1
|
Li W, Zhang H, Chen L, Huang C, Jiang Z, Zhou H, Zhu X, Liu X, Zheng Z, Yu Q, He Y, Gao Y, Ma J, Yang L. Cell membrane-derived nanovesicles as extracellular vesicle-mimetics in wound healing. Mater Today Bio 2025; 31:101595. [PMID: 40104636 PMCID: PMC11914519 DOI: 10.1016/j.mtbio.2025.101595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Cell membrane-derived nanovesicles (NVs) have emerged as promising alternatives to extracellular vesicles (EVs) for wound healing applications, addressing the limitations of traditional EVs, which include insufficient targeting capability, low production yield, and limited drug-loading capacity. Through mechanical cell extrusion methods, NVs exhibit superior characteristics, demonstrating enhanced yield, stability, and purity compared to natural EVs. These NVs can be derived from various membrane sources, including single cell types (stem cells, blood cells, immune cells, and bacterial membranes), hybrid cell membranes and cell membranes mixed with liposomes, with each offering unique therapeutic properties. The integration of genetic engineering and surface modifications has further enhanced NV functionality, enabling precise targeting and improved drug delivery capabilities. Recent advances in NV-based therapies have demonstrated their potential across multiple biomedical applications. Although challenges persist in terms of standardization, storage stability, and clinical translation, the combination of natural cell-derived functions with artificial modification potential positions NVs as a promising platform for next-generation therapeutic delivery systems, thereby offering new possibilities in wound healing applications. Finally, we explore the challenges and future prospects of translating NV-based therapeutics into clinical practice, providing insights into the future development of this innovative approach in wound healing and tissue repair.
Collapse
Affiliation(s)
- Wenwen Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huihui Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoyang Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hai Zhou
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinxi Zhu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoyang Liu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zesen Zheng
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyi Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yufang He
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
2
|
Dartora VFC, Carney R, Wang A, Qiu P, Panitch A. Extracellular matrix ligands modulate the endothelial progenitor cell secretome for enhanced angiogenesis. Acta Biomater 2025; 195:240-255. [PMID: 39954753 DOI: 10.1016/j.actbio.2025.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Wound healing is a complex physiological process fundamentally dependent on angiogenesis for effective tissue repair. Endothelial progenitor cells (EPCs) have shown significant potential in promoting angiogenesis, primarily through their secretome, rich in proteins and extracellular vesicles (EVs) essential for cell signaling and tissue regeneration. This study investigates the effect of a collagen-bound proteoglycan mimetic (LXW7-DS-SILY or LDS), that binds to the αvβ3 integrin receptor, on the EPC secretome, with a dual focus on the proteomic content and the functional properties of EVs. Utilizing high-resolution two-dimensional liquid chromatography-tandem mass spectrometry (LC-MS/MS) alongside bioinformatic analysis, we identified significant alterations in protein expression profiles, particularly in angiogenesis and wound healing pathways. The functional impact of these changes was validated through biological assays, where the whole secretome and the EV fraction from EPCs seeded on collagen-bound LDS markedly enhanced vascular network formation, driven by the increase of growth factors and angiogenic regulators such as FGFR1, NRP1, and ANGPT2 within the EV fraction. Gene Ontology (GO) enrichment analysis further highlighted the enrichment of proteins within the EVs driving biological processes, including 'response to wounding' and 'positive regulation of cell motility'. These results underscore that LDS modulates the EPC secretome and significantly enhances its angiogenic potential, offering a promising therapeutic strategy for non-healing and ischemic wounds and suggesting that biomaterials can be modified to control the EV secretome to enhance tissue repair. Functional assays validating the omics data highlight the robustness of LDS as a targeted therapeutic for enhancing angiogenesis and tissue repair in clinical settings. Moreover, the pivotal role of EVs in mediating pro-angiogenic effects offers insights into developing biomaterial therapies that exploit key regulators within the EPC secretome for wound healing. STATEMENT OF SIGNIFICANCE: This manuscript explores how a proteoglycan mimetic that binds to both collagen and the αvβ3 integrin receptor affects the proteome component of the secretome from endothelial progenitor cells (EPCs). It presents functional biological data, analytical data, and proteomic analysis of the soluble and extracellular vesical (EV) components of the secratome. The proteomic data maps to the observed enhanced angiogenic potential of the EVs. These results suggest that by controlling the cellular environment and judicially engineering how cells interact with a biomaterial can influence the proteomic composition of EVs to enhance tissue regeneration. This is the foundation of future work aimed at engineering biomaterial cell systems to influence the proteomic component of EVs for therapeutic use.
Collapse
Affiliation(s)
- Vanessa F C Dartora
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Randy Carney
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA
| | - Aijun Wang
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Peng Qiu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA
| | - Alyssa Panitch
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA, USA; Department of Surgery, University of California Davis, Sacramento, CA 95817, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, USA.
| |
Collapse
|
3
|
Aliniay-Sharafshadehi S, Yousefi MH, Ghodratie M, Kashfi M, Afkhami H, Ghoreyshiamiri SM. Exploring the therapeutic potential of different sources of mesenchymal stem cells: a novel approach to combat burn wound infections. Front Microbiol 2024; 15:1495011. [PMID: 39678916 PMCID: PMC11638218 DOI: 10.3389/fmicb.2024.1495011] [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: 09/11/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
The most prevalent and harmful injuries are burns, which are still a major global health problem. Burn injuries can cause issues because they boost the inflammatory and metabolic response, which can cause organ malfunction and systemic failure. On the other hand, a burn wound infection creates an environment that is conducive to the growth of bacteria and might put the patient at risk for sepsis. In addition, scarring is unavoidable, and this results in patients having functional and cosmetic issues. Wound healing is an amazing phenomenon with a complex mechanism that deals with different types of cells and biomolecules. Cell therapy using stem cells is one of the most challenging treatment methods that accelerates the healing of burn wounds. Since 2000, the use of mesenchymal stem cells (MSCs) in regenerative medicine and wound healing has increased. They can be extracted from various tissues, such as bone marrow, fat, the umbilical cord, and the amniotic membrane. According to studies, stem cell therapy for burn wounds increases angiogenesis, has anti-inflammatory properties, slows the progression of fibrosis, and has an excellent ability to differentiate and regenerate damaged tissue. Figuring out the main preclinical and clinical problems that stop people from using MSCs and then suggesting the right ways to improve therapy could help show the benefits of MSCs and move stem cell-based therapy forward. This review's objective was to assess mesenchymal stem cell therapy's contribution to the promotion of burn wound healing.
Collapse
Affiliation(s)
- Shahrzad Aliniay-Sharafshadehi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Ghodratie
- Department of Medical Microbiology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mojtaba Kashfi
- Fellowship in Clinical Laboratory Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | | |
Collapse
|
4
|
Zhang S, Jiang T, Li M, Sun H, Wu H, Wu W, Li Y, Jiang H. Graphene-Based Wound Dressings for Wound Healing: Mechanism, Technical Analysis, and Application Status. ACS Biomater Sci Eng 2024; 10:6790-6813. [PMID: 39467733 DOI: 10.1021/acsbiomaterials.4c01142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The development of novel wound dressings is critical in medical care. Graphene and its derivatives possess excellent biomedical properties, making them highly suitable for various applications in medical dressings. This review provides a comprehensive technical analysis and the current application status of graphene-based medical dressings. Initially, we discuss the chemical structure and the fabrication method of graphene and its derivatives. We then provide a detailed summary of the mechanisms by which graphene materials promote wound repair across the four stages of wound healing. Subsequently, we categorize the types of graphene-based wound dressings and analyze corresponding characteristics. Finally, we analyze the challenges encountered at present and propose solutions regarding future development trends. This paper aims to serve as a reference for further research in skin tissue engineering and the development of innovative graphene-based medical dressings.
Collapse
Affiliation(s)
- Shanguo Zhang
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Tianyi Jiang
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Ming Li
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Haoxiu Sun
- School of Life Sciences, Harbin Institute of Technology, No. 2 Yikuang Street, Harbin 150001, People's Republic of China
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, No. 157 Health Road, Harbin 150001, People's Republic of China
| | - Hao Wu
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Wenlong Wu
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| | - Yu Li
- School of Life Sciences, Harbin Institute of Technology, No. 2 Yikuang Street, Harbin 150001, People's Republic of China
| | - Hongyuan Jiang
- School of Mechatronics Engineering, Harbin Institute of Technology, No. 92 West Da-zhi Street, Harbin 150001, People's Republic of China
| |
Collapse
|
5
|
Wu X, Li W, Herlah L, Koch M, Wang H, Schirhagl R, Włodarczyk-Biegun MK. Melt electrowritten poly-lactic acid /nanodiamond scaffolds towards wound-healing patches. Mater Today Bio 2024; 26:101112. [PMID: 38873104 PMCID: PMC11170272 DOI: 10.1016/j.mtbio.2024.101112] [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: 01/24/2024] [Revised: 05/18/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
Multifunctional wound dressings, enriched with biologically active agents for preventing or treating infections and promoting wound healing, along with cell delivery capability, are highly needed. To address this issue, composite scaffolds with potential in wound dressing applications were fabricated in this study. The poly-lactic acid/nanodiamonds (PLA/ND) scaffolds were first printed using melt electrowriting (MEW) and then coated with quaternized β-chitin (QβC). The NDs were well-dispersed in the printed filaments and worked as fillers and bioactive additions to PLA material. Additionally, they improved coating effectiveness due to the interaction between their negative charges (from NDs) and positive charges (from QβC). NDs not only increased the thermal stability of PLA but also benefitted cellular behavior and inhibited the growth of bacteria. Scaffolds coated with QβC increased the effect of bacteria growth inhibition and facilitated the proliferation of human dermal fibroblasts. Additionally, we have observed rapid extracellular matrix (ECM) remodeling on QβC-coated PLA/NDs scaffolds. The scaffolds provided support for cell adhesion and could serve as a valuable tool for delivering cells to chronic wound sites. The proposed PLA/ND scaffold coated with QβC holds great potential for achieving fast healing in various types of wounds.
Collapse
Affiliation(s)
- Xixi Wu
- Department of Biomedical Engineering, University Medical Centre, Ant. Deusinglaan 1, 9713, AW, Groningen, the Netherlands
- Polymer Science, Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
| | - Wenjian Li
- Advanced Production Engineering, Engineering and Technology Institute of Groningen, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
| | - Lara Herlah
- Department of Biomedical Engineering, University Medical Centre, Ant. Deusinglaan 1, 9713, AW, Groningen, the Netherlands
| | - Marcus Koch
- INM – Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Hui Wang
- Nanostructured Materials and Interfaces, Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
| | - Romana Schirhagl
- Department of Biomedical Engineering, University Medical Centre, Ant. Deusinglaan 1, 9713, AW, Groningen, the Netherlands
| | - Małgorzata K. Włodarczyk-Biegun
- Polymer Science, Zernike Institute for Advanced Materials, Faculty of Science and Engineering, University of Groningen, Nijenborgh 4, 9747, AG, the Netherlands
- Biotechnology Centre, The Silesian University of Technology, Krzywoustego 8, 44-100, Gliwice, Poland
| |
Collapse
|
6
|
Shahin H, Belcastro L, Das J, Perdiki Grigoriadi M, Saager RB, Steinvall I, Sjöberg F, Olofsson P, Elmasry M, El-Serafi AT. MicroRNA-155 mediates multiple gene regulations pertinent to the role of human adipose-derived mesenchymal stem cells in skin regeneration. Front Bioeng Biotechnol 2024; 12:1328504. [PMID: 38562669 PMCID: PMC10982420 DOI: 10.3389/fbioe.2024.1328504] [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: 10/26/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction: The role of Adipose-derived mesenchymal stem cells (AD-MSCs) in skin wound healing remains to be fully characterized. This study aims to evaluate the regenerative potential of autologous AD-MSCs in a non-healing porcine wound model, in addition to elucidate key miRNA-mediated epigenetic regulations that underlie the regenerative potential of AD-MSCs in wounds. Methods: The regenerative potential of autologous AD-MSCs was evaluated in porcine model using histopathology and spatial frequency domain imaging. Then, the correlations between miRNAs and proteins of AD-MSCs were evaluated using an integration analysis in primary human AD-MSCs in comparison to primary human keratinocytes. Transfection study of AD-MSCs was conducted to validate the bioinformatics data. Results: Autologous porcine AD-MSCs improved wound epithelialization and skin properties in comparison to control wounds. We identified 26 proteins upregulated in human AD-MSCs, including growth and angiogenic factors, chemokines and inflammatory cytokines. Pathway enrichment analysis highlighted cell signalling-associated pathways and immunomodulatory pathways. miRNA-target modelling revealed regulations related to genes encoding for 16 upregulated proteins. miR-155-5p was predicted to regulate Fibroblast growth factor 2 and 7, C-C motif chemokine ligand 2 and Vascular cell adhesion molecule 1. Transfecting human AD-MSCs cell line with anti-miR-155 showed transient gene silencing of the four proteins at 24 h post-transfection. Discussion: This study proposes a positive miR-155-mediated gene regulation of key factors involved in wound healing. The study represents a promising approach for miRNA-based and cell-free regenerative treatment for difficult-to-heal wounds. The therapeutic potential of miR-155 and its identified targets should be further explored in-vivo.
