | | | | |
Collapse
|
24
|
Abstract
Chronic wounds present a unique therapeutic challenge to heal. Chronic wounds are colonized with bacteria and the presence of a biofilm that further inhibits the normal wound healing processes, and are locked into a very damaging proinflammatory response. The treatment of chronic wounds requires a coordinated approach, including debridement of devitalized tissue, minimizing bacteria and biofilm, control of inflammation, and the use of specialized dressings to address the specific aspects of the particular nonhealing ulcer.
Collapse
|
25
|
Baldan-Martin M, Martin-Rojas T, Corbacho-Alonso N, Lopez JA, Sastre-Oliva T, Gil-Dones F, Vazquez J, Arevalo JM, Mourino-Alvarez L, Barderas MG. Comprehensive Proteomic Profiling of Pressure Ulcers in Patients with Spinal Cord Injury Identifies a Specific Protein Pattern of Pathology. Adv Wound Care (New Rochelle) 2020; 9:277-294. [PMID: 32226651 PMCID: PMC7099418 DOI: 10.1089/wound.2019.0968] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/10/2019] [Indexed: 12/26/2022] Open
Abstract
Objective: Severe pressure ulcers (PUs) do not respond to conservative wound therapy and need surgical repair. To better understand the pathogenesis and to advance on new therapeutic options, we focused on the proteomic analysis of PU, which offers substantial opportunities to identify significant changes in protein abundance during the course of PU formation in an unbiased manner. Approach: To better define the protein pattern of this pathology, we performed a proteomic approach in which we compare severe PU tissue from spinal cord injury (SCI) patients with control tissue from the same patients. Results: We found 76 proteins with difference in abundance. Of these, 10 proteins were verified as proteins that define the pathology: antithrombin-III, alpha-1-antitrypsin, kininogen-1, alpha-2-macroglobulin, fibronectin, apolipoprotein A-I, collagen alpha-1 (XII) chain, haptoglobin, apolipoprotein B-100, and complement factor B. Innovation: This is the first study to analyze differential abundance protein of PU tissue from SCI patients using high-throughput protein identification and quantification by tandem mass tags followed by liquid chromatography tandem mass spectrometry. Conclusion: Differential abundance proteins are mainly involved in tissue regeneration. These proteins might be considered as future therapeutic options to enhance the physiological response and permit cellular repair of damaged tissue.
Collapse
Affiliation(s)
- Montserrat Baldan-Martin
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Tatiana Martin-Rojas
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Nerea Corbacho-Alonso
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Juan Antonio Lopez
- Department of Plastic Surgery, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Tamara Sastre-Oliva
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Felix Gil-Dones
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Jesus Vazquez
- Department of Plastic Surgery, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | | | - Laura Mourino-Alvarez
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| | - Maria G. Barderas
- Department of Vascular Physiopathology, National Hospital for Paraplegics (HNP), SESCAM, Toledo, Spain
| |
Collapse
|
26
|
Norris EG, Dalecki D, Hocking DC. Acoustic Fabrication of Collagen-Fibronectin Composite Gels Accelerates Microtissue Formation. APPLIED SCIENCES (BASEL, SWITZERLAND) 2020; 10:2907. [PMID: 33604063 PMCID: PMC7889010 DOI: 10.3390/app10082907] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ultrasound can influence biological systems through several distinct acoustic mechanisms that can be manipulated by varying reaction conditions and acoustic exposure parameters. We recently reported a new ultrasound-based fabrication technology that exploits the ability of ultrasound to generate localized mechanical forces and thermal effects to control collagen fiber microstructure non-invasively. Exposing solutions of type I collagen to ultrasound during the period of microfibril assembly produced changes in collagen fiber structure and alignment, and increased the biological activity of the resultant collagen hydrogels. In the extracellular matrix, interactions between fibronectin and collagen fibrils influence the biological activity of both proteins. Thus, in the present study, we examined how addition of fibronectin to collagen solutions prior to ultrasound exposure affects protein organization and the biological activity of the composite hydrogels. Results indicate that ultrasound can alter the distribution of fibronectin within 3D hydrogels via thermal and non-thermal mechanisms to produce composite hydrogels that support accelerated microtissue formation. The use of acoustic energy to drive changes in protein conformation to functionalize biomaterials has much potential as a unique, non-invasive technology for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Emma G. Norris
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
| | - Diane Dalecki
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Denise C. Hocking
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
27
|
McDaniel JC, Rausch J, Tan A. Impact of omega-3 fatty acid oral therapy on healing of chronic venous leg ulcers in older adults: Study protocol for a randomized controlled single-center trial. Trials 2020; 21:93. [PMID: 31948466 PMCID: PMC6966808 DOI: 10.1186/s13063-019-3970-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/06/2019] [Indexed: 12/15/2022] Open
Abstract
Background This trial addresses the global problem of chronic venous leg ulcers (CVLUs), wounds that cause significant infirmity for an estimated 9.7 million people annually, mainly older adults with comorbidities. Advanced therapies are needed because standard topical therapies are often ineffective or yield only short-term wound healing. Thus, we are testing a new oral therapy containing the bioactive elements of fish oil, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), for targeting and reducing the high numbers of activated polymorphonuclear leukocytes (PMN) in wound microenvironments that keep CVLUs “trapped” in a chronic inflammatory state. Methods This double-blind RCT will include 248 eligible adults ≥ 55 years of age with CVLUs receiving standard care at a large Midwest outpatient wound clinic. Participants are randomized to two groups: 12 weeks of daily oral therapy with EPA + DHA (1.87 g/day of EPA + 1.0 g/day of DHA) or daily oral therapy with placebo. At 0, 4, 8, and 12 weeks, across the two groups, we are pursuing three specific aims: Aim 1. Compare levels of EPA + DHA-derived lipid mediators, and inflammatory cytokines in blood and wound fluid; Subaim 1a. Compare inflammatory cytokine gene expression by PMNs in blood; Aim 2. Compare PMN activation in blood and wound fluid, and PMN-derived protease levels in wound fluid; Aim 3. Compare reduction in wound area, controlling for factors known to impact healing, and determine relationships with lipid mediators, cytokines, and PMN activation. Subaim 3a. Compare frequency of CVLU recurrence and levels of study variables in blood between the randomly assigned two subgroups (continuing EPA + DHA therapy versus placebo therapy beyond week 12) within the EPA + DHA group with healed CVLUs after 3 months of therapy. Subaim 3b. Compare symptoms of pain at all time points and quality of life at first and last time points across the two groups and two subgroups. Discussion This trial will provide new evidence about the effectiveness of EPA + DHA oral therapy to target and reduce excessive PMN activation systemically and locally in patients with CVLUs. If effective, this therapy may facilitate healing and thus be a new adjunct treatment for CVLUs in the aging population. Trial registration ClinicalTrials.gov, NCT03576989; Registered on 13 June 2018.
