1
|
Cheng X, Gao Z, Shan S, Shen H, Zheng H, Jin L, Li Q, Zhou J. Single cell transcriptomics reveals the cellular heterogeneity of keloids and the mechanism of their aggressiveness. Commun Biol 2024; 7:1647. [PMID: 39702490 DOI: 10.1038/s42003-024-07311-1] [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: 03/19/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
Keloid is a dermatofibrotic disease known for its aggressive nature and characterized by pathological scarring, which often leads to disfigurement and frequent recurrences. Effective therapies for keloids are still limited, presumably due to the inadequate comprehension of their aggressive mechanisms. In our study, we examined the unique scenario where both keloid and non-aggressive pathological scar originate from the same patient, providing a rare opportunity to explore the aggressive mechanisms of keloids through single-cell RNA sequencing. We found that the dominant fibroblast subgroup in keloids is mechanoresponsive group, which showed enhanced mechanotransduction and migration. This mechanoresponsive fibroblast subgroup is likely to be the key cell population and confer aggressive growth of keloids. The results also indicate that the endothelial cells and keratinocytes in keloid involve in endothelial-mesenchymal and epithelial-mesenchymal transitions. This study demonstrated the mechanoresponsive fibroblasts and multiple cellular mesenchymal processes could pave the way for further investigations into the keloid aggressiveness.
Collapse
Affiliation(s)
- Xinwei Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengzhou Shan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Shen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Hongkun Zheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Jin
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qingfeng Li
- 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
|
2
|
Lu P, Chen Z, Wu M, Feng S, Chen S, Cheng X, Zhao Y, Liu X, Gong L, Bian L, Yi S, Wang H. Type I collagen extracellular matrix facilitates nerve regeneration via the construction of a favourable microenvironment. BURNS & TRAUMA 2024; 12:tkae049. [PMID: 39659559 PMCID: PMC11631217 DOI: 10.1093/burnst/tkae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 12/12/2024]
Abstract
Background The extracellular matrix (ECM) provides essential physical support and biochemical cues for diverse biological activities, including tissue remodelling and regeneration, and thus is commonly applied in the construction of artificial peripheral nerve grafts. Nevertheless, the specific functions of essential peripheral nerve ECM components have not been fully determined. Our research aimed to differentially represent the neural activities of main components of ECM on peripheral nerve regeneration. Methods Schwann cells from sciatic nerves and neurons from dorsal root ganglia were isolated and cultured in vitro. The cells were seeded onto noncoated dishes, Matrigel-coated dishes, and dishes coated with the four major ECM components fibronectin, laminin, collagen I, and collagen IV. The effects of these ECM components on Schwann cell proliferation were determined via methylthiazolyldiphenyl-tetrazolium bromide (MTT), Cell Counting Kit-8, and 5-ethynyl-2'-deoxyuridine (EdU) assays, whereas their effects on cell migration were determined via wound healing and live-cell imaging. Neurite growth in neurons cultured on different ECM components was observed. Furthermore, the two types of collagen were incorporated into chitosan artificial nerves and used to repair sciatic nerve defects in rats. Immunofluorescence analysis and a behavioural assessment, including gait, electrophysiology, and target muscle analysis, were conducted. Results ECM components, especially collagen I, stimulated the DNA synthesis and movement of Schwann cells. Direct measurement of the neurite lengths of neurons cultured on ECM components further revealed the beneficial effects of ECM components on neurite outgrowth. Injection of collagen I into chitosan and poly(lactic-co-glycolic acid) artificial nerves demonstrated that collagen I facilitated axon regeneration and functional recovery after nerve defect repair by stimulating the migration of Schwann cells and the formation of new blood vessels. In contrast, collagen IV recruited excess fibroblasts and inflammatory macrophages and thus had disadvantageous effects on nerve regeneration. Conclusions These findings reveal the modulatory effects of specific ECM components on cell populations of peripheral nerves, reveal the contributing roles of collagen I in microenvironment construction and axon regeneration, and highlight the use of collagen I for the healing of injured peripheral nerves.
Collapse
Affiliation(s)
- Panjian Lu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Zhiying Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Mingjun Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Shuyue Feng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Sailing Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Xiyang Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Xingyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Lijing Bian
- Imperial College of Science, Technology and Medicine, London SW7 2AZ, United Kingdom
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| | - Hongkui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Medical School of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu 226001, China
| |
Collapse
|
3
|
Markov PA, Eremin PS, Paderin NM, Kostromina EY, Greben AI, Gilmutdinova IR. Effect of Bioplastic Material on Adhesion, Growth, and Proliferative Activity of Human Fibroblasts When Incubated in Solutions Mimic the Acidity of Wound an Acute and Chronic Inflammation. DOKL BIOCHEM BIOPHYS 2024; 519:540-546. [PMID: 39480638 DOI: 10.1134/s1607672924701187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 11/02/2024]
Abstract
INTRODUCTION One of the key stages of wound healing is the phase of inflammation, which is a transitional process between hemostasis and wound healing. Each stage of the inflammatory-reparative process is characterized by its own value of the acidity of the wound bed. For example, in the acute stage of inflammation, the acidity of the medium in the wound bed decreases to pH 5.5-6. The chronic stage of the inflammatory process, on the contrary, is accompanied by an increase in pH to 8. To date, the effect of biomaterials containing components of the intercellular matrix of the human dermis on fibroblasts under acidosis and alkalosis has not been fully investigated. AIM The aim of this study was to characterize the effect of bioplastic material based on collagen, hyaluronic acid, and elastin on the viability and proliferative activity of human fibroblasts in conditions simulating the acidity of acute and chronic wounds. MATERIALS AND METHODS Bioplastic material was made according to the method described in RF patent no. 2722744. Adhesive properties and proliferative activity of human fibroblasts were assessed visually using fluorescent microscopy. The number of apoptotic and necrotic cells was assessed by flow cytometry (BD FACSCanto II) using the commercial FITC Annexin V Apoptosis Detection Kit I (BD Pharmingen). The strength, Young's modulus, and elasticity of the gels were determined on a TA.XT-plus texture analyzer (Stable Micro Systems, Great Britain). RESULTS Using the methods of luminescent microscopy and flow cytometry, we found that the cell viability (namely, adhesive properties and proliferative activity) decreases after incubation on condition mimic of physiological acidosis. We found that bioplastic material contributes to the preservation of adhesive properties, viability, and proliferative activity of fibroblasts in physiological acidosis conditions. The results obtained indicate that bioplastic material based on soluble dermis components can be used as a biologically active component of wound dressings to increase the effectiveness of reparative regeneration, especially in cases of excessive acute inflammation.
Collapse
Affiliation(s)
- P A Markov
- National Medical Research Centre for Rehabilitation and Balneology' of the Ministry of Health of Russia, Moscow, Russia.
| | - P S Eremin
- National Medical Research Centre for Rehabilitation and Balneology' of the Ministry of Health of Russia, Moscow, Russia
| | - N M Paderin
- Institute of Physiology of Federal Research Centre "Komi Science Centre of the Urals Branch of the Russian Academy of Sciences", Syktyvkar, Russia
| | - E Yu Kostromina
- National Medical Research Centre for Rehabilitation and Balneology' of the Ministry of Health of Russia, Moscow, Russia
| | - A I Greben
- National Medical Research Centre for Rehabilitation and Balneology' of the Ministry of Health of Russia, Moscow, Russia
| | - I R Gilmutdinova
- National Medical Research Centre for Rehabilitation and Balneology' of the Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
4
|
Ma J, Li S, Zhang L, Lei B. Oxidativestress-scavenging thermo-activated MXene hydrogel for rapid repair of MRSA impaired wounds and burn wounds. MATERIALS TODAY 2024; 80:139-155. [DOI: 10.1016/j.mattod.2024.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
5
|
Zhang Z, Fan C, Xu Q, Guo F, Li W, Zeng Z, Xu Y, Yu J, Ge H, Yang C, Chang J. A New Strategy to Inhibit Scar Formation by Accelerating Normal Healing Using Silicate Bioactive Materials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407718. [PMID: 39340818 DOI: 10.1002/advs.202407718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/28/2024] [Indexed: 09/30/2024]
Abstract
Inspired by the scar-free wound healing in infants, an anti-scar strategy is proposed by accelerating wound healing using silicate bioactive materials. Bioglass/alginate composite hydrogels are applied, which significantly inhibit scar formation in rabbit ear scar models. The underlining mechanisms include stimulation of Integrin Subunit Alpha 2 expression in dermal fibroblasts to accelerate wound healing, and induction of apoptosis of hypertrophic scar fibroblasts by directly stimulating the N-Acylsphingosine Amidohydrolase 2 expression in hypertrophic scar fibroblasts, and indirectly upregulating the secretion of Cathepsin K in dermal fibroblasts. Considering specific functions of the bioactive silicate materials, two scar treatment regimes are tested. For severe scars, a regenerative intervention is applied by surgical removal of the scar followed by the treatment with bioactive hydrogels to reduce the formation of scars by activating dermal fibroblasts. For mild scars, the bioactive dressing is applied on the formed scar and reduces scar by inducing scar fibroblasts apoptosis.
Collapse
Affiliation(s)
- Zhaowenbin Zhang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Chen Fan
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
| | - Qing Xu
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Feng Guo
- Department of Plastic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200050, P. R. China
| | - Wenbo Li
- Department of Plastic Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, 200050, P. R. China
| | - Zhen Zeng
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
| | - Yuze Xu
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Jing Yu
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
| | - Hongping Ge
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
| | - Chen Yang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
| | - Jiang Chang
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, P. R. China
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
6
|
Schoberleitner I, Lackner M, Coraça-Huber DC, Augustin A, Imsirovic A, Sigl S, Wolfram D. SMI-Capsular Fibrosis and Biofilm Dynamics: Molecular Mechanisms, Clinical Implications, and Antimicrobial Approaches. Int J Mol Sci 2024; 25:11675. [PMID: 39519227 PMCID: PMC11546664 DOI: 10.3390/ijms252111675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Silicone mammary implants (SMIs) frequently result in capsular fibrosis, which is marked by the overproduction of fibrous tissue surrounding the implant. This review provides a detailed examination of the molecular and immunological mechanisms driving capsular fibrosis, focusing on the role of foreign body responses (FBRs) and microbial biofilm formation. We investigate how microbial adhesion to implant surfaces and biofilm development contribute to persistent inflammation and fibrotic responses. The review critically evaluates antimicrobial strategies, including preoperative antiseptic protocols and antimicrobial-impregnated materials, designed to mitigate infection and biofilm-related complications. Additionally, advancements in material science, such as surface modifications and antibiotic-impregnated meshes, are discussed for their potential to reduce capsular fibrosis and prevent contracture of the capsule. By integrating molecular insights with clinical applications, this review aims to elucidate the current understanding of SMI-related fibrotic responses and highlight knowledge gaps. The synthesis of these findings aims to guide future research directions of improved antimicrobial interventions and implant materials, ultimately advancing the management of capsular fibrosis and enhancing patient outcomes.
Collapse
Affiliation(s)
- Ines Schoberleitner
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Débora C. Coraça-Huber
- BIOFILM Lab, Department of Orthopedics and Traumatology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Angela Augustin
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Anja Imsirovic
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
7
|
Kübler IC, Kretzschmar J, Arredondo-Lasso MN, Keeley SD, Rößler LC, Ganss K, Sandoval-Guzmán T, Brankatschk M. Systemic and local lipid adaptations underlie regeneration in Drosophila melanogaster and Ambystoma mexicanum. NPJ Regen Med 2024; 9:33. [PMID: 39472660 PMCID: PMC11522293 DOI: 10.1038/s41536-024-00375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
In regenerating tissues, synthesis and remodeling of membranes rely on lipid turnover and transport. Our study addresses lipid adaptations in intestinal regeneration of Drosophila melanogaster and limb regeneration of Ambystoma mexicanum. We found changes in lipid profiles at different locations: transport, storage organs and regenerating tissues. We demonstrate that attenuating insulin signaling, exclusively in fat storage, inhibits the regeneration-specific response in both the fat storage and the regenerating tissue in Drosophila. Furthermore, in uninjured axolotls we found sex-specific lipid profiles in both storage and circulation, while in regenerating animals these differences subside. The regenerating limb presents a unique sterol profile, albeit with no sex differences. We postulate that regeneration triggers a systemic response, where organs storing lipids play a significant role in the regulation of systemic lipid traffic. Second, that this response may be an active and well-regulated mechanism, as observed when homeostatic sex-differences disappear in regenerating salamanders.