Collapse
Affiliation(s)
- Hady Shahin
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
- Faculty of Biotechnology, Modern Sciences and Arts University, October City, Cairo, Egypt
| | - Luigi Belcastro
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Jyotirmoy Das
- Bioinformatics Unit, Core Facility (KEF), Faculty of Medicine and Health Sciences (BKV), Linköping University, Linköping, Sweden
- Clinical Genomics Linköping, SciLife Laboratory, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Rolf B. Saager
- Department of Biomedical Engineering, Linkoping University, Linköping, Sweden
| | - Ingrid Steinvall
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Folke Sjöberg
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| | - Pia Olofsson
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Moustafa Elmasry
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
| | - Ahmed T. El-Serafi
- Department of Hand Surgery, Plastic Surgery, and Burns, Linkoping University Hospital, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linkoping University, Linköping, Sweden
| |
Collapse
|
7
|
Panda D, Nayak S. Stem Cell-Based Tissue Engineering Approaches for Diabetic Foot Ulcer: a Review from Mechanism to Clinical Trial. Stem Cell Rev Rep 2024; 20:88-123. [PMID: 37867186 DOI: 10.1007/s12015-023-10640-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2023] [Indexed: 10/24/2023]
Abstract
Diabetic foot ulcer (DFU) is a complication from incomplete or prolonged wound healing, at times requires amputation, putting substantial health and socioeconomic burden. Wound healing is a dynamic overlapping process that can be regulated by arrays of molecular factors showing redundancy in function. However, dysregulation in the mechanism of angiogenesis, extra cellular matrix (ECM) formation and immune modulation are the major causes for impair wound healing in hyperglycaemic patients. Despite development of wound care research, there is a lack of well-accepted targeted therapy with multidisciplinary approach for DFU treatment. Stem cell therapy holds a promising outcome both in preclinical and clinical trials because of its ability to promote healing via regeneration and specialized tissue differentiation. Among different types of stem cells, regenerative potential of mesenchymal stem cell (MSC) is well demonstrated in both experimental and clinical trial. Still there is a huge knowledge gap among medical practitioners for deciding the best stem cell source, administration route, and safety. This review strengthens the fact that why stem cell therapy is a promising candidate to treat DFU and cited multiple tissue engineering and biomaterial-based approaches for delivering stem cells and their aftermath paracrine events. Based on the pre-clinical and clinical studies, the review tried to come up with optimum stem cell source and delivery route for the treatment of DFU. At last, the review glances on possible direction to enhance therapeutics strategy for the same, including different approaches like: phytocompounds, exosomes, scaffold geometry, cell preconditioning and licensing etc.
Collapse
Affiliation(s)
- Debarchan Panda
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Sunita Nayak
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
8
|
Mills SJ, Kirby GT, Hofma BR, Smith LE, Statham P, Vaes B, Ting AE, Short R, Cowin AJ. Delivery of multipotent adult progenitor cells via a functionalized plasma polymerized surface accelerates healing of murine diabetic wounds. Front Bioeng Biotechnol 2023; 11:1213021. [PMID: 37675407 PMCID: PMC10477914 DOI: 10.3389/fbioe.2023.1213021] [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: 04/27/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023] Open
Abstract
Introduction: Stem cell therapies have been investigated as potential treatment modalities for chronic wounds however there has been limited success to date. Multipotent Adult Progenitor Cells (MAPCs©) have been identified as having potential as an allogenic stem cell product due to their high population doubling number and their characteristic dampening of T-cell proliferation. This helps to prevent autoimmunity and graft/cell rejection. Methods: We have developed a dressing, consisting of medical grade silicone coated with a heptylamine plasma polymer, which supports the growth and transfer of MAPCs to skin. To determine if the dressing can deliver functional stem cells into diabetic wounds, they were loaded with MAPCs and then placed over excisional wounds in both normal and diabetic mice. Results and discussion: Accelerated healing was observed in both the normal and diabetic wounds with wound gape being significantly smaller at day 3 when compared to controls. Wound analysis showed that treatment with the MAPC dressings dampened the inflammatory response with reduced numbers of neutrophils and macrophages observed. Additionally, an increase in pro-angiogenic VEGF and CD31 positive endothelial cells was observed indicating improved new blood vessel formation. The MAPC dressings had no effect on fibrosis with collagen I and III being equally affected in both control and treated wounds. Overall, the functionalized MAPC dressings improve healing responses particularly in diabetic mice with impaired healing responses and therefore, show potential for development as an advanced therapeutic approach for the treatment of chronic diabetic wounds.
Collapse
Affiliation(s)
- S. J. Mills
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - G. T. Kirby
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - B. R. Hofma
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - L. E. Smith
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - P. Statham
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| | - B. Vaes
- ReGenesys BV, Bio-Incubator Leuven, Leuven, Belgium
| | - A. E. Ting
- Athersys Inc., Cleveland, OH, United States
| | - R. Short
- Material Science Institute, Lancaster University, Lancaster, United Kingdom
| | - A. J. Cowin
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, SA, Australia
| |
Collapse
|
9
|
Ding JY, Chen MJ, Wu LF, Shu GF, Fang SJ, Li ZY, Chu XR, Li XK, Wang ZG, Ji JS. Mesenchymal stem cell-derived extracellular vesicles in skin wound healing: roles, opportunities and challenges. Mil Med Res 2023; 10:36. [PMID: 37587531 PMCID: PMC10433599 DOI: 10.1186/s40779-023-00472-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/24/2023] [Indexed: 08/18/2023] Open
Abstract
Skin wounds are characterized by injury to the skin due to trauma, tearing, cuts, or contusions. As such injuries are common to all human groups, they may at times represent a serious socioeconomic burden. Currently, increasing numbers of studies have focused on the role of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) in skin wound repair. As a cell-free therapy, MSC-derived EVs have shown significant application potential in the field of wound repair as a more stable and safer option than conventional cell therapy. Treatment based on MSC-derived EVs can significantly promote the repair of damaged substructures, including the regeneration of vessels, nerves, and hair follicles. In addition, MSC-derived EVs can inhibit scar formation by affecting angiogenesis-related and antifibrotic pathways in promoting macrophage polarization, wound angiogenesis, cell proliferation, and cell migration, and by inhibiting excessive extracellular matrix production. Additionally, these structures can serve as a scaffold for components used in wound repair, and they can be developed into bioengineered EVs to support trauma repair. Through the formulation of standardized culture, isolation, purification, and drug delivery strategies, exploration of the detailed mechanism of EVs will allow them to be used as clinical treatments for wound repair. In conclusion, MSC-derived EVs-based therapies have important application prospects in wound repair. Here we provide a comprehensive overview of their current status, application potential, and associated drawbacks.
Collapse
Affiliation(s)
- Jia-Yi Ding
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, 323000, Lishui, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Min-Jiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, 323000, Lishui, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ling-Feng Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Clinical College of the Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, Zhejiang, China
| | - Gao-Feng Shu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, 323000, Lishui, China
- Clinical College of the Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, Zhejiang, China
| | - Shi-Ji Fang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, 323000, Lishui, China
- Clinical College of the Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, Zhejiang, China
| | - Zhao-Yu Li
- Department of Overseas Education College, Jimei University, Xiamen, 361021, Fujian, China
| | - Xu-Ran Chu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Medicine II, Internal Medicine, Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, 35392, Giessen, Germany
- Pulmonary and Critical Care, Department of Medicine V, Internal Medicine, Infectious Diseases and Infection Control, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig University Giessen, 35392, Giessen, Germany
| | - Xiao-Kun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Zhou-Guang Wang
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, 323000, Lishui, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Jian-Song Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Institute of Imaging Diagnosis and Minimally Invasive Intervention Research, the Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, 323000, Lishui, China.
- Clinical College of the Affiliated Central Hospital, School of Medicine, Lishui University, Lishui, 323000, Zhejiang, China.
| |
Collapse
|
10
|
Jakovija A, Chtanova T. Skin immunity in wound healing and cancer. Front Immunol 2023; 14:1060258. [PMID: 37398649 PMCID: PMC10312005 DOI: 10.3389/fimmu.2023.1060258] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/24/2023] [Indexed: 07/04/2023] Open
Abstract
The skin is the body's largest organ. It serves as a barrier to pathogen entry and the first site of immune defense. In the event of a skin injury, a cascade of events including inflammation, new tissue formation and tissue remodeling contributes to wound repair. Skin-resident and recruited immune cells work together with non-immune cells to clear invading pathogens and debris, and guide the regeneration of damaged host tissues. Disruption to the wound repair process can lead to chronic inflammation and non-healing wounds. This, in turn, can promote skin tumorigenesis. Tumors appropriate the wound healing response as a way of enhancing their survival and growth. Here we review the role of resident and skin-infiltrating immune cells in wound repair and discuss their functions in regulating both inflammation and development of skin cancers.
Collapse
Affiliation(s)
- Arnolda Jakovija
- Immunity Theme, Garvan Institute of Medical Research, Sydney, Australia
- St. Vincent’s School of Medicine, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Tatyana Chtanova
- Immunity Theme, Garvan Institute of Medical Research, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales, Sydney, Australia
| |
Collapse
|
11
|
Zhou Z, Xun J, Wu C, Ji C, Ji S, Shu F, Wang Y, Chen H, Zheng Y, Xiao S. Acceleration of burn wound healing by micronized amniotic membrane seeded with umbilical cord-derived mesenchymal stem cells. Mater Today Bio 2023; 20:100686. [PMID: 37334186 PMCID: PMC10276167 DOI: 10.1016/j.mtbio.2023.100686] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/11/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023] Open
Abstract
Umbilical cord-derived mesenchymal stem cells (UC-MSC) are promising candidates for wound healing. However, the low amplification efficiency of MSC in vitro and their low survival rates after transplantation have limited their medical application. In this study, we fabricated a micronized amniotic membrane (mAM) as a microcarrier to amplify MSC in vitro and used mAM and MSC (mAM-MSC) complexes to repair burn wounds. Results showed that MSC could live and proliferate on mAM in a 3D culture system, exhibiting higher cell activity than in 2D culture. Transcriptome sequencing of MSC showed that the expression of growth factor-related, angiogenesis-related, and wound healing-related genes was significantly upregulated in mAM-MSC compared to traditional 2D-cultured MSC, which was verified via RT-qPCR. Gene ontology (GO) analysis of differentially expressed genes (DEGs) showed significant enrichment of terms related to cell proliferation, angiogenesis, cytokine activity, and wound healing in mAM-MSC. In a burn wound model of C57BL/6J mice, topical application of mAM-MSC significantly accelerated wound healing compared to MSC injection alone and was accompanied by longer survival of MSC and greater neovascularization in the wound.