Collapse
Affiliation(s)
- Jodi C McDaniel
- College of Nursing, The Ohio State University, 372 Newton Hall, 1585 Neil Avenue, Columbus, OH, 43210-1289, USA.
| | - Jamie Rausch
- College of Nursing, The Ohio State University, 372 Newton Hall, 1585 Neil Avenue, Columbus, OH, 43210-1289, USA
| | - Alai Tan
- College of Nursing, The Ohio State University, 372 Newton Hall, 1585 Neil Avenue, Columbus, OH, 43210-1289, USA
| |
Collapse
|
28
|
Saleh K, Strömdahl AC, Riesbeck K, Schmidtchen A. Inflammation Biomarkers and Correlation to Wound Status After Full-Thickness Skin Grafting. Front Med (Lausanne) 2019; 6:159. [PMID: 31355202 PMCID: PMC6640317 DOI: 10.3389/fmed.2019.00159] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 06/26/2019] [Indexed: 12/21/2022] Open
Abstract
Background: A surgical site infection (SSI) is believed to be the result of an exaggerated inflammatory response. Objective: Examine the relationship between clinical status and inflammation biomarkers in full-thickness skin grafting wounds. Methods: Twenty patients planned for facial full-thickness skin grafting were enrolled. A week after surgery, all graft wounds were clinically assessed using a 3-step scale for inflammation (low, moderate, high). All wounds were swabbed for routine microbiological analysis and assessment of numbers of aerobic bacteria. Tie-over dressings from all patients were collected and used for wound fluid extraction and subsequent analysis of MMPs, cytokines, and NF-κB inducing activity. Results: Wounds with a high degree of inflammation contained increased total MMP activity (P ≤ 0.05) in their corresponding fluids. Likewise, the level of the cytokines IL-1ß, IL-8, IL-6, TNF-α was analyzed, and particularly IL-1ß was discriminatory for highly inflamed wounds (P ≤ 0.01). Moreover, bacterial loads were increased in highly inflamed wounds compared to wounds with a low degree of inflammation (P ≤ 0.01). NF-κB activation in the monocytic cell line THP-1 was significantly higher when these cells were stimulated by wound fluids with a high degree of inflammation (P ≤ 0.01). Growth of S. aureus in wounds did not vary between wounds with different degrees of inflammation (chi-square 3.8, P = 0.144). Conclusion: Biomarkers analyzed from tie-over dressings correlated to clinical wound healing in full-thickness skin grafting.
Collapse
Affiliation(s)
- Karim Saleh
- Division of Dermatology, Department of Clinical Sciences, Skane University Hospital, Lund University, Lund, Sweden
| | - Ann-Charlotte Strömdahl
- Division of Dermatology, Department of Clinical Sciences, Skane University Hospital, Lund University, Lund, Sweden
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Artur Schmidtchen
- Division of Dermatology, Department of Clinical Sciences, Skane University Hospital, Lund University, Lund, Sweden.,Department of Biomedical Sciences, Copenhagen Wound Healing Center, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Abstract
Cells need to be anchored to extracellular matrix (ECM) to survive, yet the role of ECM in guiding developmental processes, tissue homeostasis, and aging has long been underestimated. How ECM orchestrates the deterioration of healthy to pathological tissues, including fibrosis and cancer, also remains poorly understood. Inquiring how alterations in ECM fiber tension might drive these processes is timely, as mechanobiology is a rapidly growing field, and many novel mechanisms behind the mechanical forces that can regulate protein, cell, and tissue functions have recently been deciphered. The goal of this article is to review how forces can switch protein functions, and thus cell signaling, and thereby inspire new approaches to exploit the mechanobiology of ECM in regenerative medicine as well as for diagnostic and therapeutic applications. Some of the mechanochemical switching concepts described here for ECM proteins are more general and apply to intracellular proteins as well.
Collapse
Affiliation(s)
- Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department for Health Sciences and Technology, ETH Zürich, CH-8093 Zürich, Switzerland;
| |
Collapse
|
30
|
Tomic-Canic M, Wong LL, Smola H. The epithelialisation phase in wound healing: options to enhance wound closure. J Wound Care 2018; 27:646-658. [DOI: 10.12968/jowc.2018.27.10.646] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Majana Tomic-Canic
- Professor and Vice Chair of Research; Director, Wound Healing and Regenerative Medicine Research Program; Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, Florida, US
| | - Lulu L. Wong
- MD Candidate; Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, Florida, US
| | - Hans Smola
- Professor of Dermatology, Medical Director, PAUL HARTMANN AG, Heidenheim and Department of Dermatology, University of Cologne, Cologne, Germany
| |
Collapse
|
31
|
Abstract
Purpose of Review Chronic wounds are a tremendous burden on the healthcare system and lead to significant patient morbidity and mortality. Normal cutaneous wound healing occurs through an intricate and delicate interplay between the immune system, keratinocytes, and dermal cells. Each cell type contributes signals that drive the normal phases of wound healing: hemostasis, inflammation, proliferation, and remodeling. This paper reviews how various immunological cell types and signaling molecules influence the way wounds develop, persist, and heal. Recent Findings Concurrent with the achievement of hemostasis, neutrophils are the first cells to migrate to the wound bed, brought in by pro-inflammatory signals including IL-8. Their apoptosis and engulfment by macrophages (efferocytosis) provides a key signal to the local immune milieu, including macrophages, to transition to an anti-inflammatory, pro-repair state, where angiogenesis occurs and granulation tissue is laid down. Myofibroblasts, activated through contractile forces and signaling molecules, then drive remodeling, where granulation tissue becomes scar. Unchecked inflammation at this stage can result in abnormal scar formation. Summary Although the derangement of immune signals at any stage can result in impaired wound healing, recent research has shown that the key transition point lies between the inflammatory and the proliferative phases. This review summarizes the events that facilitate this transition and discusses how this process can be disrupted, leading to chronic, non-healing wounds.