Collapse
Affiliation(s)
- Ines C Kübler
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jenny Kretzschmar
- Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany
- MRC Laboratory of Molecular Biology, Francis Crick Ave, Trumpington, Cambridge, UK
| | - Maria Nieves Arredondo-Lasso
- Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany
- Metabolic Programming, Technische Universität München, Freising-Weihenstephan, Germany
| | - Sean D Keeley
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Luca Claudia Rößler
- Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany
| | - Katharina Ganss
- Paul Langerhans Institute Dresden, Helmholtz Centre Munich, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Tatiana Sandoval-Guzmán
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Paul Langerhans Institute Dresden, Helmholtz Centre Munich, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Center for Regenerative Therapies TU Dresden, Technische Universität Dresden, Dresden, Germany.
| | - Marko Brankatschk
- Biotechnology Center (BIOTEC), Technische Universität Dresden, Dresden, Germany.
- Faculty of Biology Technische Universität Dresden, Dresden, Germany. Zellescher Weg 23b, Dresden, Germany.
| |
Collapse
|
8
|
Toledano-Macías E, Martínez-Pascual MA, Cecilia-Matilla A, Bermejo-Martínez M, Pérez-González A, Jara RC, Sacristán S, Hernández-Bule ML. Radiofrequency Currents Modulate Inflammatory Processes in Keratinocytes. Int J Mol Sci 2024; 25:10663. [PMID: 39408993 PMCID: PMC11476504 DOI: 10.3390/ijms251910663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Keratinocytes play an essential role in the inflammatory phase of wound regeneration. In addition to migrating and proliferating for tissue regeneration, they produce a large amount of cytokines that modulate the inflammatory process. Previous studies have shown that subthermal treatment with radiofrequency (RF) currents used in capacitive resistive electric transfer (CRET) therapy promotes the proliferation of HaCat keratinocytes and modulates their cytokine production. Although physical therapies have been shown to have anti-inflammatory effects in a variety of experimental models and in patients, knowledge of the biological basis of these effects is still limited. The aim of this study was to investigate the effect of CRET on keratinocyte proliferation, cytokine production (IL-8, MCP-1, RANTES, IL-6, IL-11), TNF-α secretion, and the expression of MMP9, MMP1, NF-κB, ERK1/2, and EGFR. Human keratinocytes (HaCat) were treated with an intermittent 448 kHz electric current (CRET signal) in subthermal conditions and for different periods of time. Cell proliferation was analyzed by XTT assay, cytokine and TNF-α production by ELISA, NF-κB expression and activation by immunofluorescence, and MMP9, MMP1, ERK1/2, and EGF receptor expression and activation by immunoblot. Compared to a control, CRET increases keratinocyte proliferation, increases the transient release of MCP-1, TNF-α, and IL-6 while decreasing IL-8. In addition, it modifies the expression of MMPs and activates EGFR, NF-κB, and ERK1/2 proteins. Our results indicate that CRET reasonably modifies cytokine production through the EGF receptor and the ERK1/2/NF-κB pathway, ultimately modulating the inflammatory response of human keratinocytes.
Collapse
Affiliation(s)
- Elena Toledano-Macías
- Photobiology and Bioelectromagnetic Lab, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain; (E.T.-M.); (M.A.M.-P.); (R.C.J.)
| | - María Antonia Martínez-Pascual
- Photobiology and Bioelectromagnetic Lab, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain; (E.T.-M.); (M.A.M.-P.); (R.C.J.)
| | - Almudena Cecilia-Matilla
- Angiology and Vascular Surgery Service, Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain; (A.C.-M.); (M.B.-M.)
| | - Mariano Bermejo-Martínez
- Angiology and Vascular Surgery Service, Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain; (A.C.-M.); (M.B.-M.)
| | - Alfonso Pérez-González
- Dermatology Service, Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain;
| | - Rosa Cristina Jara
- Photobiology and Bioelectromagnetic Lab, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain; (E.T.-M.); (M.A.M.-P.); (R.C.J.)
| | - Silvia Sacristán
- Aptamer Group, Histology Lab, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain;
| | - María Luisa Hernández-Bule
- Photobiology and Bioelectromagnetic Lab, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Ramón y Cajal, Crta. Colmenar Viejo, km. 9.100, 28034 Madrid, Spain; (E.T.-M.); (M.A.M.-P.); (R.C.J.)
| |
Collapse
|
9
|
Shang Y, Li M, Zhang L, Han C, Shen K, Wang K, Li Y, Zhang Y, Luo L, Jia Y, Guo K, Cai W, Zhang J, Wang X, Wang H, Hu D. Exosomes derived from mouse vibrissa dermal papilla cells promote hair follicle regeneration during wound healing by activating Wnt/β-catenin signaling pathway. J Nanobiotechnology 2024; 22:425. [PMID: 39030543 PMCID: PMC11264511 DOI: 10.1186/s12951-024-02689-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/01/2024] [Indexed: 07/21/2024] Open
Abstract
Hair follicle (HF) regeneration during wound healing continues to present a significant clinical challenge. Dermal papilla cell-derived exosomes (DPC-Exos) hold immense potential for inducing HF neogenesis. However, the accurate role and underlying mechanisms of DPC-Exos in HF regeneration in wound healing remain to be fully explained. This study, represents the first analysis into the effects of DPC-Exos on fibroblasts during wound healing. Our findings demonstrated that DPC-Exos could stimulate the proliferation and migration of fibroblasts, more importantly, enhance the hair-inducing capacity of fibroblasts. Fibroblasts treated with DPC-Exos were capable of inducing HF neogenesis in nude mice when combined with neonatal mice epidermal cells. In addition, DPC-Exos accelerated wound re-epithelialization and promoted HF regeneration during the healing process. Treatment with DPC-Exos led to increased expression levels of the Wnt pathway transcription factors β-catenin and Lef1 in both fibroblasts and the dermis of skin wounds. Specifically, the application of a Wnt pathway inhibitor reduced the effects of DPC-Exos on fibroblasts and wound healing. Accordingly, these results offer evidence that DPC-Exos promote HF regeneration during wound healing by enhancing the hair-inducing capacity of fibroblasts and activating the Wnt/β-catenin signaling pathway. This suggests that DPC-Exos may represent a promising therapeutic strategy for achieving regenerative wound healing.
Collapse
Affiliation(s)
- Yage Shang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Mengyang Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Lixia Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Chao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Kuo Shen
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Kejia Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yue Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yanhui Jia
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Kai Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jian Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xujie Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Hongtao Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China.
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China.
| |
Collapse
|
10
|
Liu X, Teng Y, Li H, Luo D, Li H, Shen J, Du S, Zhang Y, Wang D, Jing J. Identification of IGF2 promotes skin wound healing by co-expression analysis. Int Wound J 2024; 21:e14862. [PMID: 38572823 PMCID: PMC10993366 DOI: 10.1111/iwj.14862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2024] Open
Abstract
Oral mucosa is an ideal model for studying scarless wound healing. Researchers have shown that the key factors which promote scarless wound healing already exist in basal state of oral mucosa. Thus, to identify the other potential factors in basal state of oral mucosa will benefit to skin wound healing. In this study, we identified eight gene modules enriched in wound healing stages of human skin and oral mucosa through co-expression analysis, among which the module M8 was only module enriched in basal state of oral mucosa, indicating that the genes in module M8 may have key factors mediating scarless wound healing. Through bioinformatic analysis of genes in module M8, we found IGF2 may be the key factor mediating scarless wound healing of oral mucosa. Then, we purified IGF2 protein by prokaryotic expression, and we found that IGF2 could promote the proliferation and migration of HaCaT cells. Moreover, IGF2 promoted wound re-epithelialization and accelerated wound healing in a full-thickness skin wound model. Our findings identified IGF2 as a factor to promote skin wound healing which provide a potential target for wound healing therapy in clinic.
Collapse
Affiliation(s)
- Xingyan Liu
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
- Department of Burns and Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Ying Teng
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
- Department of Burns and Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Huan Li
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
| | - Ding Luo
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
| | - Hongkun Li
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
- Department of Burns and Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Jinghan Shen
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
- Department of Burns and Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Simin Du
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
- Department of Burns and Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Yuyue Zhang
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
- Department of Burns and Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Dali Wang
- Department of Burns and Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical UniversityZunyiChina
| | - Jie Jing
- School and Hospital of Stomatology, Zunyi Medical UniversityZunyiChina
| |
Collapse
|
11
|
Bahraoui S, Tejedor G, Mausset-Bonnefont AL, Autelitano F, Barthelaix A, Terraza-Aguirre C, Gisbert V, Arribat Y, Jorgensen C, Wei M, Djouad F. PLOD2, a key factor for MRL MSC metabolism and chondroprotective properties. Stem Cell Res Ther 2024; 15:70. [PMID: 38454524 PMCID: PMC10921602 DOI: 10.1186/s13287-024-03650-2] [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: 03/27/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Initially discovered for its ability to regenerate ear holes, the Murphy Roth Large (MRL) mouse has been the subject of multiple research studies aimed at evaluating its ability to regenerate other body tissues and at deciphering the mechanisms underlying it. These enhanced abilities to regenerate, retained during adulthood, protect the MRL mouse from degenerative diseases such as osteoarthritis (OA). Here, we hypothesized that mesenchymal stromal/stem cells (MSC) derived from the regenerative MRL mouse could be involved in their regenerative potential through the release of pro-regenerative mediators. METHOD To address this hypothesis, we compared the secretome of MRL and BL6 MSC and identified several candidate molecules expressed at significantly higher levels by MRL MSC than by BL6 MSC. We selected one candidate, Plod2, and performed functional in vitro assays to evaluate its role on MRL MSC properties including metabolic profile, migration, and chondroprotective effects. To assess its contribution to MRL protection against OA, we used an experimental model for osteoarthritis induced by collagenase (CiOA). RESULTS Among the candidate molecules highly expressed by MRL MSC, we focused our attention on procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 (PLOD2). Plod2 silencing induced a decrease in the glycolytic function of MRL MSC, resulting in the alteration of their migratory and chondroprotective abilities in vitro. In vivo, we showed that Plod2 silencing in MRL MSC significantly impaired their capacity to protect mouse from developing OA. CONCLUSION Our results demonstrate that the chondroprotective and therapeutic properties of MRL MSC in the CiOA experimental model are in part mediated by PLOD2.
Collapse
Affiliation(s)
- Sarah Bahraoui
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
- CellVax, Villejuif Bio Park, 1 Mail du Professeur Georges Mathé, 94800, Villejuif, France
| | - Gautier Tejedor
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
| | - Anne-Laure Mausset-Bonnefont
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
| | | | - Audrey Barthelaix
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
| | - Claudia Terraza-Aguirre
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
- CellVax, Villejuif Bio Park, 1 Mail du Professeur Georges Mathé, 94800, Villejuif, France
| | - Vincent Gisbert
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
| | - Yoan Arribat
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
| | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France
| | - Mingxing Wei
- CellVax, Villejuif Bio Park, 1 Mail du Professeur Georges Mathé, 94800, Villejuif, France
| | - Farida Djouad
- IRMB, University of Montpellier, INSERM U 1183, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier cedex 5, France.
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France.
| |
Collapse
|
12
|
Pan Z, Zhang X, Xie W, Cui J, Wang Y, Zhang B, Du L, Zhai W, Sun H, Li Y, Li D. Revisited and innovative perspectives of oral ulcer: from biological specificity to local treatment. Front Bioeng Biotechnol 2024; 12:1335377. [PMID: 38456005 PMCID: PMC10917957 DOI: 10.3389/fbioe.2024.1335377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Mouth ulcers, a highly prevalent ailment affecting the oral mucosa, leading to pain and discomfort, significantly impacting the patient's daily life. The development of innovative approaches for oral ulcer treatment is of great importance. Moreover, a deeper and more comprehensive understanding of mouth ulcers will facilitate the development of innovative therapeutic strategies. The oral environment possesses distinct traits as it serves as the gateway to the digestive and respiratory systems. The permeability of various epithelial layers can influence drug absorption. Moreover, oral mucosal injuries exhibit distinct healing patterns compared to cutaneous lesions, influenced by various inherent and extrinsic factors. Furthermore, the moist and dynamic oral environment, influenced by saliva and daily physiological functions like chewing and speaking, presents additional challenges in local therapy. Also, suitable mucosal adhesion materials are crucial to alleviate pain and promote healing process. To this end, the review comprehensively examines the anatomical and structural aspects of the oral cavity, elucidates the healing mechanisms of oral ulcers, explores the factors contributing to scar-free healing in the oral mucosa, and investigates the application of mucosal adhesive materials as drug delivery systems. This endeavor seeks to offer novel insights and perspectives for the treatment of oral ulcers.