Collapse
Affiliation(s)
- Zixuan Zhou
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jingnan Xun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Chenghao Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Chao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Futing Shu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Yuxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Hao Chen
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| |
Collapse
|
12
|
Lintel H, Abbas DB, Mackay DJ, Griffin M, Lavin CV, Berry CE, Guardino NJ, Guo JL, Momeni A, Mackay DR, Longaker MT, Wan DC. Topical vanadate improves tensile strength and alters collagen organisation of excisional wounds in a mouse model. Wound Repair Regen 2023; 31:77-86. [PMID: 36484112 PMCID: PMC10513738 DOI: 10.1111/wrr.13062] [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: 06/25/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 12/13/2022]
Abstract
Wound dehiscence, oftentimes a result of the poor tensile strength of early healing wounds, is a significant threat to the post-operative patient, potentially causing life-threatening complications. Vanadate, a protein tyrosine phosphatase inhibitor, has been shown to alter the organisation of deposited collagen in healing wounds and significantly improve the tensile strength of incisional wounds in rats. In this study, we sought to explore the effects of locally administered vanadate on tensile strength and collagen organisation in both the early and remodelling phases of excisional wound healing in a murine model. Wild-type mice underwent stented excisional wounding on their dorsal skin and were divided equally into three treatment conditions: vanadate injection, saline injection control and an untreated control. Tensile strength testing, in vivo suction Cutometer analysis, gross wound measurements and histologic analysis were performed during healing, immediately upon wound closure, and after 4 weeks of remodelling. We found that vanadate treatment significantly increased the tensile strength of wounds and their stiffness relative to control wounds, both immediately upon healing and into the remodelling phase. Histologic analysis revealed that these biomechanical changes were likely the result of increased collagen deposition and an altered collagen organisation composed of thicker and distinctly organised collagen bundles. Given the risk that dehiscence poses to all operative patients, vanadate presents an interesting therapeutic avenue to improve the strength of post-operative wounds and unstable chronic wounds to reduce the risk of dehiscence.
Collapse
Affiliation(s)
- Hendrik Lintel
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Darren B. Abbas
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Duncan J. Mackay
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Christopher V. Lavin
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Charlotte E. Berry
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Nicholas J. Guardino
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Jason L. Guo
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Arash Momeni
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Donald R. Mackay
- Department of Plastic Surgery, Pennsylvania State University Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University Medical Center, Stanford, California, USA
| | - Derrick C. Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
13
|
Ma J, Yong L, Lei P, Li H, Fang Y, Wang L, Chen H, Zhou Q, Wu W, Jin L, Sun D, Zhang X. Advances in microRNA from adipose-derived mesenchymal stem cell-derived exosome: focusing on wound healing. J Mater Chem B 2022; 10:9565-9577. [PMID: 36398750 DOI: 10.1039/d2tb01987f] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Skin wounds are a common condition causing economic burden and they represent an urgent clinical need, especially chronic wounds. Numerous studies have been conducted on the applications of stem cell therapy in wound healing, with adipose-derived mesenchymal stem cells (ADMSCs) playing a major role since they can be isolated easily, yielding a high number of cells, the less invasive harvesting required, the longer life span and no ethical issues. However, the lack of standardized doses and protocols, the heterogeneity of clinical trials, as well as the incompatibility of the immune system limit its application. Recent studies have demonstrated that specific stem cell functions depend on paracrine factors, including extracellular vesicles, in which microRNAs in exosomes (Exo-miRNAs) are essential in controlling their functions. This paper describes the application and mechanism whereby ADMSC-Exo-miRNA regulates wound healing. ADMSC-Exo-miRNA is involved in various stages in wounds, including modulating the immune response and inflammation, accelerating skin cell proliferation and epithelialization, promoting vascular repair, and regulating collagen remodeling thereby reducing scar formation. In summary, this acellular therapy based on ADMSC-Exo-miRNA has considerable clinical potential, and provides reference values for developing new treatment strategies for wound healing.
Collapse
Affiliation(s)
- Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Ling Yong
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610000, China
| | - Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Hua Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Lei Wang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Haojie Chen
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Qi Zhou
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325000, China. .,Wenzhou City and Kunlong Technology Co., Ltd Joint Doctoral Innovation Station, Wenzhou Association for Science and Technology, Wenzhou 325000, China
| | - Xingxing Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou325000, China.
| |
Collapse
|
14
|
Hodge JG, Zamierowski DS, Robinson JL, Mellott AJ. Evaluating polymeric biomaterials to improve next generation wound dressing design. Biomater Res 2022; 26:50. [PMID: 36183134 PMCID: PMC9526981 DOI: 10.1186/s40824-022-00291-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 08/28/2022] [Indexed: 11/24/2022] Open
Abstract
Wound healing is a dynamic series of interconnected events with the ultimate goal of promoting neotissue formation and restoration of anatomical function. Yet, the complexity of wound healing can often result in development of complex, chronic wounds, which currently results in a significant strain and burden to our healthcare system. The advancement of new and effective wound care therapies remains a critical issue, with the current therapeutic modalities often remaining inadequate. Notably, the field of tissue engineering has grown significantly in the last several years, in part, due to the diverse properties and applications of polymeric biomaterials. The interdisciplinary cohesion of the chemical, biological, physical, and material sciences is pertinent to advancing our current understanding of biomaterials and generating new wound care modalities. However, there is still room for closing the gap between the clinical and material science realms in order to more effectively develop novel wound care therapies that aid in the treatment of complex wounds. Thus, in this review, we discuss key material science principles in the context of polymeric biomaterials, provide a clinical breadth to discuss how these properties affect wound dressing design, and the role of polymeric biomaterials in the innovation and design of the next generation of wound dressings.
Collapse
Affiliation(s)
- Jacob G Hodge
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA.,Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - David S Zamierowski
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jennifer L Robinson
- Department of Chemical and Petroleum Engineering, University of Kansas, Mail Stop: 3051, 3901 Rainbow Blvd, Lawrence, KS, 66160, USA
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
15
|
Gandhi RN, Makhecha MB. A Split Face Study to Compare the Efficacy of Platelet Rich Plasma Versus Normal Saline Injections in Acne Scars and to Assess the Utility of Ultrabiomicroscopic Sonography in Evaluation of Treatment Response. J Cutan Aesthet Surg 2022; 15:375-380. [PMID: 37035589 PMCID: PMC10081466 DOI: 10.4103/jcas.jcas_80_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
Background Multiple studies have proposed the effectiveness of Platelet rich plasma (PRP) in treatment of atrophic acne scars. But an unanswered question is - whether it is the presence of growth factors in PRP or the mechanical factors during the injection that are responsible for its effectiveness? Also, no study compares PRP as a monotherapy against inert solution like normal saline (NS), that too, in a split face pattern using a non-invasive objective evaluation method. Aims Primary - To compare the efficacy of intralesional PRP and NS injections, in the treatment of atrophic acne scars. Secondary - To assess the side effect profile of the PRP injections. Study Design Prospective Interventional Comparative Split Face study. Materials and Methods 30 out of 33 patients completed the study and 20 of them were subjected to Ultrabiomicroscopic (UBM) analysis. Each patient received intralesional injections of NS and PRP in a split face pattern at two weekly intervals for five sittings. Treatment response was assessed using Goodman And Baron qualitative grading system, photographic evaluation by two blinded dermatologist and UBM analysis (scar depth & length). Results All the three methods showed a statistically significant treatment response. However, there was no significant inter group difference and PRP and NS had similar efficacy in treating acne scars. Conclusion We conclude that subscision like mechanical effect of injecting solution is more important than the nature of the solution in treatment of atrophic acne scars and UBM sonography can be proposed as an objective assessment tool for such studies.
Collapse
Affiliation(s)
- Riddhi N Gandhi
- Department of Dermatology, HBTMC & Dr R N COOPER Hospital, Mumbai, India
| | | |
Collapse
|
16
|
Kandhwal M, Behl T, Singh S, Sharma N, Arora S, Bhatia S, Al-Harrasi A, Sachdeva M, Bungau S. Role of matrix metalloproteinase in wound healing. Am J Transl Res 2022; 14:4391-4405. [PMID: 35958464 PMCID: PMC9360851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Matrix metalloproteinases (MMPs) are a group of endopeptidases that play a vital role in the restoration of damaged skin. Through mediating various cellular events such as angiogenesis and vasodilation, MMPs are very crucial for the mechanism of wound healing. These enzymes are endopeptidases that are reliant on zinc which are concealed through the extracellular matrix (ECM). MMPs have different targets in different phases of wound healing through which they are capable of promoting timely healing in the body. This review discusses all the possible role of MMPs and their inhibitors that are involved during every step of the wound healing process. This review highlights the latest advances in the respective field about the regulation and mediation of MMPs in human skin and how these studies can be applied to other branches of medical sciences as well. Published papers were searched via MEDLINE, PubMed and MDPI from the available peer reviewed journals. Research done in the past suggests that active MMPs are involved in the healing progression of the wounds or they have a positive effect towards healing of wounds. Present studies in the relative field will further enhance the knowledge about enzymes working along with their inhibitors. These studies will help in a way to resolve some of the parameters that are necessary for modulating them either positively or negatively.
Collapse
Affiliation(s)
- Mimansa Kandhwal
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of NizwaNizwa 342001, Oman
- School of Health Science, University of Petroleum and Energy StudiesDehradun 248007, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of NizwaNizwa 342001, Oman
| | - Monika Sachdeva
- Fatima College of Health SciencesAl Ain 50, United Arab Emirates
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of OradeaOradea 410028, Romania
| |
Collapse
|
17
|
Wang D, Wen JY, Wu D, Ying ZY, Wen ZM, Peng HQ, Geng C, Feng YB, Sui ZG, Lv HY, Wu J, Xu B. LPS-pretreated MSC-conditioned medium optimized with 10-kDa filter attenuates the injury of H9c2 cardiomyocytes in a model of hypoxia/reoxygenation. Can J Physiol Pharmacol 2022; 100:651-664. [PMID: 35533248 DOI: 10.1139/cjpp-2021-0745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mesenchymal stem cell-derived conditioned medium (MSC-CM) improves cardiac function, which is partly attributed to released paracrine factors. Since such cardioprotection is moderate and transient, it's essential to optimize MSC-CM effective components to alleviate myocardial injury. To optimize MSC-CM, MSCs were treated with or without lipopolysaccharides (LPSs) for 48 h (serum-free), and the supernatant was collected. Then, LPS-CM (MSC stimulated by LPS) was further treated with LPS remover (LPS Re-CM) or was concentrated with a 10-kDa cutoff filter (10 kDa-CM). ELISA showed that all pretreatments increased levels of VEGF, HGF, and IGF except LPS remover; 10 kDa-CM was superior to other-CM. CCK-8 displayed that viability of injured H9c2 cells enhanced with the increase of MSC-CM concentration. We also found 10 kDa-CM significantly alleviated H9c2 hypoxia/reoxygenation (H/R) injury, as evidenced by increased Bcl-2/Bax ratio, decreased the levels of LDH and cTn. TEM, TUNEL, and H&E staining confirmed 10 kDa-CM inhibited H/R-induced H9c2 morphological changes. Proteomic analysis identified 41 differentially expressed proteins in 10 kDa-CM, among which anti-inflammation, pro-angiogenesis, and anti-apoptosis were related to cardiac protection. This study indicates that 10 kDa-CM protects H9c2 cardiomyocytes from H/R injury by preserving most of the protective factors, such as VEGF, HGF, and IGF, in MSC-CM.