Collapse
|
32
|
Larouche J, Sheoran S, Maruyama K, Martino MM. Immune Regulation of Skin Wound Healing: Mechanisms and Novel Therapeutic Targets. Adv Wound Care (New Rochelle) 2018; 7:209-231. [PMID: 29984112 PMCID: PMC6032665 DOI: 10.1089/wound.2017.0761] [Citation(s) in RCA: 358] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022] Open
Abstract
Significance: The immune system plays a central role in orchestrating the tissue healing process. Hence, controlling the immune system to promote tissue repair and regeneration is an attractive approach when designing regenerative strategies. This review discusses the pathophysiology of both acute and chronic wounds and possible strategies to control the immune system to accelerate chronic wound closure and promote skin regeneration (scar-less healing) of acute wounds. Recent Advances: Recent studies have revealed the key roles of various immune cells and immune mediators in skin repair. Thus, immune components have been targeted to promote chronic wound repair or skin regeneration and several growth factors, cytokines, and biomaterials have shown promising results in animal models. However, these novel strategies are often struggling to meet efficacy standards in clinical trials, partly due to inadequate drug delivery systems and safety concerns. Critical Issues: Excess inflammation is a major culprit in the dysregulation of normal wound healing, and further limiting inflammation effectively reduces scarring. However, current knowledge is insufficient to efficiently control inflammation and specific immune cells. This is further complicated by inadequate drug delivery methods. Future Directions: Improving our understanding of the molecular pathways through which the immune system controls the wound healing process could facilitate the design of novel regenerative therapies. Additionally, better delivery systems may make current and future therapies more effective. To promote the entry of current regenerative strategies into clinical trials, more evidence on their safety, efficacy, and cost-effectiveness is also needed.
Collapse
Affiliation(s)
- Jacqueline Larouche
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | - Sumit Sheoran
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | - Kenta Maruyama
- WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mikaël M. Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| |
Collapse
|
33
|
Tati R, Nordin S, Abdillahi SM, Mörgelin M. Biological wound matrices with native dermis-like collagen efficiently modulate protease activity. J Wound Care 2018; 27:199-209. [DOI: 10.12968/jowc.2018.27.4.199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Ramesh Tati
- Postdoctoral Researcher, Department of Clinical Sciences, Division of Infection Medicine, Lund University, SE-221 84 Lund, Sweden
| | - Sara Nordin
- Postdoctoral Researcher, Department of Clinical Sciences, Division of Infection Medicine, Lund University, SE-221 84 Lund, Sweden
| | - Suado M Abdillahi
- Postdoctoral Researcher, Department of Clinical Sciences, Division of Infection Medicine, Lund University, SE-221 84 Lund, Sweden
| | - Matthias Mörgelin
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, SE-221 84 Lund, Sweden, Colzyx ltd, Medicon Village, Scheelevägen 2, SE-223 81 Lund, Sweden
| |
Collapse
|
34
|
Stavrou EX, Fang C, Bane KL, Long AT, Naudin C, Kucukal E, Gandhi A, Brett-Morris A, Mumaw MM, Izadmehr S, Merkulova A, Reynolds CC, Alhalabi O, Nayak L, Yu WM, Qu CK, Meyerson HJ, Dubyak GR, Gurkan UA, Nieman MT, Sen Gupta A, Renné T, Schmaier AH. Factor XII and uPAR upregulate neutrophil functions to influence wound healing. J Clin Invest 2018; 128:944-959. [PMID: 29376892 PMCID: PMC5824869 DOI: 10.1172/jci92880] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 12/14/2017] [Indexed: 01/13/2023] Open
Abstract
Coagulation factor XII (FXII) deficiency is associated with decreased neutrophil migration, but the mechanisms remain uncharacterized. Here, we examine how FXII contributes to the inflammatory response. In 2 models of sterile inflammation, FXII-deficient mice (F12-/-) had fewer neutrophils recruited than WT mice. We discovered that neutrophils produced a pool of FXII that is functionally distinct from hepatic-derived FXII and contributes to neutrophil trafficking at sites of inflammation. FXII signals in neutrophils through urokinase plasminogen activator receptor-mediated (uPAR-mediated) Akt2 phosphorylation at S474 (pAktS474). Downstream of pAkt2S474, FXII stimulation of neutrophils upregulated surface expression of αMβ2 integrin, increased intracellular calcium, and promoted extracellular DNA release. The sum of these activities contributed to neutrophil cell adhesion, migration, and release of neutrophil extracellular traps in a process called NETosis. Decreased neutrophil signaling in F12-/- mice resulted in less inflammation and faster wound healing. Targeting hepatic F12 with siRNA did not affect neutrophil migration, whereas WT BM transplanted into F12-/- hosts was sufficient to correct the neutrophil migration defect in F12-/- mice and restore wound inflammation. Importantly, these activities were a zymogen FXII function and independent of FXIIa and contact activation, highlighting that FXII has a sophisticated role in vivo that has not been previously appreciated.
Collapse
Affiliation(s)
- Evi X. Stavrou
- Department of Medicine, Louis Stokes Veterans Administration Medical Center, Cleveland, Ohio, USA
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Chao Fang
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Kara L. Bane
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Andy T. Long
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Clément Naudin
- Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Erdem Kucukal
- Department of Mechanical and Aerospace Engineering, CWRU, Cleveland, Ohio, USA
| | - Agharnan Gandhi
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Adina Brett-Morris
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Michele M. Mumaw
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Sudeh Izadmehr
- Department of Genetics and Genomics Sciences, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Alona Merkulova
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Cindy C. Reynolds
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Omar Alhalabi
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Lalitha Nayak
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Wen-Mei Yu
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | - Cheng-Kui Qu
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
| | | | | | - Umut A. Gurkan
- Department of Mechanical and Aerospace Engineering, CWRU, Cleveland, Ohio, USA
| | | | | | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Alvin H. Schmaier
- Department of Medicine, Hematology and Oncology Division, Case Western Reserve University (CWRU) School of Medicine, Cleveland, Ohio, USA
- Department of Medicine, Hematology and Oncology Division, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
35
|
Dimatteo R, Darling NJ, Segura T. In situ forming injectable hydrogels for drug delivery and wound repair. Adv Drug Deliv Rev 2018; 127:167-184. [PMID: 29567395 PMCID: PMC6003852 DOI: 10.1016/j.addr.2018.03.007] [Citation(s) in RCA: 529] [Impact Index Per Article: 75.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/18/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
Hydrogels have been utilized in regenerative applications for many decades because of their biocompatibility and similarity in structure to the native extracellular matrix. Initially, these materials were formed outside of the patient and implanted using invasive surgical techniques. However, advances in synthetic chemistry and materials science have now provided researchers with a library of techniques whereby hydrogel formation can occur in situ upon delivery through standard needles. This provides an avenue to minimally invasively deliver therapeutic payloads, fill complex tissue defects, and induce the regeneration of damaged portions of the body. In this review, we highlight these injectable therapeutic hydrogel biomaterials in the context of drug delivery and tissue regeneration for skin wound repair.