Collapse
Affiliation(s)
- Ziyi Pan
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
- School of Stomatology, Jilin University, Changchun, China
| | - Xu Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Wangni Xie
- School of Stomatology, Jilin University, Changchun, China
| | - Jing Cui
- School of Stomatology, Jilin University, Changchun, China
| | - Yue Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China
| | - Boya Zhang
- School of Stomatology, Jilin University, Changchun, China
| | - Liuyi Du
- School of Stomatology, Jilin University, Changchun, China
| | - Wenhao Zhai
- School of Stomatology, Jilin University, Changchun, China
| | - Hongchen Sun
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
- School of Stomatology, Jilin University, Changchun, China
| | - Yunfeng Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
- School of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
13
|
Frech S, Lichtenberger BM. Modulating embryonic signaling pathways paves the way for regeneration in wound healing. Front Physiol 2024; 15:1367425. [PMID: 38434140 PMCID: PMC10904466 DOI: 10.3389/fphys.2024.1367425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/05/2024] [Indexed: 03/05/2024] Open
Abstract
Epithelial tissues, including the skin, are highly proliferative tissues with the capability to constant renewal and regeneration, a feature that is essential for survival as the skin forms a protective barrier against external insults and water loss. In adult mammalian skin, every injury will lead to a scar. The scar tissue that is produced to seal the wound efficiently is usually rigid and lacks elasticity and the skin's original resilience to external impacts, but also secondary appendages such as hair follicles and sebaceous glands. While it was long thought that hair follicles develop solely during embryogenesis, it is becoming increasingly clear that hair follicles can also regenerate within a wound. The ability of the skin to induce hair neogenesis following injury however declines with age. As fetal and neonatal skin have the remarkable capacity to heal without scarring, the recapitulation of a neonatal state has been a primary target of recent regenerative research. In this review we highlight how modulating dermal signaling or the abundance of specific fibroblast subsets could be utilized to induce de novo hair follicles within the wound bed, and thus to shift wound repair with a scar to scarless regeneration.
Collapse
|
14
|
Voza FA, Huerta CT, Le N, Shao H, Ribieras A, Ortiz Y, Atkinson C, Machuca T, Liu ZJ, Velazquez OC. Fibroblasts in Diabetic Foot Ulcers. Int J Mol Sci 2024; 25:2172. [PMID: 38396848 PMCID: PMC10889208 DOI: 10.3390/ijms25042172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Fibroblasts are stromal cells ubiquitously distributed in the body of nearly every organ tissue. These cells were previously considered to be "passive cells", solely responsible for ensuring the turnover of the extracellular matrix (ECM). However, their versatility, including their ability to switch phenotypes in response to tissue injury and dynamic activity in the maintenance of tissue specific homeostasis and integrity have been recently revealed by the innovation of technological tools such as genetically modified mouse models and single cell analysis. These highly plastic and heterogeneous cells equipped with multifaceted functions including the regulation of angiogenesis, inflammation as well as their innate stemness characteristics, play a central role in the delicately regulated process of wound healing. Fibroblast dysregulation underlies many chronic conditions, including cardiovascular diseases, cancer, inflammatory diseases, and diabetes mellitus (DM), which represent the current major causes of morbidity and mortality worldwide. Diabetic foot ulcer (DFU), one of the most severe complications of DM affects 40 to 60 million people. Chronic non-healing DFU wounds expose patients to substantial sequelae including infections, gangrene, amputation, and death. A complete understanding of the pathophysiology of DFU and targeting pathways involved in the dysregulation of fibroblasts are required for the development of innovative new therapeutic treatments, critically needed for these patients.
Collapse
Affiliation(s)
- Francesca A. Voza
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Carlos Theodore Huerta
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Nga Le
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Hongwei Shao
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Antoine Ribieras
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Yulexi Ortiz
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carl Atkinson
- Department of Internal Medicine, Division of Pulmonary Critical Care & Sleep Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Tiago Machuca
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Zhao-Jun Liu
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Omaida C. Velazquez
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
15
|
Jiang X, Ma J, Xue K, Chen J, Zhang Y, Zhang G, Wang K, Yao Z, Hu Q, Lin C, Lei B, Mao C. Highly Bioactive MXene-M2-Exosome Nanocomposites Promote Angiogenic Diabetic Wound Repair through Reconstructing High Glucose-Derived Immune Inhibition. ACS NANO 2024; 18:4269-4286. [PMID: 38270104 DOI: 10.1021/acsnano.3c09721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
The repair of diabetic wounds remains challenging, primarily due to the high-glucose-derived immune inhibition which often leads to the excessive inflammatory response, impaired angiogenesis, and heightened susceptibility to infection. However, the means to reduce the immunosuppression and regulate the conversion of M2 phenotype macrophages under a high-glucose microenvironment using advanced biomaterials for diabetic wounds are not yet fully understood. Herein, we report two-dimensional carbide (MXene)-M2 macrophage exosome (Exo) nanohybrids (FM-Exo) for promoting diabetic wound repair by overcoming the high-glucose-derived immune inhibition. FM-Exo showed the sustained release of M2 macrophage-derived exosomes (M2-Exo) up to 7 days and exhibited broad-spectrum antibacterial activity. In the high-glucose microenvironment, relative to the single Exo, FM-Exo could significantly induce the optimized M2a/M2c polarization ratio of macrophages by activating the PI3K/Akt signaling pathway, promoting the proliferation, migration of fibroblasts, and angiogenic ability of endothelial cells. In the diabetic full-thickness wound model, FM-Exo effectively regulated the polarization status of macrophages and promoted their transition to the M2 phenotype, thereby inhibiting inflammation, promoting angiogenesis through VEGF secretion, and improving proper collagen deposition. As a result, the healing process was accelerated, leading to a better healing outcome with reduced scarring. Therefore, this study introduced a promising approach to address diabetic wounds by developing bioactive nanomaterials to regulate immune inhibition in a high-glucose environment.
Collapse
Affiliation(s)
- Xiaoqi Jiang
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, 322100, China
| | - Junping Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
| | - Kaikai Xue
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jinghao Chen
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yu Zhang
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Guojian Zhang
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Kangyan Wang
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhe Yao
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
- Department of Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qing Hu
- School of Material Science and Engineering, Jingdezhen Ceramic University, Jingdezhen 333001, China
| | - Cai Lin
- Department of Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Bo Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
| | - Cong Mao
- Key Laboratory of Orthopedics of Zhejiang Province, Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
16
|
Li W, Huang P, Wei J, Tan S, Liu G, Yang Q, Wang G. Down-regulation of miR-21-5p by pirfenidone to inhibit fibroblast proliferation in the treatment of acquired tracheal stenosis. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e13727. [PMID: 38151323 PMCID: PMC10775887 DOI: 10.1111/crj.13727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 12/10/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE Treatment options for acquired tracheal stenosis (ATS) are limited due to a series of pathophysiological changes including inflammation and cell proliferation. Micro ribonucleic acid-21-5p (miR-21-5p) may promote the excessive proliferation of fibroblasts. However, various types of fibrosis can be prevented with pirfenidone (PFD). Currently, the effect of PFD on miR-21-5p and its biological function has not been clarified. In this study, PFD was evaluated as a potential treatment for ATS by inducing fibroblast proliferation in lipopolysaccharide (LPS)-induced fibroblasts by targeting miR-21-5p. METHODS For 48 h, 1 g/ml LPS was used to generate fibroblasts in vitro, followed by the separation of cells into four groups: control, PFD, mimic, and mimic + PFD. The Cell Counting Kit-8 (CCK-8) technique was adopted to measure the proliferation of fibroblasts. Real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot (WB) were used to measure the relative expressions of tumor necrosis factor-α (TNF-α), transforming growth factor-β1 (TGF-β1), drosophila mothers against decapentaplegic 7 (Smad7) and collagen type I alpha 1(COL1A1) messenger RNA (mRNA) and proteins, respectively. RESULTS (1) At 0, 24, 48, and 72 h, fibroblast growth was assessed using the CCK-8 method. Compared with the control group, the mimic group showed the highest fibroblast viability, and the PFD group showed the lowest fibroblast viability. However, fibroblast viability increased in the mimic + PFD group but decreased in the mimic one. (2) RT-qPCR and WB showed that the mimic group exhibited a significant up-regulation in the relative expressions of TNF-α, TGF-β1, and COL1A1 mRNA and proteins but a down-regulation in the relative expression of Smad7 mRNA and protein compared with the control one. In the PFD group, the results were the opposite. Nevertheless, the relative expressions of TNF-α, TGF-β1, and COL1A1 mRNA and proteins were increased, whereas that of Smad7 mRNA was decreased in the mimic + PFD group. The change was less in the mimic group. CONCLUSION PFD may have a preventive and curative effect on ATS by inhibiting fibroblast proliferation and the fibrotic process and possibly through down-regulating miR-21-5p and up-regulating Smad7 and its mediated fibrotic and inflammatory responses.
Collapse
Affiliation(s)
- Wentao Li
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Pingping Huang
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jinmei Wei
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Sen Tan
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Guangnan Liu
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Qiu Yang
- Department of OphthalmologyRuikang Hospital Affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Guangfa Wang
- Department of Respiratory and Critical Care MedicinePeking University First HospitalBeijingChina
| |
Collapse
|
17
|
Helm M, Schmidt M, Del Duca E, Liu Y, Mortensen LS, Loui J, Zheng Y, Binder H, Guttman-Yassky E, Cotsarelis G, Simon JC, Ferrer RA. Repurposing DPP4 Inhibition to Improve Hair Follicle Activation and Regeneration. J Invest Dermatol 2023; 143:2132-2144.e15. [PMID: 37236597 DOI: 10.1016/j.jid.2023.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023]
Abstract
Skin injury and several diseases elicit fibrosis and induce hair follicle (HF) growth arrest and loss. The resulting alopecia and disfiguration represent a severe burden for patients, both physically and psychologically. Reduction of profibrotic factors such as dipeptidyl peptidase 4 (DPP4) might be a strategy to tackle this issue. We show DPP4 overrepresentation in settings with HF growth arrest (telogen), HF loss, and nonregenerative wound areas in mouse skin and human scalp. Topical DPP4 inhibition with Food and Drug Administration/European Medicines Agency-approved sitagliptin on preclinical models of murine HF activation/regeneration results in accelerated anagen progress, whereas treatment of wounds with sitagliptin results in reduced expression of fibrosis markers, increased induction of anagen around wounds, and HF regeneration in the wound center. These effects are associated with higher expression of Wnt target Lef1, known to be required for HF anagen/HF-activation and regeneration. Sitagliptin treatment decreases profibrotic signaling in the skin, induces a differentiation trajectory of HF cells, and activates Wnt targets related to HF activation/growth but not those supporting fibrosis. Taken together, our study shows a role for DPP4 in HF biology and shows how DPP4 inhibition, currently used as oral medication to treat diabetes, could be repurposed into a topical treatment agent to potentially reverse HF loss in alopecia and after injury.
Collapse
Affiliation(s)
- Maria Helm
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany
| | - Maria Schmidt
- Interdisciplinary Center for Bioinformatics, University Leipzig, Leipzig, Germany
| | - Ester Del Duca
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, Mount Sinai, New York City, New York, USA
| | - Ying Liu
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, Mount Sinai, New York City, New York, USA
| | - Lena Sünke Mortensen
- Interdisciplinary Center for Bioinformatics, University Leipzig, Leipzig, Germany
| | - Juliane Loui
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany
| | - Ying Zheng
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hans Binder
- Interdisciplinary Center for Bioinformatics, University Leipzig, Leipzig, Germany
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, Mount Sinai, New York City, New York, USA
| | - George Cotsarelis
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany
| | - Rubén A Ferrer
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Center, University Leipzig, Leipzig, Germany.
| |
Collapse
|
18
|
Ma J, Zhang L, Lei B. Multifunctional MXene-Based Bioactive Materials for Integrated Regeneration Therapy. ACS NANO 2023; 17:19526-19549. [PMID: 37804317 DOI: 10.1021/acsnano.3c01913] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Abstract
The reconstruction engineering of tissue defects accompanied by major diseases including cancer, infection, and inflammation is one of the important challenges in clinical medicine. The development of innovative tissue engineering strategies such as multifunctional bioactive materials presents a great potential to overcome the challenge of disease-impaired tissue regeneration. As the major representative of two-dimensional nanomaterials, MXenes have shown multifunctional physicochemical properties and have been diffusely studied as multimodal nanoplatforms in the field of biomedicine. This review summarized the recent advances in the multifunctional properties of MXenes and integrated regeneration-therapy applications of MXene-based biomaterials, including tissue regeneration-tumor therapy, tissue regeneration-infection therapy, and tissue regeneration-inflammation therapy. MXenes have been recognized as good candidates for promoting tissue regeneration and treating diseases through photothermal therapy, regulating cell behavior, and drug and gene delivery. The current challenges and future perspectives of MXene-based biomaterials in integrated regeneration-therapy are also discussed well in this review. In summary, MXene-based biomaterials have shown promising potential for integrated tissue regeneration and disease treatment due to their favorable physicochemical properties and bioactive functions. However, there are still many obstacles and challenges that must be addressed for the regeneration-therapy applications of MXene-based biomaterials, including understanding the bioactive mechanism, ensuring long-term biosafety, and improving their targeting therapy capacity.