Collapse
Affiliation(s)
- Dan Wang
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China.,Ordos Central Hospital, 586048, Department of Pharmacy, Ordos, Inner Mongolia, China;
| | - Jing-Yi Wen
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Di Wu
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Zi-Yue Ying
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Zhi-Min Wen
- The Second Affiliated Hospital of Dalian Medical University, Department of Clinical Laboratory, Dalian, Liaoning, China;
| | - Hui-Qian Peng
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Cong Geng
- The Second Affiliated Hospital of Dalian Medical University, Department of Clinical Laboratory, Dalian, Liaoning, China;
| | - Yuan-Bo Feng
- KU Leuven University Hospitals Leuven, 60182, Leuven, Flanders, Belgium;
| | - Zhi-Gang Sui
- Chinese Academy of Science, Dalian, Liaoning, China;
| | - Hui-Yi Lv
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China;
| | - Jun Wu
- The Second Affiliated Hospital of Dalian Medical University, Department of Echocardiography, Dalian, Liaoning, China;
| | - Bing Xu
- The Second Affiliated Hospital of Dalian Medical University, Department of Pharmacy, Dalian, Liaoning, China, 116023;
| |
Collapse
|
18
|
Constructing Tissue-Engineered Dressing Membranes with Adipose-Derived Stem Cells and Acellular Dermal Matrix for Diabetic Wound Healing: A Comparative Study of Hypoxia- or Normoxia-Culture Modes. Stem Cells Int 2022; 2022:2976185. [PMID: 35571531 PMCID: PMC9098365 DOI: 10.1155/2022/2976185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/22/2022] [Accepted: 03/16/2022] [Indexed: 12/11/2022] Open
Abstract
Diabetes foot ulcer (DFU) is a serious complication of diabetes, characterized by impaired vascular function, limited angiogenesis, and chronic inflammation. Direct stem cell injection on treating DFU is far from satisfactory in clinical practice, as this therapy neither protects nor localizes the injected cell suspension at the chronic ulcer site. Meanwhile, most of injected cells gradually perished within several days due to senescence or apoptosis. Acellular dermal matrix (ADM) has the potential to act as excellent cell delivery vehicles, considering it is highly biomimetic to native dermal tissue, has low immunogenicity, and suitable for stem cell attachment and proliferation. Hypoxia culture has significantly enhanced effects on the survival ability of in vitro cultured stem cells, indicating this culture mode is a suitable way for inhibiting the senescence or apoptosis of transplanted cells. In the current study, we, respectively, culture adipose-derived stem cells (ADSCs) on an ADM membrane under a hypoxia or normoxia condition to construct two kinds of tissue-engineered dressing membranes (H-ADSCs/ADM and N-ADSCs/ADM) and then comparatively evaluated their efficacy on DFU healing using a diabetic rat model. In vitro results showed that hypoxia precondition could stimulate the ADSCs secreting VEGF-A, and the culture medium from hypoxia-preconditioned ADSCs could enhance the proliferation, migration, and angiogenesis of HUVECs. In vivo results indicated that compared to the N-ADSCs/ADM membrane, the transplanted cells in the H-ADSCs/ADM membrane can survive longer at the chronic ulcer site, consequently improve angiogenesis, inhibit inflammation, and increase extracellular matrix remodeling, eventually accelerating DFU closure. This study provides an innovative covering graft for the treatment of DFU in the clinic.
Collapse
|
19
|
Luo H, Wang Y, Su Y, Liu D, Xiao H, Wu M, Zhao Y, Xue F. Paracrine effects of adipose-derived stem cells in cutaneous wound healing in streptozotocin-induced diabetic rats. J Wound Care 2022; 31:S29-S38. [PMID: 35199561 DOI: 10.12968/jowc.2022.31.sup3.s29] [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: 11/11/2022]
Abstract
OBJECTIVE The purpose of this study was to explore the paracrine effects of adipose-derived stem cells (ASCs) on cutaneous wound healing in diabetic rats. METHOD The ASCs were isolated and identified by immunofluorescent staining. The ASCs-conditioned medium (ASCs-CM) was harvested. Cell counting kit (CCK)-8 assay, scratch experiments, western blot and quantitative polymerase chain reaction (qPCR) were performed to observe the effects of ASCs-CM on fibroblasts. A full-thickness skin wound diabetic rat model was prepared, using 34 male, Sprague Dawley rats. ASCs-CM or negative-control medium (N-CM) was injected around the wound surface. The existing wound area was measured on days 4, 8, 12 and 16 after the postoperative day, and the wound tissues were collected for immunohistochemical staining and qPCR quantitative study. RESULTS In this experiment, the isolated cells were characterised as ASCs. The results of CCK-8 assay, cell scratch test, western blot and qPCR showed ASCs-CM could significantly promote the proliferation, migration and differentiation of fibroblasts. Simultaneously, the healing rate of full-thickness skin wounds in diabetic rats was significantly higher in the ASCs-CM group than the N-CM group on days 4, 8, 12 and 16. Immunohistochemical staining and qPCR results showed that the expression of vascular endothelial growth factor (VEGF, days 4 and 8), α-smooth muscle actin (SMA) (days 4 and 16), transforming growth factor (TGF)-β1 (days 4, 8 and 12) were higher in the ASCs-CM group than that of the N-CM group (p<0.05). CONCLUSION This experiment demonstrated that ASCs-CM may accelerate wound healing in diabetic rats by promoting the secretion of TGF-β1, VEGF and the proliferation, migration and differentiation of fibroblasts.
Collapse
Affiliation(s)
- Hua Luo
- Department of Orthopaedics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, Zhejiang, China.,Fengxian District Central Hospital Graduate Student Training Base, Jinzhou Medical University, Shanghai 201499, China.,Department of Laboratory Medicine & Central Laboratory, Fengxian District Central Hospital, Shanghai 201499, China
| | - Yongjian Wang
- Department of Orthopaedics, The First People's Hospital of Wenling, Taizhou, Zhejiang 317500, China
| | - Yongwei Su
- The First Affiliated Hospital of Jinzhou Medical University, Graduate Student Training Base, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Danping Liu
- The First Affiliated Hospital of Jinzhou Medical University, Graduate Student Training Base, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Haijun Xiao
- Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| | - Ming Wu
- Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| | - Yong Zhao
- Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| | - Feng Xue
- Fengxian District Central Hospital Graduate Student Training Base, Jinzhou Medical University, Shanghai 201499, China.,Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| |
Collapse
|
20
|
Three-dimensional scaffolds for tissue bioengineering cartilages. Biocybern Biomed Eng 2022. [DOI: 10.1016/j.bbe.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
21
|
Niculescu AG, Grumezescu AM. An Up-to-Date Review of Biomaterials Application in Wound Management. Polymers (Basel) 2022; 14:421. [PMID: 35160411 PMCID: PMC8839538 DOI: 10.3390/polym14030421] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/18/2022] Open
Abstract
Whether they are caused by trauma, illness, or surgery, wounds may occur throughout anyone's life. Some injuries' complexity and healing difficulty pose important challenges in the medical field, demanding novel approaches in wound management. A highly researched possibility is applying biomaterials in various forms, ranging from thin protective films, foams, and hydrogels to scaffolds and textiles enriched with drugs and nanoparticles. The synergy of biocompatibility and cell proliferative effects of these materials is reflected in a more rapid wound healing rate and improved structural and functional properties of the newly grown tissue. This paper aims to present the biomaterial dressings and scaffolds suitable for wound management application, reviewing the most recent studies in the field.
Collapse
Affiliation(s)
- Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, Politehnica University of Bucharest, 011061 Bucharest, Romania;
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Academy of Romanian Scientists, 3 Ilfov Street, 050044 Bucharest, Romania
| |
Collapse
|
22
|
Jiang W, Zhang J, Zhang X, Fan C, Huang J. VAP-PLGA microspheres (VAP-PLGA) promote adipose-derived stem cells (ADSCs)-induced wound healing in chronic skin ulcers in mice via PI3K/Akt/HIF-1α pathway. Bioengineered 2021; 12:10264-10284. [PMID: 34720043 PMCID: PMC8810082 DOI: 10.1080/21655979.2021.1990193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/23/2022] Open
Abstract
Chronic skin ulcers are a primary global health problem. Velvet antler polypeptide (VAP) regulates endothelial cell migration and angiogenic sprout. Adipose-derived stem cells (ADSCs) are reported to make pivotal impacts upon wound healing. This study aimed to explore the role of VAP combined with ADSCs in wound healing of chronic skin ulcers. The effect of VAP on phenotypes of ADSCs, and VAP (PLGA microspheres) combining with ADSCs on wound healing of chronic skin ulcers in vivo was evaluated. VAP generally promoted the proliferation, migration and invasion of ADSCs, and ADSC-induced angiogenesis in human umbilical vein endothelial cells (HUVECs) through PI3K/Akt/HIF-1α pathway. VAP-PLGA (PLGA microspheres) enhanced the promoting effect of ADSCs on wound healing, pathological changes, and angiogenesis in chronic skin ulcers in vivo. VAP-PLGA intensified the effect of ADSCs on up-regulating the levels of p-PI3K/PI3K, p-Akt/Akt, HIF-1α, vascular endothelial growth factor (VEGF), stromal cell-derived factor-1 (SDF-1), C-X-C motif chemokine receptor 4 (CXCR4), angiopoietin-4 (Ang-4), VEGF receptor (VEGFR), and transforming growth factor-β1 (TGF-β1), and down-regulating the levels of interleukin-1 β (IL-1β), IL-18 and IL-6 in wound tissues in chronic skin ulcers in vivo. Collectively, VAP promoted the growth, migration, invasion, and angiogenesis of ADSCs through activating PI3K/Akt/HIF-1α pathway, and VAP-PLGA enhanced the function of ADSCs in promoting wound healing in vivo, which was associated with angiogenesis, inflammation inhibition, and dermal collagen synthesis.
Collapse
Affiliation(s)
- Wen Jiang
- First Clinical School Medicine, Nanjing University of Chinese Medicine, Nanjing City, China
| | - Jun Zhang
- Department of Plastic Surgery, Affiliated Hospital Nanjing University of Chinese Medicine, Nanjing City, China
| | - Xudong Zhang
- Department of Aesthetic and Plastic Surgery, 903RD Hospital of Pla, Hangzhou City, China
| | - Chenghong Fan
- Aesthetic Surgery Department, Lishui Fan Chenghong Medical Aesthetic Clinic, Lishui City, China
| | - Jinlong Huang
- Department of Plastic Surgery, Affiliated Hospital Nanjing University of Chinese Medicine, Nanjing City, China
| |
Collapse
|
23
|
Zeng L, Zhou S, Chen C, Zhou LH, Shi X, Wu Z, Luo SK. Experimental study of fat derived pellets promoting wound healing in rats. Bioengineered 2021; 12:12323-12331. [PMID: 34787072 PMCID: PMC8810055 DOI: 10.1080/21655979.2021.2000257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To observe the effect of fat-derived pellets (FDP) on wound healing in rats, the inguinal fat of rats was obtained, and the FDP were obtained after centrifugation. The cell activity and growth factor secretion of FDP were measured. The wounds in rats were created, and FDP was used to treat the wounds of rats. The phenotype of macrophages and the expression of angiogenic factors expression in wounds were measured. The cell viability in FDP remains in high level after centrifugation and the expression of vascular endothelial growth factor (VEGF) and Basic Fibroblast Growth Factor (bFGF) from FDP was observed in vitro. The FDP significantly promoted the wound healing of rats compared with that in control groups. Moreover, the expression of M2 macrophages and VEGF in FDP group were significantly higher than that in the control group. FDP is a kind of stem cell product, which can be obtained from adipose tissue by physical centrifugation. The cytotherapeutic effect of FDP makes it a promising product for wound healing in clinics.