Collapse
Affiliation(s)
- Robert Dimatteo
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States.
| | - Nicole J Darling
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States.
| | - Tatiana Segura
- Department of Chemical and Biomolecular Engineering, Bioengineering, and Dermatology, School of Medicine, University of California Los Angeles, 420 Westwood Plaza, Los Angeles, CA 90095, United States.
| |
Collapse
|
36
|
Longo V, Rebulla P, Pupella S, Zolla L, Rinalducci S. Proteomic characterization of platelet gel releasate from adult peripheral and cord blood. Proteomics Clin Appl 2017; 10:870-82. [PMID: 27377258 DOI: 10.1002/prca.201500126] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/20/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022]
Abstract
PURPOSE Activated platelet gel (PG) derived from adult peripheral blood (APB) has been extensively used for topical therapy of various clinical conditions. Conversely, few observations on PG from umbilical cord blood (CB) have been reported so far. Although PG preparations are known to contain a high concentration of a large number of biological factors involved in inflammation and tissue repair, their comprehensive characterization is still missing. The innovative goal of our research was to use proteomics technologies in order to profile biologically active components in these blood derivatives. EXPERIMENTAL DESIGN Supernatants recovered from three independent APB and CB-derived PGs, prepared using batroxobin, were enriched for low-abundance proteins with ProteoMiner and subsequently analyzed by GeLC-MS/MS. RESULTS The 751 and 760 proteins were identified in the APB and CB-derived PG releasates, respectively. A core dataset including only proteins found in 2/3 and 3/3 biological replicates was generated and functionally characterized by gene ontology. Searching against Vesiclepedia database showed that 33% of our dataset consists of novel releasate proteins. Comparison between the two types of PG secretomes revealed that 117 proteins are present only in the APB-derived samples, 104 proteins are distinctive of the CB-derived samples, and 229 are in common. CONCLUSION AND CLINICAL RELEVANCE Our study highlighted a differential content of proteins supporting tissue repair and regeneration between APB and CB-derived PGs. These findings may help better identifying future appropriate clinical applications.
Collapse
Affiliation(s)
- Valentina Longo
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| | - Paolo Rebulla
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simonetta Pupella
- Italian National Blood Centre, National Institute of Health, Rome, Italy
| | - Lello Zolla
- Department of Science and Technology for Agriculture, Forestry, Nature and Energy (DAFNE), University of Tuscia, Viterbo, Italy
| | - Sara Rinalducci
- Department of Ecological and Biological Sciences (DEB), University of Tuscia, Viterbo, Italy
| |
Collapse
|
37
|
McDaniel JC, Szalacha L, Sales M, Roy S, Chafee S, Parinandi N. EPA + DHA supplementation reduces PMN activation in microenvironment of chronic venous leg ulcers: A randomized, double-blind, controlled study. Wound Repair Regen 2017; 25:680-690. [DOI: 10.1111/wrr.12558] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022]
Affiliation(s)
| | - Laura Szalacha
- College of Nursing, University of Arizona; Tucson Arizona
| | - Michelle Sales
- College of Nursing, The Ohio State University; Columbus Ohio
| | - Sashwati Roy
- College of Medicine; The Ohio State University; Columbus Ohio
- Davis Heart and Lung Research Institute; The Ohio State University; Columbus Ohio
| | - Scott Chafee
- College of Medicine; The Ohio State University; Columbus Ohio
- Davis Heart and Lung Research Institute; The Ohio State University; Columbus Ohio
| | - Narasimham Parinandi
- College of Medicine; The Ohio State University; Columbus Ohio
- Davis Heart and Lung Research Institute; The Ohio State University; Columbus Ohio
| |
Collapse
|
38
|
Merkulova Y, Shen Y, Parkinson LG, Raithatha SA, Zhao H, Westendorf K, Sharma M, Bleackley RC, Granville DJ. Granzyme B inhibits keratinocyte migration by disrupting epidermal growth factor receptor (EGFR)-mediated signaling. Biol Chem 2017; 397:883-95. [PMID: 27060743 DOI: 10.1515/hsz-2016-0129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 03/25/2016] [Indexed: 12/29/2022]
Abstract
Chronic non-healing wounds including diabetic, venous, and decubitus skin ulcers are currently lacking effective therapies. Non-healing diabetic ulcers can lead to amputations as progress into a highly chronic state before detection and existing treatments for these wounds often fail. Granzyme B (GzmB) is a serine protease that was, until recently, believed to function exclusively in cytotoxic lymphocyte-mediated apoptosis. However, during excessive or chronic inflammation, GzmB can accumulate in the extracellular milieu, retain its activity, and cleave a number of important extracellular proteins. Epidermal growth factor receptor (EGFR) is a transmembrane receptor involved in cellular processes such as proliferation and migration. EGFR signaling is integral to the wound healing process. The present study investigated the effects of GzmB on keratinocyte cell migration using HaCaT cell line. Using electric cell-substrate impedance sensing and scratch assays, the present study demonstrates that GzmB inhibits keratinocyte migration by interfering with the EGFR pathway. GzmB limited cell transition into a migratory morphology and was found to reduce ligand-induced EGFR phosphorylation. Inhibition of GzmB reversed the aforementioned effects. In summary, data from the present study suggest key role for GzmB in the pathogenesis of impaired wound healing through the impairment of EGFR signaling and cell migration.