Collapse
Affiliation(s)
- Junping Ma
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
| | - Long Zhang
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Bo Lei
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an 710000, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710000, China
| |
Collapse
|
19
|
Fernández-Guarino M, Hernández-Bule ML, Bacci S. Cellular and Molecular Processes in Wound Healing. Biomedicines 2023; 11:2526. [PMID: 37760967 PMCID: PMC10525842 DOI: 10.3390/biomedicines11092526] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
This review summarizes the recent knowledge of the cellular and molecular processes that occur during wound healing. However, these biological mechanisms have yet to be defined in detail; this is demonstrated by the fact that alterations of events to pathological states, such as keloids, consisting of the excessive formation of scars, have consequences yet to be defined in detail. Attention is also dedicated to new therapies proposed for these kinds of pathologies. Awareness of these scientific problems is important for experts of various disciplines who are confronted with these kinds of presentations daily.
Collapse
Affiliation(s)
- Montserrat Fernández-Guarino
- Dermatology Service, Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis), 28034 Madrid, Spain;
| | - Maria Luisa Hernández-Bule
- Bioelectromagnetic Lab, Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis), 28034 Madrid, Spain;
| | - Stefano Bacci
- Research Unit of Histology and Embriology, Department of Biology, University of Florence, Viale Pieraccini 6, 50134 Firenze, Italy
| |
Collapse
|
20
|
Son B, Kim M, Won H, Jung A, Kim J, Koo Y, Lee NK, Baek SH, Han U, Park CG, Shin H, Gweon B, Joo J, Park HH. Secured delivery of basic fibroblast growth factor using human serum albumin-based protein nanoparticles for enhanced wound healing and regeneration. J Nanobiotechnology 2023; 21:310. [PMID: 37658367 PMCID: PMC10474766 DOI: 10.1186/s12951-023-02053-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/05/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Basic fibroblast growth factor (bFGF) is one of the critical components accelerating angiogenesis and tissue regeneration by promoting the migration of dermal fibroblasts and endothelial cells associated with matrix formation and remodeling in wound healing process. However, clinical applications of bFGF are substantially limited by its unstable nature due to rapid decomposition under physiological microenvironment. RESULTS In this study, we present the bFGF-loaded human serum albumin nanoparticles (HSA-bFGF NPs) as a means of enhanced stability and sustained release platform during tissue regeneration. Spherical shape of the HSA-bFGF NPs with uniform size distribution (polydispersity index < 0.2) is obtained via a simple desolvation and crosslinking process. The HSA-bFGF NPs securely load and release the intact soluble bFGF proteins, thereby significantly enhancing the proliferation and migration activity of human dermal fibroblasts. Myofibroblast-related genes and proteins were also significantly down-regulated, indicating decrease in risk of scar formation. Furthermore, wound healing is accelerated while achieving a highly organized extracellular matrix and enhanced angiogenesis in vivo. CONCLUSION Consequently, the HSA-bFGF NPs are suggested not only as a delivery vehicle but also as a protein stabilizer for effective wound healing and tissue regeneration.
Collapse
Affiliation(s)
- Boram Son
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Minju Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Hyosub Won
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Ara Jung
- Department of Mechanical Engineering, Sejong University, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jihyun Kim
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Yonghoe Koo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Na Kyeong Lee
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University (SKKU), Suwon, Republic of Korea
| | - Seung-Ho Baek
- Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan, Korea
| | - Uiyoung Han
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, CA, USA
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea
| | - Bomi Gweon
- Department of Mechanical Engineering, Sejong University, Seoul, Republic of Korea.
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea.
- Materials Research Science and Engineering Center, University of California, San Diego, La Jolla, United States.
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Republic of Korea.
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Knoedler S, Broichhausen S, Guo R, Dai R, Knoedler L, Kauke-Navarro M, Diatta F, Pomahac B, Machens HG, Jiang D, Rinkevich Y. Fibroblasts - the cellular choreographers of wound healing. Front Immunol 2023; 14:1233800. [PMID: 37646029 PMCID: PMC10461395 DOI: 10.3389/fimmu.2023.1233800] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Injuries to our skin trigger a cascade of spatially- and temporally-synchronized healing processes. During such endogenous wound repair, the role of fibroblasts is multifaceted, ranging from the activation and recruitment of innate immune cells through the synthesis and deposition of scar tissue to the conveyor belt-like transport of fascial connective tissue into wounds. A comprehensive understanding of fibroblast diversity and versatility in the healing machinery may help to decipher wound pathologies whilst laying the foundation for novel treatment modalities. In this review, we portray the diversity of fibroblasts and delineate their unique wound healing functions. In addition, we discuss future directions through a clinical-translational lens.
Collapse
Affiliation(s)
- Samuel Knoedler
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Sonja Broichhausen
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Trauma Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Ruiji Guo
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Ruoxuan Dai
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Leonard Knoedler
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Martin Kauke-Navarro
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Fortunay Diatta
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Bohdan Pomahac
- Division of Plastic Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Hans-Guenther Machens
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| |
Collapse
|
22
|
van Beijnum H, Koopmans T, Tomasso A, Disela V, Te Lindert S, Bakkers J, Alemany A, Berezikov E, Bartscherer K. Spatial transcriptomics reveals asymmetric cellular responses to injury in the regenerating spiny mouse ( Acomys) ear. Genome Res 2023; 33:1424-1437. [PMID: 37726147 PMCID: PMC10547259 DOI: 10.1101/gr.277538.122] [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: 11/25/2022] [Accepted: 07/19/2023] [Indexed: 09/21/2023]
Abstract
In contrast to other mammals, the spiny mouse (Acomys) regenerates skin and ear tissue, which includes hair follicles, glands, and cartilage, in a scar-free manner. Ear punch regeneration is asymmetric with only the proximal wound side participating in regeneration. Here, we show that cues originating from the proximal side are required for normal regeneration and use spatially resolved transcriptomics (tomo-seq) to understand the molecular and cellular events underlying this process. Analyzing gene expression across the ear and comparing expression modules between proximal and distal wound sides, we identify asymmetric gene expression patterns and pinpoint regenerative processes in space and time. Moreover, using a comparative approach with nonregenerative rodents (Mus, Meriones), we strengthen a hypothesis in which particularities in the injury-induced immune response may be one of the crucial determinants for why spiny mice regenerate whereas their relatives do not. Our data are available in SpinyMine, an easy-to-use and expandable web-based tool for exploring Acomys regeneration-associated gene expression.
Collapse
Affiliation(s)
- Henriëtte van Beijnum
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), 3584CT Utrecht, The Netherlands
- Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Tim Koopmans
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), 3584CT Utrecht, The Netherlands
- Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Antonio Tomasso
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), 3584CT Utrecht, The Netherlands
- Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Vanessa Disela
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), 3584CT Utrecht, The Netherlands
- Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Severin Te Lindert
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), 3584CT Utrecht, The Netherlands
- Wageningen University, Wageningen, 6708WE, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), 3584CT Utrecht, The Netherlands
- University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Anna Alemany
- Department of Anatomy and Embryology, Leiden University Medical Center, and the Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden node, 2300RC Leiden, The Netherlands
| | - Eugene Berezikov
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Kerstin Bartscherer
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), 3584CT Utrecht, The Netherlands;
- Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| |
Collapse
|
23
|
Ruksiriwanich W, Linsaenkart P, Muangsanguan A, Sringarm K, Jantrawut P, Arjin C, Sommano SR, Phimolsiripol Y, Barba FJ. Wound Healing Effect of Supercritical Carbon Dioxide Datura metel L. Leaves Extracts: An In Vitro Study of Anti-Inflammation, Cell Migration, MMP-2 Inhibition, and the Modulation of the Sonic Hedgehog Pathway in Human Fibroblasts. PLANTS (BASEL, SWITZERLAND) 2023; 12:2546. [PMID: 37447107 DOI: 10.3390/plants12132546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
Datura metel L. (thorn apple) has been used in Thai folk wisdom for wound care. In this study, we chose supercritical carbon dioxide extraction (scCO2) to develop crude extraction from the leaves of the thorn apple. The phytochemical profiles were observed using liquid chromatography-quadrupole time-of-flight mass spectrometry (LC-QTOF-MS). The biological activities of D. metel were performed through antioxidant assays, anti-inflammation based on the Griess reaction, the migration assay, the expression of matrix metalloproteinase-2 (MMP-2), and regulatory genes in fibroblasts. Dm1 and Dm2 extracts were obtained from scCO2 procedures at different pressures of 300 and 500 bar, respectively. Bioactive compounds, including farnesyl acetone, schisanhenol B, and loliolide, were identified in both extracts. The antioxidant properties of both D. metel extracts were comparable to those of l-ascorbic acid in hydrogen peroxide-induced fibroblasts with no significant difference. Additionally, Dm1 and Dm2 significantly inhibited the nitrite production levels of 1.23 ± 0.19 and 1.52 ± 0.05 μM, respectively, against the lipopolysaccharide-treated group (3.82 ± 0.39 μM). Interestingly, Dm1 obviously demonstrated the percentage of wound closure with 58.46 ± 7.61 and 82.62 ± 6.66% after 36 and 48 h of treatment, which were comparable to the commercial deproteinized dialysate from the calf blood extract. Moreover, both extracts were comparable to l-ascorbic acid treatment in their ability to suppress the expression of MMP-2: an enzyme that breaks down collagen. The gene expressions of SHH, SMO, and GLI1 that control the sonic hedgehog pathway were also clearly upregulated by Dm1. Consequently, the scCO2 technique could be applied in D. metel extraction and contribute to potentially effective wound closure.
Collapse
Affiliation(s)
- Warintorn Ruksiriwanich
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Cluster of Valorization and Bio-Green Transformation for Translation Research Innovation of Raw Materials and Products, Chiang Mai University, Chiang Mai 50200, Thailand
- Cluster of Agro Bio-Circular-Green Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Pichchapa Linsaenkart
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Anurak Muangsanguan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Korawan Sringarm
- Cluster of Valorization and Bio-Green Transformation for Translation Research Innovation of Raw Materials and Products, Chiang Mai University, Chiang Mai 50200, Thailand
- Cluster of Agro Bio-Circular-Green Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pensak Jantrawut
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Cluster of Valorization and Bio-Green Transformation for Translation Research Innovation of Raw Materials and Products, Chiang Mai University, Chiang Mai 50200, Thailand
- Cluster of Agro Bio-Circular-Green Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Chaiwat Arjin
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sarana Rose Sommano
- Cluster of Valorization and Bio-Green Transformation for Translation Research Innovation of Raw Materials and Products, Chiang Mai University, Chiang Mai 50200, Thailand
- Cluster of Agro Bio-Circular-Green Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Yuthana Phimolsiripol
- Cluster of Agro Bio-Circular-Green Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
- School of Agro-Industry, Faculty of Agro-Industry, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Francisco J Barba
- Department of Preventive Medicine and Public Health, Food Science, Toxicology and Forensic Medicine, Faculty of Pharmacy, University of Valencia, Burjassot, 46100 Valencia, Spain
| |
Collapse
|
24
|
Ansari L, Mashayekhi-Sardoo H, Baradaran Rahimi V, Yahyazadeh R, Ghayour-Mobarhan M, Askari VR. Curcumin-based nanoformulations alleviate wounds and related disorders: A comprehensive review. Biofactors 2023; 49:736-781. [PMID: 36961254 DOI: 10.1002/biof.1945] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/24/2023] [Indexed: 03/25/2023]
Abstract
Despite numerous advantages, curcumin's (CUR) low solubility and low bioavailability limit its employment as a free drug. CUR-incorporated nanoformulation enhances the bioavailability and angiogenesis, collagen deposition, fibroblast proliferation, reepithelization, collagen synthesis, neovascularization, and granulation tissue formation in different wounds. Designing nanoformulations with controlled-release properties ensure the presence of CUR in the defective area during treatment. Different nanoformulations encompassing nanofibers, nanoparticles (NPs), nanospray, nanoemulsion, nanosuspension, nanoliposome, nanovesicle, and nanomicelle were described in the present study comprehensively. Moreover, for some other systems which contain nano-CUR or CUR nanoformulations, including some nanofibers, films, composites, scaffolds, gel, and hydrogels seems the CUR-loaded NPs incorporation has better control of the sustained release, and thereby, the presence of CUR until the final stages of wound healing is more possible. Incorporating CUR-loaded chitosan NPs into nanofiber increased the release time, while 80% of CUR was released during 240 h (10 days). Therefore, this system can guarantee the presence of CUR during the entire healing period. Furthermore, porous structures such as sponges, aerogels, some hydrogels, and scaffolds disclosed promising performance. These architectures with interconnected pores can mimic the native extracellular matrix, thereby facilitating attachment and infiltration of cells at the wound site, besides maintaining a free flow of nutrients and oxygen within the three-dimensional structure essential for rapid and proper wound healing, as well as enhancing mechanical strength.