Collapse
Affiliation(s)
- Li Zeng
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou City, 510317
| | - Shaolong Zhou
- Yichun University, No. 576, Xuefu Road, Yichun, Jiangxi, China
| | - Chen Chen
- Yichun University, No. 576, Xuefu Road, Yichun, Jiangxi, China
| | - Lin-Hua Zhou
- Yichun University, No. 576, Xuefu Road, Yichun, Jiangxi, China
| | - Xiujiang Shi
- Yichun University, No. 576, Xuefu Road, Yichun, Jiangxi, China
| | - Zongjian Wu
- Yichun University, No. 576, Xuefu Road, Yichun, Jiangxi, China
| | - Sheng-Kang Luo
- Department of Plastic and Reconstructive Surgery, Guangdong Second Provincial General Hospital, Guangzhou City, 510317
| |
Collapse
|
24
|
Zhang L, Yan X, An L, Wang M, Xu X, Ma Z, Nie M, Du F, Zhang J, Yu S. Novel pneumatically assisted atomization device for living cell delivery: application of sprayed mesenchymal stem cells for skin regeneration. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00144-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
25
|
Abstract
Numerous individuals suffer from impaired wound healing, such as chronic ulcers, severe burns and immune disorders, resulting in both public health and economic burdens. Skin is the first line of defense and the largest organ of the human body, however, an incomplete understanding of underlying cellular and molecular mechanisms of dermal repair leads to a lack of effective therapy for healing impaired wounds. There are strong clinical and social needs for improved therapeutic approaches to enhance endogenous tissue repair and regenerative capacity. The purpose of this review is to illuminate the cellular and molecular aspects of the healing process and highlight potential therapeutic strategies to accelerate translational research and the development of clinical therapies in dermal wounds.
Collapse
Affiliation(s)
- Fan Yang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xiangjun Bai
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xiaojing Dai
- MD Anderson Cancer Center, The Advanced Technology Genomics Core, Houston, TX 77030, USA
| | - Yong Li
- Department of Orthopedic Surgery & Biomedical Engineering, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI 49008, USA
| |
Collapse
|
26
|
Ayavoo T, Murugesan K, Gnanasekaran A. Roles and mechanisms of stem cell in wound healing. Stem Cell Investig 2021; 8:4. [PMID: 33829056 DOI: 10.21037/sci-2020-027] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 01/27/2021] [Indexed: 01/02/2023]
Abstract
Wound healing phases comprise of highly synchronized process that begins due to a damage and restores the integrity of the injured tissues. Wound healing reduces the damage in tissue and supply sufficient oxygen and tissue perfusion, provide proper nourishment and humid wound healing atmosphere to re-establish the essential status of exaggerated parts. The untreated wound becomes susceptible for pus development, bacterial infection and complications like sepsis. Traditional and modern approaches are in practice to treat acute, open and chronic injuries, however, present wound care management has met with challenges and minimal positive effects. Stem cells have possible wound healing capability to overwhelm restrictions of the current wound care practices as it produces faster tissue regeneration in wound repair. Stem cells are unspecialized cells derived from adult body tissues and embryos that differentiate into any cell of an organism and capable of self-regeneration. The understanding on molecular mechanisms of stem cells has become the central and promising field in scientific study. This review focuses on the pre-existing traditional and modern treatments for wound healing, and types and roles of stem cells in wound care management. This review also focuses on the fundamental molecular characterization and factors influencing the molecular mechanisms of stem cells in wound healing.
Collapse
Affiliation(s)
- Thurga Ayavoo
- Centre of Research for Infectious Diseases and Phytochemical Studies, Quest International University Perak, Perak, Malaysia
| | - Karthikeyan Murugesan
- Centre of Research for Infectious Diseases and Phytochemical Studies, Quest International University Perak, Perak, Malaysia.,Department of Microbiology, Faculty of Medicine, Quest International University Perak, Perak, Malaysia
| | - Ashok Gnanasekaran
- Centre of Research for Infectious Diseases and Phytochemical Studies, Quest International University Perak, Perak, Malaysia.,Department of Microbiology, Faculty of Medicine, Quest International University Perak, Perak, Malaysia
| |
Collapse
|
27
|
Shafiee S, Heidarpour M, Sabbagh S, Amini E, Saffari H, Dolati S, Meamar R. Stem cell transplantation therapy for diabetic foot ulcer: a narrative review. ASIAN BIOMED 2021; 15:3-18. [PMID: 37551298 PMCID: PMC10388749 DOI: 10.2478/abm-2021-0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Diabetes mellitus is a chronic metabolic disease associated with high cardiovascular risk. A vascular complication of diabetes is foot ulcers. Diabetic foot ulcers are prevalent and substantially reduce the quality of life of patients who have them. Currently, diabetic foot ulcer is a major problem for wound care specialists, and its treatment requires considerable health care resources. So far, various therapeutic modalities have been proposed to treat diabetic foot ulcers and one of them is stem cell-based therapy. Stem cell-based therapy has shown great promise for the treatment of diabetic foot ulcers. This strategy has been shown to be safe and effective in both preclinical and clinical trials. In this review, we provide an overview of the stem cell types and possible beneficial effects of stem cell transplantation therapy for diabetic foot ulcers, and an overview of the current status of stem cell research in both preclinical and clinical trial stages of treatment strategies for diabetic foot ulcers.
Collapse
Affiliation(s)
- Sahar Shafiee
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Maryam Heidarpour
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan81737-35131, Iran
| | - Sima Sabbagh
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Elham Amini
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Hanieh Saffari
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Sara Dolati
- Clinical Development Research Center, Islamic Azad University, Najafabad Branch, Isfahan81737-35131, Iran
| | - Rokhsareh Meamar
- Isfahan Clinical Toxicology Research Center, Isfahan University of Medical Sciences, Isfahan81746-73461, Iran
| |
Collapse
|
28
|
Sharma P, Kumar A, Dey AD, Behl T, Chadha S. Stem cells and growth factors-based delivery approaches for chronic wound repair and regeneration: A promise to heal from within. Life Sci 2021; 268:118932. [PMID: 33400933 DOI: 10.1016/j.lfs.2020.118932] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/03/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023]
Abstract
The sophisticated chain of cellular and molecular episodes during wound healing includes cell migration, cell proliferation, deposition of extracellular matrix, and remodelling and are onerous to replicate. Encapsulation of growth factors (GFs) and Stem cell-based (SCs) has been proclaimed to accelerate healing by transforming every phase associated with wound healing to enhance skin regeneration. Therapeutic application of mesenchymal stem cells (MSCs), embryonic stem cells (ESCs) and induced pluripotent stem cells (PSCs) provides aid in wound fixing, tissue integrity restoration and function of impaired tissue. Several scientific studies have established the essential role GFs in wound healing and their reduced degree in the chronic wound. The overall limitation includes half-life, unfriendly microhabitat abundant with protease, and inadequate delivery approaches results in decreased delivery of effective amounts in a suitable time-based fashion. Advancements in the area of reformative medicine as well as tissue engineering have offered techniques competent of dispensing SCs and GFs in site-oriented manner. The progress in nanotechnology-based approaches attracts researcher to study and evaluate the potential of this SCs and GFs based therapy in chronic wounds. These techniques embrace the polymeric regime viz., nano-formulations, hydrogels, liposomes, scaffolds, nanofibers, metallic nanoparticles, lipid-based nanoparticles and dendrimers that have established better retort through targeting tissues when GFs and SCs are transported via these humans made devices. Assumed the current problems, improvements in delivery approaches and difficulties offered by chronic wounds, we hope to show that encapsulation of SCs and GFs loaded nanoformulations therapies is the rational next step in improving wound care.
Collapse
Affiliation(s)
- Preety Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Asmita Deka Dey
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
29
|
Shi X, Jiang L, Zhao X, Chen B, Shi W, Cao Y, Chen Y, Li X, He Y, Li C, Liu X, Li X, Lu H, Chen C, Liu J. Adipose-Derived Stromal Cell-Sheets Sandwiched, Book-Shaped Acellular Dermal Matrix Capable of Sustained Release of Basic Fibroblast Growth Factor Promote Diabetic Wound Healing. Front Cell Dev Biol 2021; 9:646967. [PMID: 33842472 PMCID: PMC8027315 DOI: 10.3389/fcell.2021.646967] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The management of diabetic wounds is a therapeutic challenge in clinical settings. Current tissue engineering strategies for diabetic wound healing are insufficient, owing to the lack of an appropriate scaffold that can load a large number of stem cells and induce the interaction of stem cells to form granulation tissue. Herein we fabricated a book-shaped decellularized dermal matrix (BDDM), which shows a high resemblance to native dermal tissue in terms of its histology, microstructure, and ingredients, is non-cytotoxic and low-immunogenic, and allows adipose-derived stromal cell (ASC) attachment and proliferation. Then, a collagen-binding domain (CBD) capable of binding collagen was fused into basic fibroblast growth factor (bFGF) to synthetize a recombinant growth factor (termed as CBD-bFGF). After that, CBD-bFGF was tethered onto the collagen fibers of BDDM to improve its endothelial inducibility. Finally, a functional scaffold (CBD-bFGF/BDDM) was fabricated. In vitro and in vivo experiments demonstrated that CBD-bFGF/BDDM can release tethered bFGF with a sustained release profile, steadily inducing the interaction of stem cells down to endothelial differentiation. ASCs were cultured to form a cell sheet and then sandwiched by CBD-bFGF/BDDM, thus enlarging the number of stem cells loaded into the scaffold. Using a rat model, the ASC sheets sandwiched with CBD-bFGF/BDDM (ASCs/CBD-bFGF/BDDM) were capable of enhancing the formation of granulation tissue, promoting angiogenesis, and facilitating collagen deposition and remodeling. Therefore, the findings of this study demonstrate that ASCs/CBD-bFGF/BDDM could be applicable for diabetic wound healing.
Collapse
Affiliation(s)
- Xin Shi
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Liyuan Jiang
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Zhao
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Bei Chen
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Wei Shi
- Department of Emergency, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yanpeng Cao
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Yaowu Chen
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Xiying Li
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Yusheng He
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Chengjie Li
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Xiaoren Liu
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
| | - Xing Li
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Lu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Can Chen
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Can Chen,
| | - Jun Liu
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Southern Medical University, Chenzhou, China
- The First School of Clinical Medicine, Xiangnan University, Chenzhou, China
- Jun Liu,
| |
Collapse
|
30
|
Shafiee A, Cavalcanti AS, Saidy NT, Schneidereit D, Friedrich O, Ravichandran A, De-Juan-Pardo EM, Hutmacher DW. Convergence of 3D printed biomimetic wound dressings and adult stem cell therapy. Biomaterials 2020; 268:120558. [PMID: 33307369 DOI: 10.1016/j.biomaterials.2020.120558] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Biomimetically designed medical-grade polycaprolactone (mPCL) dressings are 3D-printed with pore architecture and anisotropic mechanical characteristics that favor skin wound healing with reduced scarring. Melt electrowritten mPCL dressings are seeded with human gingival tissue multipotent mesenchymal stem/stromal cells and cryopreserved using a clinically approved method. The regenerative potential of fresh or frozen cell-seeded mPCL dressing is compared in a splinted full-thickness excisional wound in a rat model over six weeks. The application of 3D-printed mPCL dressings decreased wound contracture and significantly improved skin regeneration through granulation and re-epithelialization compared to control groups. Combining 3D-printed biomimetic wound dressings and precursor cell delivery enhances physiological wound closure with reduced scar tissue formation.