Collapse
|
39
|
Lauridsen HM, Pellowe AS, Ramanathan A, Liu R, Miller-Jensen K, McNiff JM, Pober JS, Gonzalez AL. Tumor Necrosis Factor-α and IL-17A Activation Induces Pericyte-Mediated Basement Membrane Remodeling in Human Neutrophilic Dermatoses. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1893-1906. [PMID: 28609645 DOI: 10.1016/j.ajpath.2017.04.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/03/2017] [Accepted: 04/24/2017] [Indexed: 12/11/2022]
Abstract
Sweet syndrome (SS) is a prototypical neutrophilic dermatosis, a class of inflammatory diseases marked by elevated levels of tumor necrosis factor (TNF)-α and IL-17A, pathologic neutrophil recruitment, and microvascular remodeling. Histologic analyses of four matrix proteins-collagen I and IV, laminin, and fibronectin-in skin biopsies of patients with SS reveal that the basement membrane of dermal postcapillary venules undergoes changes in structure and composition. Increased neutrophil recruitment in vivo was associated with increases in collagen IV, decreases in laminin, and varied changes in fibronectin. In vitro studies using TNF-α and IL-17A were conducted to dissect basement membrane remodeling. Prolonged dual activation of cultured human pericytes with TNF-α and IL-17A augmented collagen IV production, similar to in vivo remodeling. Co-activation of pericytes with TNF-α and IL-17A also elevated fibronectin levels with little direct effect on laminin. However, the expression of fibronectin- and laminin-specific matrix metalloproteinases (MMPs), particularly MMP-3, was significantly up-regulated. Interactions between pericytes and neutrophils in culture yielded even higher levels of active MMPs, facilitating fibronectin and laminin degradation, and likely contributing to the varied levels of detectable fibronectin and the decreases in laminin observed in vivo. These data indicate that pericyte-neutrophil interactions play a role in mediating microvascular changes in SS and suggest that targeting MMP-3 may be effective in protecting vascular wall integrity.
Collapse
Affiliation(s)
- Holly M Lauridsen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Amanda S Pellowe
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Anand Ramanathan
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Rebecca Liu
- Department of Immunobiology, Yale University, New Haven, Connecticut
| | | | - Jennifer M McNiff
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Jordan S Pober
- Department of Immunobiology, Yale University, New Haven, Connecticut; Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Anjelica L Gonzalez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut.
| |
Collapse
|
40
|
Topical Negative Pressure on Burns: An Innovative Method for Wound Exudate Collection. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2016; 4:e1117. [PMID: 27975022 PMCID: PMC5142488 DOI: 10.1097/gox.0000000000001117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/13/2016] [Indexed: 12/16/2022]
Abstract
Supplemental Digital Content is available in the text. Burn wound exudate is an important source of information on the wound-healing process and systemic improvement of burn patients. Identification of biomarkers for the prediagnosis of local or systemic complications in patients will have a great impact on adapting personalized procedures in burn treatment. No efficient exudate collection method exists that offers a direct and continuous collection over time. We developed an innovative system based on the negative pressure wound therapy technique to directly collect exudate from burn wounds over several days after burn. This method did not cause any complication or pain for patients, and positive influence on wound healing was seen. Exudate samples were further used in different projects for studying biochemical profile, trace element content, kinetics of bacterial growth, and cell cytotoxicity.
Collapse
|
41
|
The neglected role of copper ions in wound healing. J Inorg Biochem 2016; 161:1-8. [DOI: 10.1016/j.jinorgbio.2016.02.012] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/19/2016] [Accepted: 02/10/2016] [Indexed: 12/30/2022]
|
42
|
Förster Y, Schmidt JR, Wissenbach DK, Pfeiffer SEM, Baumann S, Hofbauer LC, von Bergen M, Kalkhof S, Rammelt S. Microdialysis Sampling from Wound Fluids Enables Quantitative Assessment of Cytokines, Proteins, and Metabolites Reveals Bone Defect-Specific Molecular Profiles. PLoS One 2016; 11:e0159580. [PMID: 27441377 PMCID: PMC4956113 DOI: 10.1371/journal.pone.0159580] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/04/2016] [Indexed: 12/16/2022] Open
Abstract
Bone healing involves a variety of different cell types and biological processes. Although certain key molecules have been identified, the molecular interactions of the healing progress are not completely understood. Moreover, a clinical routine for predicting the quality of bone healing after a fracture in an early phase is missing. This is mainly due to a lack of techniques to comprehensively screen for cytokines, growth factors and metabolites at their local site of action. Since all soluble molecules of interest are present in the fracture hematoma, its in-depth assessment could reveal potential markers for the monitoring of bone healing. Here, we describe an approach for sampling and quantification of cytokines and metabolites by using microdialysis, combined with solid phase extractions of proteins from wound fluids. By using a control group with an isolated soft tissue wound, we could reveal several bone defect-specific molecular features. In bone defect dialysates the neutrophil chemoattractants CXCL1, CXCL2 and CXCL3 were quantified with either a higher or earlier response compared to dialysate from soft tissue wound. Moreover, by analyzing downstream adaptions of the cells on protein level and focusing on early immune response, several proteins involved in the immune cell migration and activity could be identified to be specific for the bone defect group, e.g. immune modulators, proteases and their corresponding inhibitors. Additionally, the metabolite screening revealed different profiles between the bone defect group and the control group. In summary, we identified potential biomarkers to indicate imbalanced healing progress on all levels of analysis.