Collapse
Affiliation(s)
- Legha Ansari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Roghayeh Yahyazadeh
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
25
|
Hernández-Bule ML, Toledano-Macías E, Pérez-González LA, Martínez-Pascual MA, Fernández-Guarino M. Anti-Fibrotic Effects of RF Electric Currents. Int J Mol Sci 2023; 24:10986. [PMID: 37446165 DOI: 10.3390/ijms241310986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Hypertrophic scars and keloids are two different manifestations of excessive dermal fibrosis and are caused by an alteration in the normal wound-healing process. Treatment with radiofrequency (RF)-based therapies has proven to be useful in reducing hypertrophic scars. In this study, the effect of one of these radiofrequency therapies, Capacitive Resistive Electrical Transfer Therapy (CRET) on biomarkers of skin fibrosis was investigated. For this, in cultures of human myofibroblasts treated with CRET therapy or sham-treated, proliferation (XTT Assay), apoptosis (TUNEL Assay), and cell migration (Wound Closure Assay) were analyzed. Furthermore, in these cultures the expression and/or localization of extracellular matrix proteins such as α-SMA, Col I, Col III (immunofluorescence), metalloproteinases MMP1 and MMP9, MAP kinase ERK1/2, and the transcription factor NFκB were also investigated (immunoblot). The results have revealed that CRET decreases the expression of extracellular matrix proteins, modifies the expression of the metalloproteinase MMP9, and reduces the activation of NFκB with respect to controls, suggesting that this therapy could be useful for the treatment of fibrotic pathologies.
Collapse
Affiliation(s)
- María Luisa Hernández-Bule
- Bioelectromagnetic Laboratory, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis), Carretera de Colmenar Viejo, km. 9.100, 28034 Madrid, Spain
| | - Elena Toledano-Macías
- Bioelectromagnetic Laboratory, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis), Carretera de Colmenar Viejo, km. 9.100, 28034 Madrid, Spain
| | - Luis Alfonso Pérez-González
- Dermatology Service, Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis), Carretera de Colmenar Viejo, km. 9.100, 28034 Madrid, Spain
| | - María Antonia Martínez-Pascual
- Bioelectromagnetic Laboratory, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis), Carretera de Colmenar Viejo, km. 9.100, 28034 Madrid, Spain
| | - Montserrat Fernández-Guarino
- Dermatology Service, Hospital Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (Irycis), Carretera de Colmenar Viejo, km. 9.100, 28034 Madrid, Spain
| |
Collapse
|
26
|
Roman J. Fibroblasts-Warriors at the Intersection of Wound Healing and Disrepair. Biomolecules 2023; 13:945. [PMID: 37371525 DOI: 10.3390/biom13060945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/07/2023] [Accepted: 05/17/2023] [Indexed: 06/29/2023] Open
Abstract
Wound healing is triggered by inflammation elicited after tissue injury. Mesenchymal cells, specifically fibroblasts, accumulate in the injured tissues, where they engage in tissue repair through the expression and assembly of extracellular matrices that provide a scaffold for cell adhesion, the re-epithelialization of tissues, the production of soluble bioactive mediators that promote cellular recruitment and differentiation, and the regulation of immune responses. If appropriately deployed, these processes promote adaptive repair, resulting in the preservation of the tissue structure and function. Conversely, the dysregulation of these processes leads to maladaptive repair or disrepair, which causes tissue destruction and a loss of organ function. Thus, fibroblasts not only serve as structural cells that maintain tissue integrity, but are key effector cells in the process of wound healing. The review will discuss the general concepts about the origins and heterogeneity of this cell population and highlight the specific fibroblast functions disrupted in human disease. Finally, the review will explore the role of fibroblasts in tissue disrepair, with special attention to the lung, the role of aging, and how alterations in the fibroblast phenotype underpin disorders characterized by pulmonary fibrosis.
Collapse
Affiliation(s)
- Jesse Roman
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care and The Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
27
|
Chen M, Xu Z, Chen Y, Yang Q, Lu R, Dong Y, Li X, Xie J, Xu R, Jia H, Kang Y, Wu Y. EGFR marks a subpopulation of dermal mesenchymal cells highly expressing IGF1 which enhances hair follicle regeneration. J Cell Mol Med 2023; 27:1697-1707. [PMID: 37165726 PMCID: PMC10273066 DOI: 10.1111/jcmm.17766] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/14/2023] [Accepted: 04/28/2023] [Indexed: 05/12/2023] Open
Abstract
The skin harbours transcriptionally and functionally heterogeneous mesenchymal cells that participate in various physiological activities by secreting biochemical cues. In this study, we aimed to identify a new subpopulation of dermal mesenchymal cells that enhance hair follicle regeneration through a paracrine mechanism. Integrated single-cell RNA sequencing (scRNA-seq) data analysis revealed epidermal growth factor receptor (EGFR) as a marker of distinct fibroblast subpopulation in the neonatal murine dermis. Immunofluorescence staining and fluorescence-activated cell sorting (FACS) were used to validate the existence of the cell population in Krt14-rtTA-H2BGFP mouse. The difference of gene expression between separated cell subpopulation was examined by real-time PCR. Potential effect of the designated factor on hair follicle regeneration was observed after the application on excisional wounds in Krt14-rtTA-H2BGFP mouse. Immunofluorescence staining demonstrated the existence of dermal EGFR+ cells in neonatal and adult mouse dermis. The EGFR+ mesenchymal population, sorted by FACS, displayed a higher expression level of Igf1 (insulin-like growth factor 1). Co-localisation of IGF1 with EGFR in the mouse dermis and upregulated numbers of hair follicles in healed wounds following the application of exogenous IGF1 illustrated the contribution of EGFR+ cells in promoting wound-induced hair follicle neogenesis. Our results indicate that EGFR identifies a subpopulation of dermal fibroblasts that contribute to IGF1 promotion of hair follicle neogenesis. It broadens the understanding of heterogeneity and the mesenchymal cell function in skin and may facilitate the potential translational application of these cells.
Collapse
Affiliation(s)
- Min Chen
- Tsinghua‐Berkeley Shenzhen InstituteTsinghua UniversityShenzhenChina
- State Key Laboratory of Chemical Oncogenomics, and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate SchoolTsinghua UniversityShenzhenChina
| | - Zaoxu Xu
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Yu Chen
- State Key Laboratory of Chemical Oncogenomics, and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate SchoolTsinghua UniversityShenzhenChina
| | - Qingyang Yang
- Tsinghua‐Berkeley Shenzhen InstituteTsinghua UniversityShenzhenChina
| | - Ruiqing Lu
- State Key Laboratory of Chemical Oncogenomics, and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate SchoolTsinghua UniversityShenzhenChina
| | - Yankai Dong
- State Key Laboratory of Chemical Oncogenomics, and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate SchoolTsinghua UniversityShenzhenChina
| | - Xiaosong Li
- State Key Laboratory of Chemical Oncogenomics, and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate SchoolTsinghua UniversityShenzhenChina
| | - Jundong Xie
- State Key Laboratory of Chemical Oncogenomics, and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate SchoolTsinghua UniversityShenzhenChina
| | - Ren‐He Xu
- Faculty of Health SciencesUniversity of MacauTaipaChina
| | | | - Yan Kang
- Shanghai Jahwa United Co., LtdShanghaiChina
| | - Yaojiong Wu
- Tsinghua‐Berkeley Shenzhen InstituteTsinghua UniversityShenzhenChina
- State Key Laboratory of Chemical Oncogenomics, and the Institute of Biopharmaceutical and Health Engineering (iBHE), Shenzhen International Graduate SchoolTsinghua UniversityShenzhenChina
| |
Collapse
|
28
|
Vorstandlechner V, Copic D, Klas K, Direder M, Golabi B, Radtke C, Ankersmit HJ, Mildner M. The Secretome of Irradiated Peripheral Mononuclear Cells Attenuates Hypertrophic Skin Scarring. Pharmaceutics 2023; 15:pharmaceutics15041065. [PMID: 37111549 PMCID: PMC10143262 DOI: 10.3390/pharmaceutics15041065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Hypertrophic scars can cause pain, movement restrictions, and reduction in the quality of life. Despite numerous options to treat hypertrophic scarring, efficient therapies are still scarce, and cellular mechanisms are not well understood. Factors secreted by peripheral blood mononuclear cells (PBMCsec) have been previously described for their beneficial effects on tissue regeneration. In this study, we investigated the effects of PBMCsec on skin scarring in mouse models and human scar explant cultures at single-cell resolution (scRNAseq). Mouse wounds and scars, and human mature scars were treated with PBMCsec intradermally and topically. The topical and intradermal application of PBMCsec regulated the expression of various genes involved in pro-fibrotic processes and tissue remodeling. We identified elastin as a common linchpin of anti-fibrotic action in both mouse and human scars. In vitro, we found that PBMCsec prevents TGFβ-mediated myofibroblast differentiation and attenuates abundant elastin expression with non-canonical signaling inhibition. Furthermore, the TGFβ-induced breakdown of elastic fibers was strongly inhibited by the addition of PBMCsec. In conclusion, we conducted an extensive study with multiple experimental approaches and ample scRNAseq data demonstrating the anti-fibrotic effect of PBMCsec on cutaneous scars in mouse and human experimental settings. These findings point at PBMCsec as a novel therapeutic option to treat skin scarring.
Collapse
Affiliation(s)
- Vera Vorstandlechner
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dragan Copic
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna, 1090 Vienna, Austria
| | - Katharina Klas
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Martin Direder
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
- Department of Orthopedics and Trauma-Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Bahar Golabi
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
| | - Christine Radtke
- Department of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Hendrik J. Ankersmit
- Laboratory for Cardiac and Thoracic Diagnosis, Regeneration and Applied Immunology, Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
- Aposcience AG, 1200 Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence:
| |
Collapse
|
29
|
Iannello G, Patel A, Sirabella D, Diaz AG, Hoover BN, Sarmah H, Corneo B. Simple, Fast, and Efficient Method for Derivation of Dermal Fibroblasts From Skin Biopsies. Curr Protoc 2023; 3:e714. [PMID: 36912580 DOI: 10.1002/cpz1.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Primary fibroblasts are a precious resource in the field of translational regenerative medicine. Dermal fibroblasts derived from human subject biopsies are being used as donor tissues for the derivation of patient-specific iPSC lines, which in turn are used for disease modeling, drug screening, tissue engineering, and cell transplantation. We developed a fast and simple protocol to grow dermal fibroblasts from skin biopsies. Using this protocol, we simply and firmly fix the biopsy piece on the surface of a tissue culture-treated plate and allow the fibroblasts to grow. This novel method eliminates any need for enzymatic digestion or mechanical dissociation of the biopsy piece. By using this newly developed protocol, we have successfully established around 100 fibroblast lines characterized by the expression of specific markers [Serpin H1 (Hsp-47), F-actin, and Vimentin]. Finally, we have used many of these fibroblast lines as donor tissues to successfully derive iPSC lines. We have developed a method that is simple, fast, convenient, efficient, and gentle on the cells to derive dermal fibroblasts from human skin biopsies. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Skin biopsy collection and fibroblast derivation Support Protocol 1: Culturing, freezing, and thawing dermal fibroblasts derived from a skin biopsy Support Protocol 2: Characterization of dermal fibroblasts by immunocytochemistry.