Collapse
Affiliation(s)
- Abbas Shafiee
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia; UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, 4102, Australia; Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD, 4029, Australia.
| | - Amanda S Cavalcanti
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Navid T Saidy
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia; The University of Queensland, School of Dentistry, Herston, Queensland, Australia
| | - Dominik Schneidereit
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nuremberg, Paul-Gordan-Str.3, 91052, Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nuremberg, Paul-Gordan-Str.3, 91052, Erlangen, Germany
| | - Akhilandeshwari Ravichandran
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Elena M De-Juan-Pardo
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Kelvin Grove, Brisbane, QLD, 4059, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, Queensland University of Technology (QUT), Kelvin Grove, QLD, 4059, Australia.
| |
Collapse
|
31
|
Nour S, Imani R, Chaudhry GR, Sharifi AM. Skin wound healing assisted by angiogenic targeted tissue engineering: A comprehensive review of bioengineered approaches. J Biomed Mater Res A 2020; 109:453-478. [PMID: 32985051 DOI: 10.1002/jbm.a.37105] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/23/2020] [Accepted: 09/26/2020] [Indexed: 12/16/2022]
Abstract
Skin injuries and in particular, chronic wounds, are one of the major prevalent medical problems, worldwide. Due to the pivotal role of angiogenesis in tissue regeneration, impaired angiogenesis can cause several complications during the wound healing process and skin regeneration. Therefore, induction or promotion of angiogenesis can be considered as a promising approach to accelerate wound healing. This article presents a comprehensive overview of current and emerging angiogenesis induction methods applied in several studies for skin regeneration, which are classified into the cell, growth factor, scaffold, and biological/chemical compound-based strategies. In addition, the advantages and disadvantages of these angiogenic strategies along with related research examples are discussed in order to demonstrate their potential in the treatment of wounds.
Collapse
Affiliation(s)
- Shirin Nour
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - G Rasul Chaudhry
- OU-WB Institute for Stem Cell and Regenerative Medicine, Department of Biological Sciences, Oakland University, Rochester, Michigan, USA
| | - Ali Mohammad Sharifi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.,Tissue Engineering Group (NOCERAL), Department of Orthopedics Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Ahangar P, Mills SJ, Cowin AJ. Mesenchymal Stem Cell Secretome as an Emerging Cell-Free Alternative for Improving Wound Repair. Int J Mol Sci 2020; 21:ijms21197038. [PMID: 32987830 PMCID: PMC7583030 DOI: 10.3390/ijms21197038] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
The use of mesenchymal stem cells (MSC) for the treatment of cutaneous wounds is currently of enormous interest. However, the broad translation of cell therapies into clinical use is hampered by their efficacy, safety, manufacturing and cost. MSCs release a broad repertoire of trophic factors and immunomodulatory cytokines, referred to as the MSC secretome, that has considerable potential for the treatment of cutaneous wounds as a cell-free therapy. In this review, we outline the current status of MSCs as a treatment for cutaneous wounds and introduce the potential of the MSC secretome as a cell-free alternative for wound repair. We discuss the challenges and provide insights and perspectives for the future development of the MSC secretome as well as identify its potential clinical translation into a therapeutic treatment.
Collapse
Affiliation(s)
- Parinaz Ahangar
- Future Industries Institute, University of South Australia, Adelaide, SA 5000, Australia; (P.A.); (S.J.M.)
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Stuart J. Mills
- Future Industries Institute, University of South Australia, Adelaide, SA 5000, Australia; (P.A.); (S.J.M.)
| | - Allison J. Cowin
- Future Industries Institute, University of South Australia, Adelaide, SA 5000, Australia; (P.A.); (S.J.M.)
- Correspondence: ; Tel.: +61-8-8302-5018
| |
Collapse
|
33
|
Multipotent adult progenitor cells grown under xenobiotic-free conditions support vascularization during wound healing. Stem Cell Res Ther 2020; 11:389. [PMID: 32894199 PMCID: PMC7487685 DOI: 10.1186/s13287-020-01912-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022] Open
Abstract
Background Cell therapy has been evaluated pre-clinically and clinically as a means to improve wound vascularization and healing. While translation of this approach to clinical practice ideally requires the availability of clinical grade xenobiotic-free cell preparations, studies proving the pre-clinical efficacy of the latter are mostly lacking. Here, the potential of xenobiotic-free human multipotent adult progenitor cell (XF-hMAPC®) preparations to promote vascularization was evaluated. Methods The potential of XF-hMAPC cells to support blood vessel formation was first scored in an in vivo Matrigel assay in mice. Next, a dose-response study was performed with XF-hMAPC cells in which they were tested for their ability to support vascularization and (epi) dermal healing in a physiologically relevant splinted wound mouse model. Results XF-hMAPC cells supported blood vessel formation in Matrigel by promoting the formation of mature (smooth muscle cell-coated) vessels. Furthermore, XF-hMAPC cells dose-dependently improved wound vascularization associated with increasing wound closure and re-epithelialization, granulation tissue formation, and dermal collagen organization. Conclusions Here, we demonstrated that the administration of clinical-grade XF-hMAPC cells in mice represents an effective approach for improving wound vascularization and healing that is readily applicable for translation in humans.
Collapse
|
34
|
Ahangar P, Mills SJ, Smith LE, Strudwick XL, Ting AE, Vaes B, Cowin AJ. Human multipotent adult progenitor cell-conditioned medium improves wound healing through modulating inflammation and angiogenesis in mice. Stem Cell Res Ther 2020; 11:299. [PMID: 32680566 PMCID: PMC7368692 DOI: 10.1186/s13287-020-01819-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/15/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Stem cell therapies have been widely investigated for their healing effects. However, the translation of these therapies has been hampered by the requirement to deliver live allogeneic or autologous cells directly to the wound in a clinical setting. Multipotent adult progenitor cells (MAPC® cells) are a subpopulation of bone marrow-derived adherent stem cells that secrete a wide range of factors known to accelerate the wound healing process. The aim of this study was to determine the impact of MAPC cells secretome on healing outcomes without the presence of MAPC cells. METHODS The effect of MAPC-conditioned medium (MAPC-CM) on the capacity of keratinocytes, fibroblasts and endothelial cells to migrate and proliferate was determined in vitro using scratch wound closure and WST1 assay, respectively. The effect of MAPC-CM on collagen deposition and angiogenesis was also assessed using in vitro methods. Additionally, two excisional wounds were created on the dorsal surface of mice (n = 8/group) and 100 μL of 20× MAPC-CM were intradermally injected to the wound margins. Wound tissues were collected at 3, 7 and 14 days post-wounding and stained with H&E for microscopic analysis. Immunohistochemistry was performed to investigate inflammation, angiogenesis and collagen deposition in the wounds. RESULTS Skin fibroblasts, keratinocytes and endothelial cells treated with MAPC-CM all showed improved rates of scratch closure and increased cellular proliferation. Moreover, fibroblasts treated with MAPC-CM deposited more collagens I and III and endothelial cells treated with MAPC-CM showed increased capillary tube formation. Murine excisional wounds intradermally injected with MAPC-CM showed a significant reduction in the wound area and an increase in the rate of reepithelialisation. The results also showed that inflammatory cell infiltration was decreased while an increase in angiogenesis, as well as collagens I and III expressions, was observed. CONCLUSION These findings suggest that factors produced by MAPC cells can have an important effect on cutaneous wound healing by affecting skin cell proliferation and migration, balancing inflammation and improving the formation of extracellular matrix and angiogenesis. Development of stem cell-free therapy for the treatment of wounds may be a more clinically translatable approach for improving healing outcomes.
Collapse
Affiliation(s)
- Parinaz Ahangar
- Future Industries Institute, University of South Australia, Adelaide, SA, 5000, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide, SA, 5000, Australia
| | - Stuart J Mills
- Future Industries Institute, University of South Australia, Adelaide, SA, 5000, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide, SA, 5000, Australia
| | - Louise E Smith
- Future Industries Institute, University of South Australia, Adelaide, SA, 5000, Australia.,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide, SA, 5000, Australia
| | - Xanthe L Strudwick
- Future Industries Institute, University of South Australia, Adelaide, SA, 5000, Australia
| | | | - Bart Vaes
- ReGenesys BVBA, Bio-Incubator Leuven, Gaston Geenslaan 1, 3001, Heverlee, Belgium
| | - Allison J Cowin
- Future Industries Institute, University of South Australia, Adelaide, SA, 5000, Australia. .,Cell Therapy Manufacturing Cooperative Research Centre, Adelaide, SA, 5000, Australia.
| |
Collapse
|
35
|
Gorecka J, Gao X, Fereydooni A, Dash BC, Luo J, Lee SR, Taniguchi R, Hsia HC, Qyang Y, Dardik A. Induced pluripotent stem cell-derived smooth muscle cells increase angiogenesis and accelerate diabetic wound healing. Regen Med 2020; 15:1277-1293. [PMID: 32228292 PMCID: PMC7304438 DOI: 10.2217/rme-2019-0086] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: To assess the potential of human induced pluripotent stem cell-derived smooth muscle cells (hiPSC-SMC) to accelerate diabetic wound healing. Methods: hiPSC-SMC were embedded in 3D collagen scaffolds and cultured in vitro for 72 h; scaffolds were then applied to diabetic, nude mouse, splinted back wounds to assess in vivo healing. Cultured medium after scaffold incubation was collected and analyzed for expression of pro-angiogenic cytokines. Results: hiPSC-SMC secrete increased concentration of pro-angiogenic cytokines, compared with murine adipose derived stem cells. Delivery of hiPSC-SMC-containing collagen scaffolds accelerates diabetic wound healing and is associated with an increased number of total and M2 type macrophages. Conclusion: hiPSC-SMC promote angiogenesis and accelerate diabetic wound healing, making them a promising new candidate for treatment of diabetic wounds.
Collapse
Affiliation(s)
- Jolanta Gorecka
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Xixiang Gao
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA.,Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Arash Fereydooni
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Biraja C Dash
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jiesi Luo
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA.,Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA.,Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519 USA.,Department of Pathology, YaleUniversity, New Haven, CT 06520, USA
| | - Shin Rong Lee
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Ryosuke Taniguchi
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA
| | - Henry C Hsia
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT 06511, USA.,Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA.,Vascular Biology & Therapeutics Program, Yale School of Medicine, New Haven, CT 06519 USA.,Department of Pathology, YaleUniversity, New Haven, CT 06520, USA
| | - Alan Dardik
- Vascular Biology & Therapeutics Program & The Department of Surgery, Yale School of Medicine, Yale University, New Haven, CT 06519 USA.,Section of Vascular & Endovascular Surgery, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
36
|
Bi H, Li H, Zhang C, Mao Y, Nie F, Xing Y, Sha W, Wang X, Irwin DM, Tan H. Stromal vascular fraction promotes migration of fibroblasts and angiogenesis through regulation of extracellular matrix in the skin wound healing process. Stem Cell Res Ther 2019; 10:302. [PMID: 31623669 PMCID: PMC6798485 DOI: 10.1186/s13287-019-1415-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/07/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A refractory wound is a typical complication of diabetes and is a common outcome after surgery. Current approaches have difficulty in improving wound healing. Recently, non-expanded stromal vascular fraction (SVF), which is derived from mature fat, has opened up new directions for the treatment of refractory wound healing. The aim of the current study is to systematically investigate the impact of SVF on wound healing, including the rate and characteristics of wound healing, ability of fibroblasts to migrate, and blood transport reconstruction, with a special emphasis on their precise molecular mechanisms. METHODS SVF was isolated by digestion, followed by filtration and centrifugation, and then validated by immunocytochemistry, a MTS proliferation assay and multilineage potential analysis. A wound model was generated by creating 6-mm-diameter wounds, which include a full skin defect, on the backs of streptozocin-induced hyperglycemic mice. SVF or human adipose-derived stem cell (hADSC) suspensions were subcutaneously injected, and the wounds were characterized over a 9-day period by photography and measurements. A scratch test was used to determine whether changes in the migratory ability of fibroblasts occurred after co-culture with hADSCs. Angiogenesis was observed with human umbilical vein endothelial cells. mRNA from fibroblasts, endotheliocyte, and skin tissue were sequenced by high-throughput RNAseq, and differentially expressed genes, and pathways, potentially regulated by SVF or hADSCs were bioinformatically analyzed. RESULTS Our data show that hADSCs have multiple characteristics of MSC. SVF and hADSCs significantly improved wound healing in hyperglycemic mice. hADSCs improve the migratory ability of fibroblasts and capillary structure formation in HUVECs. SVF promotes wound healing by focusing on angiogenesis and matrix remodeling. CONCLUSIONS Both SVF and hADSCs improve the function of fibroblast and endothelial cells, regulate gene expression, and promote skin healing. Various mechanisms likely are involved, including migration of fibroblasts, tubulogenesis of endothelial cells through regulation of cell adhesion, and cytokine pathways.