Collapse
Affiliation(s)
- Yvonne Förster
- University Center of Orthopedics and Trauma Surgery and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital “Carl Gustav Carus”, TU Dresden, Dresden, Germany
- * E-mail:
| | - Johannes R. Schmidt
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Dirk K. Wissenbach
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Susanne E. M. Pfeiffer
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
| | - Sven Baumann
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
- Institute of Pharmacy, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
| | - Lorenz C. Hofbauer
- Division of Endocrinology and Metabolic Bone Diseases, Department of Medicine III, University Hospital “Carl Gustav Carus”, TU Dresden, Dresden, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
- Centre for Microbial Communities, University of Aalborg, Aalborg East, Denmark
| | - Stefan Kalkhof
- Department of Molecular Systems Biology, Helmholtz-Centre for Environmental Research - UFZ, Leipzig, Germany
- Department of Bioanalytics, University of Applied Sciences and Arts of Coburg, Coburg, Germany
| | - Stefan Rammelt
- University Center of Orthopedics and Trauma Surgery and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital “Carl Gustav Carus”, TU Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| |
Collapse
|
43
|
Zollino I, Zuolo M, Gianesini S, Pedriali M, Sibilla MG, Tessari M, Carinci F, Occhionorelli S, Zamboni P. Autologous adipose-derived stem cells: Basic science, technique, and rationale for application in ulcer and wound healing. Phlebology 2016; 32:160-171. [PMID: 27056621 DOI: 10.1177/0268355516641546] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Objectives The present review represents a translational boundary between basic research and surgery, particularly focusing on the promising application of adipose-derived stem cells harvested intra-operatively during debridement of venous leg ulcers. Methods We reviewed 830 out of 5578 articles on MEDLINE starting from 1997 and sorted by the relevance option. Results The technique currently used for adipose-derived stem cells intra-operative harvesting is presented, including a safety evaluation on a cohort of 5089 revised patients who underwent plastic surgery and maxillo-facial surgical procedures. Complications were reported in 169 cases (3.3%). One hundred and forty-one (2.77%) patients were classified as having minor complications, specifically: nodularity/induration 93 (1.83%), dysesthesia 14 (0.26%), hematoma 12 (0.23%), superficial infection 11 (0.21%), pain 7 (0.13%), poor cosmesis 3 (0.06%), and abnormal breast secretion 1 (0.02%), while 28 patients (0.55%) were classified as having major complications, specifically: deep infection 22 (0.43%), sepsis 3 (0.06%), abdominal hematoma 2 (0.04%), and pneumothorax 1 (0.02%). Application of cell therapy in venous leg ulcer is currently used only for patients not responding to the standard treatment. The review shows the lack of randomized clinical trials for application of adipose-derived stem cells among treatments for venous leg ulcer. Finally, adipose-derived stem cells implantation at the wound site promotes a new tissue formation rich in vascular structures and remodeling collagen. Conclusion Adipose-derived stem cells strategy represents a great opportunity for the treatment of chronic wounds, due to the simplicity of the technique and the application of cell treatment in the operating room immediately following debridement. However, clinical studies and data from randomized trials are currently lacking.
Collapse
Affiliation(s)
- Ilaria Zollino
- 1 Department of Morphology, Surgery and Experimental Medicine, Section of Translational of Medicine and Surgery, University of Ferrara, Italy
| | - Michele Zuolo
- 2 Unit of Translational Surgery and Vascular Diseases Center, Sant'Anna University Hospital, Ferrara, Italy
| | - Sergio Gianesini
- 1 Department of Morphology, Surgery and Experimental Medicine, Section of Translational of Medicine and Surgery, University of Ferrara, Italy.,2 Unit of Translational Surgery and Vascular Diseases Center, Sant'Anna University Hospital, Ferrara, Italy
| | - Massimo Pedriali
- 3 Department of Experimental and Diagnostic Medicine, Sant'Anna University Hospital, Ferrara, Italy
| | - Maria Grazia Sibilla
- 2 Unit of Translational Surgery and Vascular Diseases Center, Sant'Anna University Hospital, Ferrara, Italy
| | - Mirko Tessari
- 1 Department of Morphology, Surgery and Experimental Medicine, Section of Translational of Medicine and Surgery, University of Ferrara, Italy.,2 Unit of Translational Surgery and Vascular Diseases Center, Sant'Anna University Hospital, Ferrara, Italy
| | - Francesco Carinci
- 1 Department of Morphology, Surgery and Experimental Medicine, Section of Translational of Medicine and Surgery, University of Ferrara, Italy
| | - Savino Occhionorelli
- 1 Department of Morphology, Surgery and Experimental Medicine, Section of Translational of Medicine and Surgery, University of Ferrara, Italy.,2 Unit of Translational Surgery and Vascular Diseases Center, Sant'Anna University Hospital, Ferrara, Italy
| | - Paolo Zamboni
- 1 Department of Morphology, Surgery and Experimental Medicine, Section of Translational of Medicine and Surgery, University of Ferrara, Italy.,2 Unit of Translational Surgery and Vascular Diseases Center, Sant'Anna University Hospital, Ferrara, Italy
| |
Collapse
|
44
|
Dunyach-Remy C, Ngba Essebe C, Sotto A, Lavigne JP. Staphylococcus aureus Toxins and Diabetic Foot Ulcers: Role in Pathogenesis and Interest in Diagnosis. Toxins (Basel) 2016; 8:toxins8070209. [PMID: 27399775 PMCID: PMC4963842 DOI: 10.3390/toxins8070209] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 06/29/2016] [Accepted: 07/01/2016] [Indexed: 12/17/2022] Open
Abstract
Infection of foot ulcers is a common, often severe and costly complication in diabetes. Diabetic foot infections (DFI) are mainly polymicrobial, and Staphylococcus aureus is the most frequent pathogen isolated. The numerous virulence factors and toxins produced by S. aureus during an infection are well characterized. However, some particular features could be observed in DFI. The aim of this review is to describe the role of S. aureus in DFI and the implication of its toxins in the establishment of the infection. Studies on this issue have helped to distinguish two S. aureus populations in DFI: toxinogenic S. aureus strains (harboring exfoliatin-, EDIN-, PVL- or TSST-encoding genes) and non-toxinogenic strains. Toxinogenic strains are often present in infections with a more severe grade and systemic impact, whereas non-toxinogenic strains seem to remain localized in deep structures and bone involving diabetic foot osteomyelitis. Testing the virulence profile of bacteria seems to be a promising way to predict the behavior of S. aureus in the chronic wounds.
Collapse
Affiliation(s)
- Catherine Dunyach-Remy
- Institut National de la Santé Et de la Recherche Médicale U1047, Université de Montpellier, UFR de Médecine, Nîmes 30908, France.
- Service de Microbiologie, Centre Hospitalo-Universitaire Carémeau, Nîmes 30029, France.
| | - Christelle Ngba Essebe
- Institut National de la Santé Et de la Recherche Médicale U1047, Université de Montpellier, UFR de Médecine, Nîmes 30908, France.
| | - Albert Sotto
- Institut National de la Santé Et de la Recherche Médicale U1047, Université de Montpellier, UFR de Médecine, Nîmes 30908, France.