Collapse
Affiliation(s)
- Grazia Iannello
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, New York, New York
| | | | - Dario Sirabella
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, New York, New York
| | | | - Benjamin N Hoover
- Department of Neurology, Eleanor and Lou Gehrig ALS Center, Columbia University Irving Medical Center, New York, New York
| | - Hemanta Sarmah
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, New York, New York
| | - Barbara Corneo
- Columbia Stem Cell Initiative, Stem Cell Core, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
30
|
Li S, Zhao Z, Li Q, Li J, Pang Y. Lamprey Wound Healing and Regenerative Effects: The Collaborative Efforts of Diverse Drivers. Int J Mol Sci 2023; 24:ijms24043213. [PMID: 36834626 PMCID: PMC9965152 DOI: 10.3390/ijms24043213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/08/2023] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
Skin is a natural barrier between the body and the external environment, and this important multifunctional organ plays roles in body temperature regulation, sensory stimulation, mucus secretion, metabolite excretion and immune defense. Lampreys, as ancient vertebrates, rarely experience infection of damaged skin during farming and efficiently promote skin wound healing. However, the mechanism underlying these wound healing and regenerative effects is unclear. Our histology and transcriptomics results demonstrate that lampreys regenerate a nearly complete skin structure in damaged epidermis, including the secretory glands, and will almost not be infected, even if experiencing full-thickness damage. In addition, ATGL, DGL and MGL participate in the lipolysis process to provide space for infiltrating cells. A large number of red blood cells migrate to the site of injury and exert proinflammatory effects, upregulating the expression of proinflammatory factors such as IL-8 and IL-17. Based on a lamprey skin damage healing model, adipocytes and red blood cells in the subcutaneous fat layer can promote wound healing, which provides a new approach for the study of skin healing mechanisms. Transcriptome data reveal that mechanical signal transduction pathways are mainly regulated by focal adhesion kinase and that the actin cytoskeleton plays an important role in the healing of lamprey skin injuries. We identified RAC1 as a key regulatory gene that is necessary and partially sufficient for wound regeneration. Insights into the mechanisms of lamprey skin injury and healing will provide a theoretical basis for overcoming the challenges associated with chronic healing and scar healing in the clinic.
Collapse
Affiliation(s)
- Shushen Li
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| | - Zhiyuan Zhao
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| | - Qingwei Li
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
| | - Jun Li
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
- Correspondence: (J.L.); (Y.P.)
| | - Yue Pang
- College of Life Sciences, Liaoning Normal University, Dalian 116081, China
- Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
- Correspondence: (J.L.); (Y.P.)
| |
Collapse
|
31
|
Neves LMG, Wilgus TA, Bayat A. In Vitro, Ex Vivo, and In Vivo Approaches for Investigation of Skin Scarring: Human and Animal Models. Adv Wound Care (New Rochelle) 2023; 12:97-116. [PMID: 34915768 DOI: 10.1089/wound.2021.0139] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Significance: The cutaneous repair process naturally results in different types of scarring that are classified as normal or pathological. Affected individuals are often affected from an esthetic, physical (functional), and psychosocial perspective. The distinct nature of scarring in humans, particularly the formation of pathological scars, makes the study of skin scarring a challenge for researchers in this area. Several established experimental models exist for studying scar formation. However, the increasing development and validation of newly emerging models have made it possible to carry out studies focused on different variables that influence this unique process. Recent Advances: Experimental models such as in vitro, ex vivo, and in vivo models have obtained different degrees of success in the reproduction of the scar formation in its native milieu and true environment. These models also differ in their ability to elucidate the molecular, cellular, and structural mechanisms involved in scarring, as well as for testing new agents and approaches for therapies. The models reviewed here, including cells derived from human skin and in vivo animal models, have contributed to the advancement of skin scarring research. Critical Issues and Future Directions: The absence of experimental models that faithfully reproduce the typical characteristics of the different types of human skin scars makes the improvement of validated models and the establishment of new ones a critical unmet need. The fields of wound healing research combined with tissue engineering have offered newer alternatives for experimental studies with the potential to provide clinically useful knowledge about scar formation.
Collapse
Affiliation(s)
- Lia M G Neves
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom
| | - Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, Ohio, USA
| | - Ardeshir Bayat
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom.,Medical Research Council (MRC) Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
32
|
Ma Y, Liu Z, Miao L, Jiang X, Ruan H, Xuan R, Xu S. Mechanisms underlying pathological scarring by fibroblasts during wound healing. Int Wound J 2023. [PMID: 36726192 DOI: 10.1111/iwj.14097] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Pathological scarring is an abnormal outcome of wound healing, which often manifests as excessive proliferation and transdifferentiation of fibroblasts (FBs), and excessive deposition of the extracellular matrix. FBs are the most important effector cells involved in wound healing and scar formation. The factors that promote pathological scar formation often act on the proliferation and function of FB. In this study, we describe the factors that lead to abnormal FB formation in pathological scarring in terms of the microenvironment, signalling pathways, epigenetics, and autophagy. These findings suggest that understanding the causes of abnormal FB formation may aid in the development of precise and effective preventive and treatment strategies for pathological scarring that are associated with improved quality of life of patients.
Collapse
Affiliation(s)
- Yizhao Ma
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Zhifang Liu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - LinLin Miao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Xinyu Jiang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Hongyu Ruan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Rongrong Xuan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Suling Xu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
33
|
Shen Y, Ning J, Zhao L, Liu W, Wang T, Yu J, Wang Y. Matrix remodeling associated 7 proteins promote cutaneous wound healing through vimentin in coordinating fibroblast functions. Inflamm Regen 2023; 43:5. [PMID: 36647132 PMCID: PMC9841631 DOI: 10.1186/s41232-023-00256-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Abstract
Wound healing depends largely on the remodeling of the extracellular matrix around and reorganization of tissue-resident cells. Matrix remodeling associated 7 (MXRA7) is a member of the matrix remodeling-associated gene family and is involved in matrix remodeling-associated processes, such as inflammatory neovascularization, liver injury, and autoimmune skin disease. To investigate whether and how MXRA7 participate in cutaneous wound healing, an ear-punching model was utilized in wild-type (WT) and MXRA7-deficient mice, and the dermal fibroblasts from these mice were further studied in vitro. Results showed that the MXRA7 deficiency impaired the wound healing process in mice. Quantitative PCR indicated that lack of MXRA7 impaired the expression of several extracellular matrix genes (e.g., MMP-2) and inhibited signaling pathways (e.g., STAT3) in healing ear tissues. In in vitro culture system, migration, contraction, or proliferation of fibroblasts was impaired upon MXRA7 deficiency. Pull-down and mass spectrum assay revealed that vimentin was among the proteins that bound MXRA7 proteins in cells, and further investigations indicate MXRA7 was an autocrine factor in fibroblasts that involved vimentin in certain ways, such as JNK and STAT3/STAT5 signaling pathways in our study. In conclusion, MXRA7 proteins promote wound healing through vimentin in coordinating fibroblast functions.
Collapse
Affiliation(s)
- Ying Shen
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Jinling Ning
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Lu Zhao
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Wei Liu
- grid.429222.d0000 0004 1798 0228Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006 China
| | - Ting Wang
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Jie Yu
- grid.429222.d0000 0004 1798 0228Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006 China
| | - Yiqiang Wang
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China ,grid.12955.3a0000 0001 2264 7233Eye Institute of Xiamen University, Xiamen University, Xiamen, 361104 China ,grid.440701.60000 0004 1765 4000Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou, 215123 China
| |
Collapse
|
34
|
Wood FM. The Role of Cell-Based Therapies in Acute Burn Wound Skin Repair: A Review. J Burn Care Res 2023; 44:S42-S47. [PMID: 36567469 DOI: 10.1093/jbcr/irac146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tissue engineering solutions for skin have been developed over the last few decades with a focus initially on a two-layered structure with epithelial and dermal repair. An essential element of skin restoration is a source of cells capable of differentiating into the appropriate phenotype. The need to repair areas of skin when traditional techniques were not adequate addressed led to cell based therapies being developed initially as a laboratory-based tissue expansion opportunity, both as sheets of cultured epithelial autograft and in composite laboratory-based skin substitutes. The time to availability of the cell-based therapies has been solved in a number of ways, from using allograft cell-based solutions to the use of point of care skin cell harvesting for immediate clinical use. More recently pluripotential cells have been explored providing a readily available source of cells and cells which can express the broad range of phenotypes seen in the mature skin construct. The lessons learnt from the use of cell based techniques has driven the exploration of the use of 3D printing technology, with controlled accurate placement of the cells within a specific printed construct to optimise the phenotypic expression and tissue generation.
Collapse
Affiliation(s)
- Fiona M Wood
- University of Western Australia, Fiona Stanley Hospital, Perth Children's Hospital, Burns Service of WA, Level 4 Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch Western, Australia 6150
| |
Collapse
|
35
|
Cooke JP, Lai L. Role of angiogenic transdifferentiation in vascular recovery. Front Cardiovasc Med 2023; 10:1155835. [PMID: 37200975 PMCID: PMC10187761 DOI: 10.3389/fcvm.2023.1155835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/10/2023] [Indexed: 05/20/2023] Open
Abstract
Tissue repair requires the orchestration of multiple processes involving a multiplicity of cellular effectors, signaling pathways, and cell-cell communication. The regeneration of the vasculature is a critical process for tissue repair and involves angiogenesis, adult vasculogenesis, and often arteriogenesis, which processes enable recovery of perfusion to deliver oxygen and nutrients to the repair or rebuild of the tissue. Endothelial cells play a major role in angiogenesis, whereas circulating angiogenic cells (primarily of hematopoietic origin) participate in adult vasculogenesis, and monocytes/macrophages have a defining role in the vascular remodeling that is necessary for arteriogenesis. Tissue fibroblasts participate in tissue repair by proliferating and generating the extracellular matrix as the structural scaffold for tissue regeneration. Heretofore, fibroblasts were not generally believed to be involved in vascular regeneration. However, we provide new data indicating that fibroblasts may undergo angiogenic transdifferentiation, to directly expand the microvasculature. Transdifferentiation of fibroblasts to endothelial cells is initiated by inflammatory signaling which increases DNA accessibility and cellular plasticity. In the environment of under-perfused tissue, the activated fibroblasts with increased DNA accessibility can now respond to angiogenic cytokines, which provide the transcriptional direction to induce fibroblasts to become endothelial cells. Periphery artery disease (PAD) involves the dysregulation of vascular repair and inflammation. Understanding the relationship between inflammation, transdifferentiation, and vascular regeneration may lead to a new therapeutic approach to PAD.
Collapse
|
36
|
Li Y, Liu D, Tan F, Yin W, Li Z. Umbilical cord derived mesenchymal stem cell-GelMA microspheres for accelerated wound healing. Biomed Mater 2022; 18. [PMID: 36541452 DOI: 10.1088/1748-605x/aca947] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are an ideal seed cell for tissue engineering and stem cell transplantation. MSCs combined with biological scaffolds play an important role in promoting the repair of cutaneous wound. However, direct administration of MSCs is challenging for MSCs survival and integration into tissues. Providing MSCs with a biocompatible scaffold can improve MSCs survival, but the effect of gelatin methacrylate (GelMA) loaded MSCs from umbilical cord MSCs (UC-MSCs) in wound healing remains unknown. Here, we investigated the ability of GelMA with UC-MSCs complexes to promote migration and proliferation and the effect on wound healing in mouse models. We discovered that UC-MSCs attached to GelMA and promoted the proliferation and migration of fibroblasts. Both UC-MSCs and UC-MSCs-derived extracellular vesicles accelerated wound healing. MSC + Gelatin methacrylate microspheres (GMs) application decreased expression of transforming growth factor-β(TGF-β) and Type III collagen (Col3)in vivo, leading to new collagen deposition and angiogenesis, and accelerate wound healing and skin tissue regeneration. Taken together, these findings indicate MSC + GMs can promote wound healing by regulating wound healing-related factors in the paracrine. Therefore, our research proves that GelMA is an ideal scaffold for the top management of UC-MSCs in wound healing medical practice.