Collapse
Affiliation(s)
- Hongsen Bi
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191 China
| | - Hui Li
- Department of Pharmacology, Peking University, Health Science Center, Beijing, 100191 China
| | - Chen Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191 China
| | - Yiqing Mao
- Department of Pharmacology, Peking University, Health Science Center, Beijing, 100191 China
| | - Fangfei Nie
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191 China
| | - Ying Xing
- Department of Pharmacology, Peking University, Health Science Center, Beijing, 100191 China
| | - Wuga Sha
- Department of Pharmacology, Peking University, Health Science Center, Beijing, 100191 China
| | - Xi Wang
- Department of Pharmacology, Peking University, Health Science Center, Beijing, 100191 China
| | - David M. Irwin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S1A8 Canada
| | - Huanran Tan
- Department of Pharmacology, Peking University, Health Science Center, Beijing, 100191 China
| |
Collapse
|
37
|
Kathawala MH, Ng WL, Liu D, Naing MW, Yeong WY, Spiller KL, Van Dyke M, Ng KW. Healing of Chronic Wounds: An Update of Recent Developments and Future Possibilities. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:429-444. [PMID: 31068101 DOI: 10.1089/ten.teb.2019.0019] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chronic wounds are the result of disruptions in the body's usual process of healing. They are not only a source of significant pain and discomfort but also, more importantly, an unguarded port of entry for pathogens into the body. While our current understanding of this phenomenon is far from complete, findings in physiological patterns and advancements in wound healing technologies have helped develop wound management and healing solutions to this long-standing medical challenge. This review presents an overview of known wound healing mechanics, abnormalities that lead to chronic wounds, and a summary of established and new wound healing technologies. Various approaches to heal wounds are discussed, from dermal replacements to advanced biomaterial-based treatments, from cell-, synthetic-, and composite-based approaches to preclinical approaches, which make developing such products possible. While tested breakthrough products are described, the authors focused more on recently developed innovations, which are at varying stages of maturity. The review concludes with a note on future perspectives and opinions on where the field and industry are headed and where they should be. Impact Statement Wound healing is an important area of research and clinical practice, and has captured the attention of tissue engineers since the nascent beginnings of the discipline. Tissue-engineered skin was the first FDA-approved product, achieved in 1996. Despite this success, and the passage of time, healing wounds, particularly chronic wounds, remains a vexing challenge. This comprehensive review article will provide readers with a synopsis of current issues, research approaches, animal models, technologies, and products that span the continuum from early development to clinical studies, in the hope of fueling new interests and ideas to overcome this long-standing medical challenge.
Collapse
Affiliation(s)
| | - Wei Long Ng
- Singapore Centre for 3D Printing (SC3DP), School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Dan Liu
- Singapore Institute of Manufacturing Technology (SIMTECH), Singapore, Singapore
| | - May Win Naing
- Singapore Institute of Manufacturing Technology (SIMTECH), Singapore, Singapore
| | - Wai Yee Yeong
- Singapore Centre for 3D Printing (SC3DP), School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, Singapore
| | - Kara L Spiller
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania
| | - Mark Van Dyke
- Department of Biomedical Engineering and Mechanics (BEAM), Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Kee Woei Ng
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore.,Skin Research Institute of Singapore (SRIS), Singapore, Singapore.,Environmental Chemistry & Materials Centre, Nanyang Environment and Water Research Institute (NEWRI), Singapore, Singapore
| |
Collapse
|
38
|
Wang X, Jiang B, Sun H, Zheng D, Zhang Z, Yan L, Li E, Wu Y, Xu RH. Noninvasive application of mesenchymal stem cell spheres derived from hESC accelerates wound healing in a CXCL12-CXCR4 axis-dependent manner. Theranostics 2019; 9:6112-6128. [PMID: 31534540 PMCID: PMC6735514 DOI: 10.7150/thno.32982] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/29/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSC) derived from adult tissues effectively promote wound healing. However, MSC quality varies, and the quantity of MSC is limited, as MSC are acquired through donations. Moreover, the survival and functioning of dissociated MSC delivered to an inflammatory lesion are subject to challenges. Methods: Here, spheres (EMSCSp) generated from human embryonic stem cell-derived MSC (EMSC) were directly dropped onto excised wounds in mice; the effects of EMSCSp were compared to those of dissociated EMSC (EMSCDiss). Following transplantation, we measured the extent of wound closure, dissected the histological features of the wounds, determined transcriptomic changes in cells isolated from the treated and control wounds, and evaluated the molecular mechanism of the effects of EMSC. Results: The application of EMSCSp onto murine dermal wounds substantially increased survival and efficacy of EMSC compared to the topical application of EMSCDiss. RNA sequencing (RNA-Seq) of cells isolated from the wounds highlighted the involvement of CXCL12-CXCR4 signaling in the effects of EMSCSp, which was verified in EMSC via CXCL12 knockdown and in target cells (vascular endothelial cells, epithelial keratinocytes, and macrophages) via CXCR4 inhibition. Finally, we enhanced the biosafety of EMSCSp by engineering cells with an inducible suicide gene. Conclusions: Together, these data suggest the topical application of EMSCSp as an unlimited, quality-assured, safe, and noninvasive therapy for wound healing and the CXCL12-CXCR4 axis as a key player in this treatment.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Bin Jiang
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Huiyan Sun
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Dejin Zheng
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Zhenwu Zhang
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Li Yan
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Enqin Li
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yaojiong Wu
- The Shenzhen Key Laboratory of Health Sciences and Technology, Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Ren-He Xu
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
39
|
Venugopal B, Shenoy SJ, Mohan S, Anil Kumar PR, Kumary TV. Bioengineered corneal epithelial cell sheet from mesenchymal stem cells-A functional alternative to limbal stem cells for ocular surface reconstruction. J Biomed Mater Res B Appl Biomater 2019; 108:1033-1045. [PMID: 31400069 DOI: 10.1002/jbm.b.34455] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/25/2019] [Accepted: 07/17/2019] [Indexed: 12/13/2022]
Abstract
Limbal stem cell deficiency (LSCD) is the loss of limbal stem cells that reside in the corneoscleral junction resulting in vision loss or blindness. Bilateral LSCD is usually treated by allogeneic corneal transplantation, with instances of tissue rejection or failure in long-term follow-up. This study aims to use adipose mesenchymal stem cells (ASC) as an alternative autologous cell source for treating bilateral limbal deficiency conditions. ASCs derived from rabbit fat tissue were differentiated into corneal epithelial lineage using limbal explant condition media. Apart from transdifferentiation, ASC sheets were developed to facilitate effective delivery of these cells to the damage site. A thermoresponsive polymer N-isopropylacrylamide-co-glycidylmethacrylate (NGMA) was synthesized and characterized to demonstrate ASC sheet formation. Transdifferentiated ASCs showed positive expression of corneal epithelial marker CK3/12 on immunostaining, supported by gene expression studies. in vivo studies by transplanting cell sheet in rabbit models of corneal injury showed clear and smooth cornea in comparison to the sham models. Histology revealed a sheet of cells aligned and integrated on to the injured corneal surface, 1 month posttransplantation. Identifying ASCs as an alternative cell source along with cell sheet technology will be a novel step in the field of corneal surface therapies.
Collapse
Affiliation(s)
- Balu Venugopal
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Sachin J Shenoy
- Division of in vivo Modes and Testing, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - Sumitha Mohan
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - P R Anil Kumar
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | - T V Kumary
- Division of Tissue Culture, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| |
Collapse
|
40
|
Secretome of Adipose Tissue-Derived Stem Cells (ASCs) as a Novel Trend in Chronic Non-Healing Wounds: An Overview of Experimental In Vitro and In Vivo Studies and Methodological Variables. Int J Mol Sci 2019; 20:ijms20153721. [PMID: 31366040 PMCID: PMC6696601 DOI: 10.3390/ijms20153721] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/27/2019] [Accepted: 07/28/2019] [Indexed: 12/11/2022] Open
Abstract
Wound healing is a complex process with a linear development that involves many actors in a multistep timeline commonly divided into four stages: Hemostasis, inflammation, proliferation, and remodeling. Chronic non-healing wounds fail to progress beyond the inflammatory phase, thus precluding the next steps and, ultimately, wound repair. Many intrinsic or extrinsic factors may contribute to such an occurrence, including patient health conditions, age-related diseases, metabolic deficiencies, advanced age, mechanical pressure, and infections. Great interest is being focused on the adipose tissue-derived stem cell’s (ASC) paracrine activity for its potential therapeutic impact on chronic non-healing wounds. In this review, we summarize the results of in vitro and in vivo experimental studies on the pro-wound healing effects of ASC-secretome and/or extracellular vesicles (EVs). To define an overall picture of the available literature data, experimental conditions and applied methodologies are described as well as the in vitro and in vivo models chosen in the reported studies. Even if a comparative analysis of the results obtained by the different groups is challenging due to the large variability of experimental conditions, the available findings are undoubtedly encouraging and fully support the use of cell-free therapies for the treatment of chronic non-healing wounds.
Collapse
|
41
|
Millán-Rivero JE, Martínez CM, Romecín PA, Aznar-Cervantes SD, Carpes-Ruiz M, Cenis JL, Moraleda JM, Atucha NM, García-Bernal D. Silk fibroin scaffolds seeded with Wharton's jelly mesenchymal stem cells enhance re-epithelialization and reduce formation of scar tissue after cutaneous wound healing. Stem Cell Res Ther 2019; 10:126. [PMID: 31029166 PMCID: PMC6487033 DOI: 10.1186/s13287-019-1229-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/25/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The treatment of extensive and/or chronic skin wounds is a widespread and costly public health problem. Mesenchymal stem cells (MSCs) have been proposed as a potential cell therapy for inducing wound healing in different clinical settings, alone or in combination with biosynthetic scaffolds. Among them, silk fibroin (SF) seeded with MSCs has been shown to have increased efficacy in skin wound healing experimental models. METHODS In this report, we investigated the wound healing effects of electrospun SF scaffolds cellularized with human Wharton's jelly MSCs (Wj-MSCs-SF) using a murine excisional wound splinting model. RESULTS Immunohistopathological examination after transplant confirmed the presence of infiltrated human fibroblast-like CD90-positive cells in the dermis of the Wj-MSCs-SF-treated group, yielding neoangiogenesis, decreased inflammatory infiltrate and myofibroblast proliferation, less collagen matrix production, and complete epidermal regeneration. CONCLUSIONS These findings indicate that Wj-MSCs transplanted in the wound bed on a silk fibroin scaffold contribute to the generation of a well-organized and vascularized granulation tissue, enhance reepithelization of the wound, and reduce the formation of fibrotic scar tissue, highlighting the potential therapeutic effects of Wj-MSC-based tissue engineering approaches to non-healing wound treatment.