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalo-Universitaire Carémeau, Nîmes 30029, France.
| | - Jean-Philippe Lavigne
- Institut National de la Santé Et de la Recherche Médicale U1047, Université de Montpellier, UFR de Médecine, Nîmes 30908, France.
- Service de Microbiologie, Centre Hospitalo-Universitaire Carémeau, Nîmes 30029, France.
| |
Collapse
|
45
|
Dhall S, Alamat R, Castro A, Sarker AH, Mao JH, Chan A, Hang B, Martins-Green M. Tobacco toxins deposited on surfaces (third hand smoke) impair wound healing. Clin Sci (Lond) 2016; 130:1269-84. [PMID: 27129193 DOI: 10.1042/cs20160236] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/28/2016] [Indexed: 02/04/2023]
Abstract
Third hand smoke (THS) is the accumulation of second hand smoke (SHS) toxins on surfaces in homes, cars, clothing and hair of smokers. It is known that 88M US nonsmokers ≥3 years old living in homes of smokers are exposed to THS toxicants and show blood cotinine levels of ≥0.05 ng/ml, indicating that the toxins are circulating in their circulatory systems. The goal of the present study is to investigate the mechanisms by which THS causes impaired wound healing. We show that mice living under conditions that mimic THS exposure in humans display delayed wound closure, impaired collagen deposition, altered inflammatory response, decreased angiogenesis, microvessels with fibrin cuffs and a highly proteolytic wound environment. Moreover, THS-exposed mouse wounds have high levels of oxidative stress and significantly lower levels of antioxidant activity leading to molecular damage, including protein nitration, lipid peroxidation and DNA damage that contribute to tissue dysfunction. Furthermore, we show that elastase is elevated, suggesting that elastin is degraded and the plasticity of the wound tissue is decreased. Taken together, our results lead us to conclude that THS toxicants delay and impair wound healing by disrupting the sequential processes that lead to normal healing. In addition, the lack of elastin results in loss of wound plasticity, which may be responsible for reopening of wounds.
Collapse
Affiliation(s)
- Sandeep Dhall
- Department of Cell Biology and Neuroscience, University of California at Riverside, Riverside, CA 92521, U.S.A
| | - Raquelle Alamat
- Department of Cell Biology and Neuroscience, University of California at Riverside, Riverside, CA 92521, U.S.A
| | - Anthony Castro
- Department of Cell Biology and Neuroscience, University of California at Riverside, Riverside, CA 92521, U.S.A
| | - Altaf H Sarker
- Biological Systems & Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, U.S.A
| | - Jian-Hua Mao
- Biological Systems & Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, U.S.A
| | - Alex Chan
- Department of Cell Biology and Neuroscience, University of California at Riverside, Riverside, CA 92521, U.S.A
| | - Bo Hang
- Biological Systems & Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, U.S.A
| | - Manuela Martins-Green
- Department of Cell Biology and Neuroscience, University of California at Riverside, Riverside, CA 92521, U.S.A.
| |
Collapse
|
46
|
Yager DR, Kulina RA, Gilman LA. Wound Fluids: A Window Into the Wound Environment? INT J LOW EXTR WOUND 2016; 6:262-72. [DOI: 10.1177/1534734607307035] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Wound healing of the skin is a complex biologic process involving temporal interactions between numerous types of cells, extracellular matrix molecules, and soluble factors. The process of repair can be viewed as involving 3 or 4 phases: homeostasis, inflammation, synthesis, and remodeling. These phases occur at different times and differ in their cellular, biochemical, and physiologic requirements. Disruption of one or more of these interactions can significantly interfere with the repair process. Such comorbidities as age, nutrition, immune status, and underlying disease status (eg, diabetes or venous stasis) contribute additional intricacy to the repair process. Because of this complexity, care of chronic wounds remains highly individualized, and it should not come as a surprise that treatment of these wounds as a group with single target therapies have met with only modest success. A major hurdle in the progression toward improved treatment regimens has been the lack of objective biochemical and physiological landmarks that can be used to assess wound status. Collection and biochemical characterization of wound fluids presents the opportunity to noninvasively obtain information reflecting the status of the wound and of specific biomarkers. This review discusses the collection of wound fluid and highlights biomarkers that may be useful to this end.
Collapse
Affiliation(s)
- Dorne R. Yager
- Department of Surgery, Viginia Commonwealth University
Medical Center, Richmond, Virginia, Department of Physiology, Virginia Commonweath University
Medical Center, Richmond, Virginia, Department of Biochemistry, Virginia Commonwealth University
Medical Center, Richmond, Virginia,
| | - Robert A. Kulina
- Department of Surgery, Viginia Commonwealth University
Medical Center, Richmond, Virginia
| | - Laura A. Gilman
- Department of Surgery, Viginia Commonwealth University
Medical Center, Richmond, Virginia
| |
Collapse
|
47
|
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: 985] [Impact Index Per Article: 109.4] [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
|
48
|
Craig M, Altskär A, Nordstierna L, Holmberg K. Bacteria-triggered degradation of nanofilm shells for release of antimicrobial agents. J Mater Chem B 2015; 4:672-682. [PMID: 32262949 DOI: 10.1039/c5tb01274k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Due to an increase in lifestyle diseases in the developed world, the number of chronic wounds is increasing at a fast pace. Chronic wound infections are common and systemic antibiotics are usually used as a treatment. In this paper we describe an approach to encapsulate antimicrobial agents in hollow microcapsules covered with a nanofilm shell that degrades through the action of a virulence factor from Pseudomonas aeruginosa. The shell was assembled using the layer-by-layer (LbL) technique with poly-l-lysine and hyaluronic acid. The microcapsules were loaded with a model substrate or a drug. By crosslinking the components in the nanofilm, the film remained intact when exposed to human wound proteases. However, the film was degraded and the drug exposed when in contact with Pseudomonas aeruginosa's Lys-X specific protease IV. The antimicrobial efficacy of the drug-loaded microcapsules was confirmed by exposure to virulent Pseudomonas aeruginosa. The current study contributes to the establishment of a release platform for targeted treatment of topical infections with the aim of minimizing both overexposure to drugs and development of bacterial resistance.