Collapse
Affiliation(s)
- Yanqun Li
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Dongyu Liu
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Fengming Tan
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Wenling Yin
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Zhihuan Li
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| |
Collapse
|
37
|
Trentini M, Zanolla I, Zanotti F, Tiengo E, Licastro D, Dal Monego S, Lovatti L, Zavan B. Apple Derived Exosomes Improve Collagen Type I Production and Decrease MMPs during Aging of the Skin through Downregulation of the NF-κB Pathway as Mode of Action. Cells 2022; 11:3950. [PMID: 36552714 PMCID: PMC9776931 DOI: 10.3390/cells11243950] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Skin ageing is strictly related to chronic inflammation of the derma and the decay of structural proteins of the extracellular matrix. Indeed, it has become common practice to refer to this phenomenon as inflammageing. Biotech innovation is always in search of new active principles that induce a youthful appearance. In this paper, apple-derived nanovesicles (ADNVs) were investigated as novel anti-inflammatory compounds, which are able to alter the extracellular matrix production of dermal fibroblasts. Total RNA sequencing analysis revealed that ADNVs negatively influence the activity of Toll-like Receptor 4 (TLR4), and, thus, downregulate the NF-κB pro-inflammatory pathway. ADNVs also reduce extracellular matrix degradation by increasing collagen synthesis (COL3A1, COL1A2, COL8A1 and COL6A1) and downregulating metalloproteinase production (MMP1, MMP8 and MMP9). Topical applications for skin regeneration were evaluated by the association of ADNVs with hyaluronic-acid-based hydrogel and patches.
Collapse
Affiliation(s)
- Martina Trentini
- Department Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ilaria Zanolla
- Department Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Federica Zanotti
- Department Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Elena Tiengo
- Department Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | | | | | - Luca Lovatti
- Department Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Barbara Zavan
- Department Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
38
|
Concentration-Dependent Inhibition of Hypertrophic Scar Formation by Botulinum Toxin Type A in a Rabbit Ear Model. Aesthetic Plast Surg 2022; 46:3072-3079. [PMID: 35864206 DOI: 10.1007/s00266-022-03008-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/26/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypertrophic scar (HS), as a disappointing result of wound healing, adversely affects the patient, both physically and psychologically. Botulinum toxin type A (BTXA) has been revealed to prevent and improve HS. We conducted this study to assess the effect of different BTXA concentrations on inhibiting HS in a rabbit ear model. METHODS Eight healthy New Zealand long-eared rabbits were included in the experiment for modeling. Four wounds of 1 cm in diameter were created on both ears, which separately received an injection of a given BTXA concentration immediately after surgery. On postoperative days 40, scar tissue was obtained and subjected to hematoxylin and eosin (HE) staining for the hypertrophic index (HI) and immunohistochemical staining for CD31, Ki67, and transforming growth factor-beta 1 (TGF-β1) expression. The HI was assessed for scar proliferation, and CD31 and Ki67 expression were used to assess the effect of BTXA on angiogenesis and fibroblast proliferation, respectively. RESULTS All rabbits healed well without infection or mortality. From the HE staining, the HI showed a significant decrease with increasing BTXA concentration (p < 0.05). BTXA also inhibited angiogenesis and TGF-β1 expression in a concentration-dependent manner, with significant differences between the groups (p < 0.05). BTXA inhibited fibroblast proliferation with increasing BTXA concentration. However, there was no significant difference between the 0.5 U/0.1 ml and 0 U/0.1 ml groups (p > 0.05). CONCLUSION Immediate postoperative BTXA injection inhibited angiogenesis, fibroblast proliferation, and TGF-β1 expression in a concentration-dependent manner, thus suppressing HS formation in rabbit ears. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors https://www.springer.com/journal/00266 .
Collapse
|
39
|
Ultrastructural Characterization of Human Gingival Fibroblasts in 3D Culture. Cells 2022; 11:cells11223647. [PMID: 36429075 PMCID: PMC9688082 DOI: 10.3390/cells11223647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022] Open
Abstract
Cell spheroids are applied in various fields of research, such as the fabrication of three-dimensional artificial tissues in vitro, disease modeling, stem cell research, regenerative therapy, and biotechnology. A preclinical 3D culture model of primary human gingival fibroblasts free of external factors and/or chemical inducers is presented herein. The ultrastructure of the spheroids was characterized to establish a cellular model for the study of periodontal tissue regeneration. The liquid overlay technique was used with agarose to generate spheroids. Fibroblasts in 2D culture and cell spheroids were characterized by immunofluorescence, and cell spheroids were characterized by optical and scanning electron microscopy, energy-dispersive X-ray spectroscopy, backscattered electrons, and Fourier transform infrared spectroscopy. Ostegenic related genes were analyzed by RT-qPCR. Gingival fibroblasts formed spheroids spontaneously and showed amorphous calcium phosphate nanoparticle deposits on their surface. The results suggest that human gingival fibroblasts have an intrinsic potential to generate a mineralized niche in 3D culture.
Collapse
|
40
|
Markov PA, Vinogradov II, Kostromina E, Eremin PS, Gilmutdinova IR, Kudryashova IS, Greben A, Rachin AP, Nechaev AN. A wound dressing based on a track-etched membrane modified by a biopolymer nanoframe: physical, chemical, and biological characteristics. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
41
|
Cialdai F, Risaliti C, Monici M. Role of fibroblasts in wound healing and tissue remodeling on Earth and in space. Front Bioeng Biotechnol 2022; 10:958381. [PMID: 36267456 PMCID: PMC9578548 DOI: 10.3389/fbioe.2022.958381] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Wound healing (WH) and the role fibroblasts play in the process, as well as healing impairment and fibroblast dysfunction, have been thoroughly reviewed by other authors. We treat these topics briefly, with the only aim of contextualizing the true focus of this review, namely, the microgravity-induced changes in fibroblast functions involved in WH. Microgravity is a condition typical of spaceflight. Studying its possible effects on fibroblasts and WH is useful not only for the safety of astronauts who will face future interplanetary space missions, but also to help improve the management of WH impairment on Earth. The interesting similarity between microgravity-induced alterations of fibroblast behavior and fibroblast dysfunction in WH impairment on Earth is highlighted. The possibility of using microgravity-exposed fibroblasts and WH in space as models of healing impairment on Earth is suggested. The gaps in knowledge on fibroblast functions in WH are analyzed. The contribution that studies on fibroblast behavior in weightlessness can make to fill these gaps and, consequently, improve therapeutic strategies is considered.
Collapse
|
42
|
Psenakova Z, Smondrk M, Barabas J, Benova M, Brociek R, Wajda A, Kowol P, Coco S, Sciuto GL. Computational Analysis of a Multi-Layered Skin and Cardiac Pacemaker Model Based on Neural Network Approach. SENSORS (BASEL, SWITZERLAND) 2022; 22:6359. [PMID: 36080817 PMCID: PMC9459797 DOI: 10.3390/s22176359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 06/15/2023]
Abstract
The presented study discusses the possible disturbing effects of the electromagnetic field of antennas used in mobile phones or WiFi technologies on the pacemaker in the patient's body. This study aims to obtain information on how the thickness of skin layers (such as the thickness of the hypodermis) can affect the activity of a pacemaker exposed to a high-frequency electromagnetic field. This study describes the computational mathematical analysis and modeling of the heart pacemaker inserted under the skin exposed to various electromagnetic field sources, such as a PIFA antenna and a tuned dipole antenna. The finite integration technique (FIT) for a pacemaker model was implemented within the commercially available CST Microwave simulation software studio. Likewise, the equations that describe the mathematical relationship between the subcutaneous layer thickness and electric field according to different exposures of a tuned dipole and a PIFA antenna are used and applied for training a neural network. The main output of this study is the creation of a mathematical model and a multilayer feedforward neural network, which can show the dependence of the thickness of the hypodermis on the size of the electromagnetic field, from the simulated data from CST Studio.
Collapse
Affiliation(s)
- Zuzana Psenakova
- Department of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering, University of Zilina, Univerzitna 1, 01026 Zilina, Slovakia
| | - Maros Smondrk
- Department of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering, University of Zilina, Univerzitna 1, 01026 Zilina, Slovakia
| | - Jan Barabas
- Department of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering, University of Zilina, Univerzitna 1, 01026 Zilina, Slovakia
| | - Mariana Benova
- Department of Electromagnetic and Biomedical Engineering, Faculty of Electrical Engineering, University of Zilina, Univerzitna 1, 01026 Zilina, Slovakia
| | - Rafał Brociek
- Department of Mathematics Applications and Methods for Artificial Intelligence, Faculty of Applied Mathematics, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Agata Wajda
- Institute of Energy and Fuel Processing Technology, 41-803 Zabrze, Poland
| | - Paweł Kowol
- Department of Mechatronics, Silesian University of Technology, Akademicka 10a, 44-100 Gliwice, Poland
| | - Salvatore Coco
- Department of Electrical, Electronics and Informatics Engineering, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Grazia Lo Sciuto
- Department of Mechatronics, Silesian University of Technology, Akademicka 10a, 44-100 Gliwice, Poland
- Department of Electrical, Electronics and Informatics Engineering, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
43
|
Gefen A. The influence zone: a critical performance measure for negative pressure wound therapy systems. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2022; 31:S8-S12. [PMID: 35980923 DOI: 10.12968/bjon.2022.31.15.s8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This article provides an introduction to the theory of, what is termed, the 'influence zone' in the context of negative pressure wound therapy (NPWT). It is a quantitative bioengineering performance measure for NPWT systems, to indicate their effectiveness, namely, how far from the wound bed edges a specific system is able to deliver effective mechano-stimulation into the periwound, and at which intensity. The influence zone therefore provides objective and standardised metrics of one of the fundamental modes of action of NPWT systems: the ability to effectively and optimally deform both the wound and periwound macroscopically and microscopically. Most important is the mechanical deformation of the periwound area to activate cells responsible for tissue repair, particularly (myo)fibroblasts. Notably, the influence zone must extend sufficiently into the periwound to stimulate (myo)fibroblasts in order that they migrate and progress the wound healing process, facilitating the formation of scar tissue, without overstretching the periwound tissues so as not cause or escalate further cell and tissue damage. The inclusion of the influence zone theory within research to investigate the efficacy of NPWT systems facilitates systematic comparisons of commercially available and potentially new systems. This approach has the capacity to guide not only research and development work, but also clinical decision-making. Recently published research found that inducing an effective influence zone first and foremost requires continuous delivery of the intended pressure to the wound bed.
Collapse
Affiliation(s)
- Amit Gefen
- Professor of Biomedical Engineering, The Herbert J Berman Chair in Vascular Bioengineering, Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Israel
| |
Collapse
|
44
|
Orlov A, Gefen A. Effective negative pressure wound therapy for open wounds: The importance of consistent pressure delivery. Int Wound J 2022; 20:328-344. [PMID: 35818745 PMCID: PMC9885467 DOI: 10.1111/iwj.13879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/14/2022] [Indexed: 02/03/2023] Open
Abstract
Two distinct design concepts exist for single-use negative pressure wound therapy systems: Canister-based versus canisterless. The canister-based technology provides intrinsic stable delivery of the intended negative pressure, because exudate is constantly transferred from the wound into a canister, thereby preventing dressing saturation. In contrast, with a canisterless system, where delivery of the negative pressure depends on continuous evaporation of wound fluids from its dressing, loss of the intended wound-bed pressure may occur due to dressing saturation. To investigate whether these two designs differ in their mechanobiological effect with respect to magnitudes and distributions of tissue strain fields under the absorptive dressing, termed the influence zone, we integrated computational modelling with an animal study. This influence zone must be of biologically influential strain levels and extend sufficiently into the peri-wound for stimulating fibroblasts to migrate and progress the healing. We found that an effective influence zone requires continuous delivery of the intended pressure to the wound-bed. Loss of negative pressure at the wound-bed below 40 mmHg adversely lowered the peri-wound stimulation around a 120 × 70 mm sized wound to less than one-third of the baseline stimulation, and further pressure decreases to 20 mmHg or lower resulted in complete lack of peri-wound mechano-stimulation.
Collapse
Affiliation(s)
- Aleksei Orlov
- Department of Biomedical Engineering, Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
| | - Amit Gefen
- Department of Biomedical Engineering, Faculty of EngineeringTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
45
|
Nilforoushzadeh MA, Khodaverdi Darian E, Afzali H, Amirkhani MA, Razzaghi M, Naser R, Amiri AB, Alimohammadi A, Nikkhah N, Zare S. Role of Cultured Skin Fibroblasts in Regenerative Dermatology. Aesthetic Plast Surg 2022; 46:1463-1471. [PMID: 35676559 DOI: 10.1007/s00266-022-02940-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 05/04/2022] [Indexed: 11/26/2022]
Abstract
The skin, as the largest organ, covers the entire outer part of the body, and since this organ is directly exposed to microbial, thermal, mechanical and chemical damage, it may be destroyed by factors such as acute trauma, chronic wounds or even surgical interventions. Cell therapy is one of the most important procedures to treat skin lesions. Fibroblasts are cells that are responsible for the synthesis of collagen, elastin, and the organization of extracellular matrix (ECM) components and have many vital functions in wound healing processes. Today, cultured autologous fibroblasts are used to treat wrinkles, scars, wounds and subcutaneous atrophy. The results of many studies have shown that fibroblasts can be effective and beneficial in the treatment of skin lesions. On the other hand, skin substitutes are used as a regenerative model to improve and regenerate the skin. The use of these alternatives, restorative medicine and therapeutic cells such as fibroblasts has tremendous potential in the treatment of skin diseases and can be a new window for the treatment of diseases with no definitive treatment. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description ofthese Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Mohammad Ali Nilforoushzadeh
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Jordan Dermatology and Hair Transplantation Center, Tehran, Iran
| | - Ebrahim Khodaverdi Darian
- Department of Medical Biotechnology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Biotechnology Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamideh Afzali
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammadreza Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Naser
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Behtash Amiri
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Alimohammad Alimohammadi
- Forensic Medicine Specialist, Research Center of Legal Medicine Organization of Iran, Tehran, Iran
| | - Nahid Nikkhah
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sona Zare
- Skin and Stem Cell Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Mu R, Campos de Souza S, Liao Z, Dong L, Wang C. Reprograming the immune niche for skin tissue regeneration - From cellular mechanisms to biomaterials applications. Adv Drug Deliv Rev 2022; 185:114298. [PMID: 35439569 DOI: 10.1016/j.addr.2022.114298] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023]
Abstract
Despite the rapid development of therapeutic approaches for skin repair, chronic wounds such as diabetic foot ulcers remain an unaddressed problem that affects millions of people worldwide. Increasing evidence has revealed the crucial and diverse roles of the immune cells in the development and repair of the skin tissue, prompting new research to focus on further understanding and modulating the local immune niche for comprehensive, 'perfect' regeneration. In this review, we first introduce how different immunocytes and certain stromal cells involved in innate and adaptive immunity coordinate to maintain the immune niche and tissue homeostasis, with emphasis on their specific roles in normal and pathological wound healing. We then discuss novel engineering approaches - particularly biomaterials systems and cellular therapies - to target different players of the immune niche, with three major aims to i) overcome 'under-healing', ii) avoid 'over-healing', and iii) promote functional restoration, including appendage development. Finally, we highlight how these strategies strive to manage chronic wounds and achieve full structural and functional skin recovery by creating desirable 'soil' through modulating the immune microenvironment.
Collapse
|
47
|
Tran NT, Park IS, Truong MD, Park DY, Park SH, Min BH. Conditioned media derived from human fetal progenitor cells improves skin regeneration in burn wound healing. Cell Tissue Res 2022; 389:289-308. [PMID: 35624315 DOI: 10.1007/s00441-022-03638-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/11/2022] [Indexed: 01/06/2023]
Abstract
Stem cells are known to have excellent regenerative ability, which is primarily facilitated by indirect paracrine factors, rather than via direct cell replacement. The regenerative process is mediated by the release of extracellular matrix molecules, cytokines, and growth factors, which are also present in the media during cultivation. Herein, we aimed to demonstrate the functionality of key factors and mechanisms in skin regeneration through the analysis of conditioned media derived from fetal stem cells. A series of processes, including 3D pellet cultures, filtration and lyophilization is developed to fabricate human fetal cartilage-derived progenitor cells-conditioned media (hFCPCs-CM) and its useful properties are compared with those of human bone marrow-derived MSCs-conditioned media (hBMSCs-CM) in terms of biochemical characterization, and in vitro studies of fibroblast behavior, macrophage polarization, and burn wound healing. The hFCPCs-CM show to be devoid of cellular components but to contain large amounts of total protein, collagen, glycosaminoglycans, and growth factors, including IGFBP-2, IGFBP-6, HGF, VEGF, TGF β3, and M-CSF, and contain a specific protein, collagen alpha-1(XIV) compare with hBMSCs-CM. The therapeutic potential of hFCPCs-CM observes to be better than that of hBMSCs-CM in the viability, proliferation, and migration of fibroblasts, and M2 macrophage polarization in vitro, and efficient acceleration of wound healing and minimization of scar formation in third-degree burn wounds in a rat model. The current study shows the potential therapeutic effect of hFCPCs and provides a rationale for using the secretome released from fetal progenitor cells to promote the regeneration of skin tissues, both quantitatively and qualitatively. The ready-to-use product of human fetal cartilage-derived progenitor cells-conditioned media (hFCPCs-CM) are fabricated via a series of techniques, including a 3D culture of hFCPCs, filtration using a 3.5 kDa cutoff dialysis membrane, and lyophilization of the CM. hFCPCs-CM contains many ECM molecules and biomolecules that improves wound healing through efficient acceleration of M2 macrophage polarization and reduction of scar formation.
Collapse
Affiliation(s)
- Ngoc-Trinh Tran
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Korea
| | - In-Su Park
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Korea
| | | | - Do-Young Park
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Korea
| | - Sang-Hyug Park
- Advanced Translational Engineering and Medical Science, Seoul, Korea.
- Department of Biomecial Engineering, Pukyong National University, Busan, 48513, Korea.
| | - Byoung-Hyun Min
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea.
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Korea.
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, Korea.
- Advanced Translational Engineering and Medical Science, Seoul, Korea.
| |
Collapse
|
48
|
Eremenko E, Ding J, Kwan P, Tredget EE. The Biology of Extracellular Matrix Proteins in Hypertrophic Scarring. Adv Wound Care (New Rochelle) 2022; 11:234-254. [PMID: 33913776 DOI: 10.1089/wound.2020.1257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Significance: Hypertrophic scars (HTS) are a fibroproliferative disorder that occur following deep dermal injury and affect up to 72% of burn patients. These scars result in discomfort, impaired mobility, disruption of normal function and cosmesis, and significant psychological distress. Currently, there are no satisfactory methods to treat or prevent HTS, as the cellular and molecular mechanisms are complex and incompletely understood. This review summarizes the biology of proteins in the dermal extracellular matrix (ECM), which are involved in wound healing and hypertrophic scarring. Recent Advances: New basic research continues toward understanding the diversity of cellular and molecular mechanisms of normal wound healing and hypertrophic scarring. Broadening the understanding of these mechanisms creates insight into novel methods for preventing and treating HTS. Critical Issues: Although there is an abundance of research conducted on collagen in the ECM and its relationship to HTS, there is a significant gap in understanding the role of proteoglycans and their specific isoforms in dermal fibrosis. Future Directions: Exploring the biological roles of ECM proteins and their unique isoforms in HTS, mature scars, and normal skin will further the understanding of abnormal wound healing and create a more robust understanding of what constitutes dermal fibrosis. Research into the biological roles of ECM protein isoforms and their regulation during wound healing warrants a more extensive investigation to identify their distinct biological functions in cellular processes and outcomes.
Collapse
Affiliation(s)
- Elizabeth Eremenko
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Jie Ding
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
| | - Peter Kwan
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Edward E. Tredget
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Canada
- Division of Plastic Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| |
Collapse
|
49
|
Decorin Inhibits Dermal Mesenchymal Cell Migration and Induces Scar Formation. Plast Reconstr Surg Glob Open 2022; 10:e4245. [PMID: 35425688 PMCID: PMC9000046 DOI: 10.1097/gox.0000000000004245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/07/2022] [Indexed: 11/27/2022]
Abstract
Background Variations in skin healing capacities are observed during different murine embryonic developmental stages. Through embryonic day 16 (E16), embryos are able to regenerate dermal architecture following flank skin wounding; however, after E17, wounds heal incompletely, inducing scar formation. The regenerative ability of the E16 fetal dermis depends on the migration of dermal mesenchymal cells. Decorin is a small molecule known to affect tissue tensile strength, cell phenotype, and tissue repair, including skin wound healing. In the current study, we evaluated the expression and roles of decorin in wound healing. Methods Surgical injury was induced at E16 and E17 in ICR mouse embryos. Decorin expression was evaluated in tissue samples from these embryos using immunohistochemistry and reverse transcription quantitative polymerase chain reaction. Cell migration assays were used to evaluate wound healing capability of separated dermal and fascial tissues. Results Our results showed that decorin exhibited distinct expression patterns during wound healing at E16 versus E17. Additionally, decorin expression altered cell migration in vitro. Dermal and fascial mesenchymal cells were found to exhibit distinct migration patterns concomitant with altered decorin expression. Specifically, decorin inhibited migration and favored scar formation. Conclusion Decorin expression may contribute to scar formation in the late stage of mouse embryos by inhibiting the migration of dermal mesenchymal cells.
Collapse
|
50
|
Capolupo L, Khven I, Lederer AR, Mazzeo L, Glousker G, Ho S, Russo F, Montoya JP, Bhandari DR, Bowman AP, Ellis SR, Guiet R, Burri O, Detzner J, Muthing J, Homicsko K, Kuonen F, Gilliet M, Spengler B, Heeren RMA, Dotto GP, La Manno G, D'Angelo G. Sphingolipids control dermal fibroblast heterogeneity. Science 2022; 376:eabh1623. [PMID: 35420948 DOI: 10.1126/science.abh1623] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human cells produce thousands of lipids that change during cell differentiation and can vary across individual cells of the same type. However, we are only starting to characterize the function of these cell-to-cell differences in lipid composition. Here, we measured the lipidomes and transcriptomes of individual human dermal fibroblasts by coupling high-resolution mass spectrometry imaging with single-cell transcriptomics. We found that the cell-to-cell variations of specific lipid metabolic pathways contribute to the establishment of cell states involved in the organization of skin architecture. Sphingolipid composition is shown to define fibroblast subpopulations, with sphingolipid metabolic rewiring driving cell-state transitions. Therefore, cell-to-cell lipid heterogeneity affects the determination of cell states, adding a new regulatory component to the self-organization of multicellular systems.
Collapse
Affiliation(s)
- Laura Capolupo
- Interfaculty Institute of Bioengineering and Global Health Institute, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Irina Khven
- Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Alex R Lederer
- Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Luigi Mazzeo
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Galina Glousker
- School of Life Sciences, Swiss Institute for Experimental Cancer Research, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Sylvia Ho
- Interfaculty Institute of Bioengineering and Global Health Institute, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Francesco Russo
- Institute of Biochemistry and Cellular Biology, National Research Council of Italy, 80131 Napoli, Italy
| | - Jonathan Paz Montoya
- Interfaculty Institute of Bioengineering and Global Health Institute, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Dhaka R Bhandari
- Institute for Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Andrew P Bowman
- Maastricht MultiModal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, 6629 ER Maastricht, Netherlands
| | - Shane R Ellis
- Maastricht MultiModal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, 6629 ER Maastricht, Netherlands.,Molecular Horizons and School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, New South Wales 2522, Australia
| | - Romain Guiet
- Faculté des Sciences de la Vie, Bioimaging and Optics Platform, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015 Vaud, Switzerland
| | - Olivier Burri
- Faculté des Sciences de la Vie, Bioimaging and Optics Platform, École Polytechnique Fédérale de Lausanne, Lausanne, CH-1015 Vaud, Switzerland
| | - Johanna Detzner
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany
| | - Johannes Muthing
- Institute of Hygiene, University of Münster, D-48149 Münster, Germany
| | - Krisztian Homicsko
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland.,Swiss Cancer Center Leman, CH-1015 Lausanne, Switzerland.,The Ludwig Institute for Cancer Research, Lausanne Branch, CH-1066 Epalinges, Switzerland
| | - François Kuonen
- Département de Dermatologie et Vénéréologie, Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland
| | - Michel Gilliet
- Département de Dermatologie et Vénéréologie, Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland
| | - Bernhard Spengler
- Institute for Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Ron M A Heeren
- Maastricht MultiModal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, 6629 ER Maastricht, Netherlands
| | - G Paolo Dotto
- Personalized Cancer Prevention Research Unit, Head and Neck Surgery Division, Centre Hospitalier Universitaire Vaudois, CH-1011 Lausanne, Switzerland.,Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland.,Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Gioele La Manno
- Brain Mind Institute, Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Giovanni D'Angelo
- Interfaculty Institute of Bioengineering and Global Health Institute, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland.,Institute of Biochemistry and Cellular Biology, National Research Council of Italy, 80131 Napoli, Italy
| |
Collapse
|