Collapse
Affiliation(s)
- Marina Craig
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, SE-41296, Gothenburg, Sweden.
| | | | | | | |
Collapse
|
49
|
Buz A, Görgülü T, Olgun A, Kargi E. Efficacy of glutathione mesotherapy in burns: an experimental study. Eur J Trauma Emerg Surg 2015; 42:775-783. [PMID: 26614529 DOI: 10.1007/s00068-015-0607-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/16/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND Thermal burns are the leading cause of trauma worldwide. Currently, no consensus on optimal treatment of deep partial-thickness (second-degree) burns has emerged, as reflected by the wide variability in available wound-care materials. The relative efficacies of products used for treatment of partial-thickness thermal burns remain unclear. Mesotherapy features intradermal administration of various agents, depending on burn location. In the present experimental study, we explored the efficacy of mesotherapy used to treat partial-thickness thermal burns in 50 male Wistar rats divided into five groups of equal number. No procedure was performed after infliction of thermal burns in control group (Group 1). Mesotherapy was applied with physiological saline in sham group (Group 2), glutathione, taurine, and L-carnitine were separately applied in Group 3, Group 4, and Group 5, respectively. MATERIALS AND METHODS Mesotherapeutic agents were injected intradermally into the reticular layer of the dermis using the point technique. The first course of mesotherapy was given within the first 2 h after infliction of thermal burns, and therapy was continued to day 10. On day 22, unhealed thermal burn areas were measured prior to sacrifice, and biopsies covering the total areas of burns were performed to allow of pathological evaluation. RESULTS Group 3 (the glutathione group) showed the best extent of healing, followed by Group 4 (the taurine group) and Group 5 (the L-carnitine group). The healed thermal burn areas in these groups were significantly greater than those in the control and sham groups (P = 0.001). All of healing, acute and chronic inflammation, the amount of granulation tissue, the level of fibroblast maturation, the amount of collagen, the extent of re-epithelization and neovascularization, and ulcer depth were scored upon pathological examination of tissue cross-sections. The best outcomes were evident in the glutathione group, with statistical significance. Although wound healing in the L-carnitine and taurine groups was better than in the control and sham groups, the differences were not statistically significant. CONCLUSION Thus, glutathione mesotherapy was effective when used to treat partial-thickness thermal burns and may be a useful treatment option for various human burns.
Collapse
Affiliation(s)
- A Buz
- Department of Plastic, Reconstructive and Aesthetic Surgery, Bulent Ecevit University Medical Faculty, A blok Kat:3, Kozlu, Zonguldak, Turkey
| | - T Görgülü
- Department of Plastic, Reconstructive and Aesthetic Surgery, Bulent Ecevit University Medical Faculty, A blok Kat:3, Kozlu, Zonguldak, Turkey.
| | - A Olgun
- Department of Plastic, Reconstructive and Aesthetic Surgery, Bulent Ecevit University Medical Faculty, A blok Kat:3, Kozlu, Zonguldak, Turkey
| | - E Kargi
- Department of Plastic, Reconstructive and Aesthetic Surgery, Bulent Ecevit University Medical Faculty, A blok Kat:3, Kozlu, Zonguldak, Turkey
| |
Collapse
|
50
|
Dhall S, Wijesinghe DS, Karim ZA, Castro A, Vemana HP, Khasawneh FT, Chalfant CE, Martins-Green M. Arachidonic acid-derived signaling lipids and functions in impaired healing. Wound Repair Regen 2015; 23:644-56. [PMID: 26135854 DOI: 10.1111/wrr.12337] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 05/26/2015] [Indexed: 12/30/2022]
Abstract
Very little is known about lipid function during wound healing, and much less during impaired healing. Such understanding will help identify what roles lipid signaling plays in the development of impaired/chronic wounds. We took a lipidomics approach to study the alterations in lipid profile in the LIGHT(-/-) mouse model of impaired healing which has characteristics that resemble those of impaired/chronic wounds in humans, including high levels of oxidative stress, excess inflammation, increased extracellular matrix degradation and blood vessels with fibrin cuffs. The latter suggests excess coagulation and potentially increased platelet aggregation. We show here that in these impaired wounds there is an imbalance in the arachidonic acid (AA) derived eicosonoids that mediate or modulate inflammatory reactions and platelet aggregation. In the LIGHT(-/-) impaired wounds there is a significant increase in enzymatically derived breakdown products of AA. We found that early after injury there was a significant increase in the eicosanoids 11-, 12-, and 15-hydroxyeicosa-tetranoic acid, and the proinflammatory leukotrienes (LTD4 and LTE) and prostaglandins (PGE2 and PGF2α ). Some of these eicosanoids also promote platelet aggregation. This led us to examine the levels of other eicosanoids known to be involved in the latter process. We found that thromboxane (TXA2 /B2 ), and prostacyclins 6kPGF1α are elevated shortly after wounding and in some cases during healing. To determine whether they have an impact in platelet aggregation and hemostasis, we tested LIGHT(-/-) mouse wounds for these two parameters and found that, indeed, platelet aggregation and hemostasis are enhanced in these mice when compared with the control C57BL/6 mice. Understanding lipid signaling in impaired wounds can potentially lead to development of new therapeutics or in using existing nonsteroidal anti-inflammatory agents to help correct the course of healing.
Collapse
Affiliation(s)
- Sandeep Dhall
- Department of Cell Biology and Neuroscience, University of California, Riverside, California.,Department of Bioengineering Interdepartmental Graduate Program, University of California, Riverside, California
| | - Dayanjan Shanaka Wijesinghe
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia.,Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, Virginia.,The Massey Cancer Center, Richmond, VA, Virginia Commonwealth University, Richmond, Virginia.,Virginia Commonwealth University Reanimation Engineering Science Center (VCURES)
| | - Zubair A Karim
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Anthony Castro
- Department of Cell Biology and Neuroscience, University of California, Riverside, California
| | - Hari Priya Vemana
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, California
| | - Fadi T Khasawneh
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Charles E Chalfant
- Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, Virginia.,The Massey Cancer Center, Richmond, VA, Virginia Commonwealth University, Richmond, Virginia.,Virginia Commonwealth University Reanimation Engineering Science Center (VCURES).,Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Manuela Martins-Green
- Department of Cell Biology and Neuroscience, University of California, Riverside, California.,Department of Bioengineering Interdepartmental Graduate Program, University of California, Riverside, California
| |
Collapse
|