Collapse
Affiliation(s)
- José E. Millán-Rivero
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
- Internal Medicine Department, Medicine School, University of Murcia, Avenida Buenavista s/n. El Palmar, Murcia, Spain
| | - Carlos M. Martínez
- Experimental Pathology Unit, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain
| | - Paola A. Romecín
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
| | - Salvador D. Aznar-Cervantes
- Biotechnology Department, Instituto Murciano de Investigación y Desarrollo Agrario y Alimentario (IMIDA), Murcia, Spain
| | - Marina Carpes-Ruiz
- Experimental Pathology Unit, Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain
| | - José L. Cenis
- Biotechnology Department, Instituto Murciano de Investigación y Desarrollo Agrario y Alimentario (IMIDA), Murcia, Spain
| | - Jose M. Moraleda
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
- Internal Medicine Department, Medicine School, University of Murcia, Avenida Buenavista s/n. El Palmar, Murcia, Spain
| | - Noemí M. Atucha
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
- Physiology Department, Medicine School, University of Murcia, Murcia, Spain
| | - David García-Bernal
- Hematopoietic Transplant and Cellular Therapy Unit, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Virgen de la Arrixaca University Hospital, University of Murcia, Murcia, Spain
- Internal Medicine Department, Medicine School, University of Murcia, Avenida Buenavista s/n. El Palmar, Murcia, Spain
| |
Collapse
|
42
|
Gorecka J, Kostiuk V, Fereydooni A, Gonzalez L, Luo J, Dash B, Isaji T, Ono S, Liu S, Lee SR, Xu J, Liu J, Taniguchi R, Yastula B, Hsia HC, Qyang Y, Dardik A. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res Ther 2019; 10:87. [PMID: 30867069 PMCID: PMC6416973 DOI: 10.1186/s13287-019-1185-1] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Wound healing is the physiologic response to a disruption in normal skin architecture and requires both spatial and temporal coordination of multiple cell types and cytokines. This complex process is prone to dysregulation secondary to local and systemic factors such as ischemia and diabetes that frequently lead to chronic wounds. Chronic wounds such as diabetic foot ulcers are epidemic with great cost to the healthcare system as they heal poorly and recur frequently, creating an urgent need for new and advanced therapies. Stem cell therapy is emerging as a potential treatment for chronic wounds, and adult-derived stem cells are currently employed in several commercially available products; however, stem cell therapy is limited by the need for invasive harvesting techniques, immunogenicity, and limited cell survival in vivo. Induced pluripotent stem cells (iPSC) are an exciting cell type with enhanced therapeutic and translational potential. iPSC are derived from adult cells by in vitro induction of pluripotency, obviating the ethical dilemmas surrounding the use of embryonic stem cells; they are harvested non-invasively and can be transplanted autologously, reducing immune rejection; and iPSC are the only cell type capable of being differentiated into all of the cell types in healthy skin. This review focuses on the use of iPSC in animal models of wound healing including limb ischemia, as well as their limitations and methods aimed at improving iPSC safety profile in an effort to hasten translation to human studies.
Collapse
Affiliation(s)
- Jolanta Gorecka
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Valentyna Kostiuk
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jiesi Luo
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Biraja Dash
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, PO Box 208062, New Haven, CT, 06520-8062, USA
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shun Ono
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shirley Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Shin Rong Lee
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jianbiao Xu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Jia Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Bogdan Yastula
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA
| | - Henry C Hsia
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, PO Box 208062, New Haven, CT, 06520-8062, USA
| | - Yibing Qyang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA.,Yale Stem Cell Center, Yale University, New Haven, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, USA.,Department of Pathology, Yale University, New Haven, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT, 06520-8089, USA.
| |
Collapse
|
43
|
Yeo GC, Kosobrodova E, Kondyurin A, McKenzie DR, Bilek MM, Weiss AS. Plasma‐Activated Substrate with a Tropoelastin Anchor for the Maintenance and Delivery of Multipotent Adult Progenitor Cells. Macromol Biosci 2018; 19:e1800233. [DOI: 10.1002/mabi.201800233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/19/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Giselle C. Yeo
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of Life and Environmental SciencesUniversity of Sydney NSW 2006 Australia
- Bosch InstituteUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Elena Kosobrodova
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- School of AerospaceMechanical and Mechatronic EngineeringUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Alexey Kondyurin
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - David R. McKenzie
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Marcela M. Bilek
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of PhysicsUniversity of Sydney NSW 2006 Australia
- School of AerospaceMechanical and Mechatronic EngineeringUniversity of Sydney NSW 2006 Australia
- Australian Institute of Nanoscale Science and TechnologyUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| | - Anthony S. Weiss
- Charles Perkins CentreUniversity of Sydney NSW 2006 Australia
- School of Life and Environmental SciencesUniversity of Sydney NSW 2006 Australia
- Bosch InstituteUniversity of Sydney NSW 2006 Australia
- The Cooperative Research Centre for Cell Therapy ManufacturingUniversity of South Australia City West Campus Adelaide SA 5000 Australia
| |
Collapse
|
44
|
Farias C, Lyman R, Hemingway C, Chau H, Mahacek A, Bouzos E, Mobed-Miremadi M. Three-Dimensional (3D) Printed Microneedles for Microencapsulated Cell Extrusion. Bioengineering (Basel) 2018; 5:E59. [PMID: 30065227 PMCID: PMC6164407 DOI: 10.3390/bioengineering5030059] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
Cell-hydrogel based therapies offer great promise for wound healing. The specific aim of this study was to assess the viability of human hepatocellular carcinoma (HepG2) cells immobilized in atomized alginate capsules (3.5% (w/v) alginate, d = 225 µm ± 24.5 µm) post-extrusion through a three-dimensional (3D) printed methacrylate-based custom hollow microneedle assembly (circular array of 13 conical frusta) fabricated using stereolithography. With a jetting reliability of 80%, the solvent-sterilized device with a root mean square roughness of 158 nm at the extrusion nozzle tip (d = 325 μm) was operated at a flowrate of 12 mL/min. There was no significant difference between the viability of the sheared and control samples for extrusion times of 2 h (p = 0.14, α = 0.05) and 24 h (p = 0.5, α = 0.05) post-atomization. Factoring the increase in extrusion yield from 21.2% to 56.4% attributed to hydrogel bioerosion quantifiable by a loss in resilience from 5470 (J/m³) to 3250 (J/m³), there was no significant difference in percentage relative payload (p = 0.2628, α = 0.05) when extrusion occurred 24 h (12.2 ± 4.9%) when compared to 2 h (9.9 ± 2.8%) post-atomization. Results from this paper highlight the feasibility of encapsulated cell extrusion, specifically protection from shear, through a hollow microneedle assembly reported for the first time in literature.
Collapse
Affiliation(s)
- Chantell Farias
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053-0583, USA.
| | - Roman Lyman
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053-0583, USA.
| | - Cecilia Hemingway
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053-0583, USA.
| | - Huong Chau
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053-0583, USA.
| | - Anne Mahacek
- SCU Maker Lab, Santa Clara University, Santa Clara, CA 95053-0583, USA.
| | - Evangelia Bouzos
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053-0583, USA.
| | - Maryam Mobed-Miremadi
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053-0583, USA.
| |
Collapse
|
45
|
Leitão L, Alves CJ, Alencastre IS, Sousa DM, Neto E, Conceição F, Leitão C, Aguiar P, Almeida-Porada G, Lamghari M. Bone marrow cell response after injury and during early stage of regeneration is independent of the tissue-of-injury in 2 injury models. FASEB J 2018; 33:857-872. [PMID: 30044924 DOI: 10.1096/fj.201800610rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Selectively recruiting bone marrow (BM)-derived stem and progenitor cells to injury sites is a promising therapeutic approach. The coordinated action of soluble factors is thought to trigger the mobilization of stem cells from the BM and recruit them to lesions to contribute to tissue regeneration. Nevertheless, the temporal response profile of the major cellular players and soluble factors involved in priming the BM and recruiting BM-derived cells to promote regeneration is unknown. We show that injury alters the BM cellular composition, introducing population-specific fluctuations during tissue regeneration. We demonstrate that injury causes an immediate, transient response of mesenchymal stromal cells and endothelial cells followed by a nonoverlapping increase in hematopoietic stem and progenitor cells. Moreover, BM reaction is identical whether the injury is inflicted on skin and muscle or also involves a bone defect, but these 2 injury paradigms trigger distinct systemic cytokine responses. Together, our results indicate that the BM response to injury in the early stages of regeneration is independent of the tissue-of-injury based on the 2 models used, but the injured tissue dictates the systemic cytokine response.-Leitão, L., Alves, C. J., Alencastre, I. S., Sousa, D. M., Neto, E., Conceição, F., Leitão, C., Aguiar, P., Almeida-Porada, G., Lamghari, M. Bone marrow cell response after injury and during early stage of regeneration is independent of the tissue-of-injury in 2 injury models.
Collapse
Affiliation(s)
- Luís Leitão
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cecília J Alves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Inês S Alencastre
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Daniela M Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Estrela Neto
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | - Francisco Conceição
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Catarina Leitão
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal; and
| | - Paulo Aguiar
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Meriem Lamghari
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
46
|
Zhou D, Yang T, Qian W, Xing M, Luo G. Study of the mechanism of environmentally friendly translucent balsa-modified lysozyme dressing for facilitating wound healing. Int J Nanomedicine 2018; 13:4171-4187. [PMID: 30046241 PMCID: PMC6054277 DOI: 10.2147/ijn.s165075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objective This study aimed to prepare an eco-friendly dressing using a balsa-derived membrane with lysozymes designed for antibacterial purposes. Methods The groups included controls, balsa (group A), translucent balsa (group B), translucent balsa–lysozymes (group C), and translucent balsa-modified lysozymes (group D). Physical and chemical methods were used to characterize the materials, and the function of the materials was evaluated by in vivo and in vitro experiments. Results Antibacterial activity against Escherichia coli and Staphylococcus aureus was ordered D > C > B ≈ A (P<0.05). Healing rates in the control, A, B, C, and D groups were 30.6%, 48.3%, 56.7%, 70.9%, and 79.2%, respectively at 7 days after injury. The lengths of new epithelia of the wound surface were ordered D > C > B ≈ A > control (P<0.05). Reverse-transcription polymerase chain reaction showed that expression of Wnt3a, β-catenin, and PCNA mRNA were ordered D > C > B ≈ A > control (P<0.05). The order of expression of PCNA was D > C > B ≈ A > control (P<0.05). There were no differences in GSK3β expression (P>0.05). The order of expression of axin was D < C < B ≈ A < control (P<0.05). The cell-migration rate at 24 hours was ordered D > C > B ≈ A > control (P<0.05). Conclusion This translucent balsa-modified lysozyme dressing is characterized by strong antibacterial properties, stable and persistent release, no cytotoxicity, and capacity to promote antibacterial ability and epithelial growth, as well as cell proliferation and migration.
Collapse
Affiliation(s)
- Daijun Zhou
- Institute of Burn Research; State Key Laboratory of Trauma, Burn and Combined Injury; Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China, ;
| | - Tao Yang
- Institute of Burn Research; State Key Laboratory of Trauma, Burn and Combined Injury; Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China, ;
| | - Wei Qian
- Institute of Burn Research; State Key Laboratory of Trauma, Burn and Combined Injury; Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China, ;
| | - Malcolm Xing
- Institute of Burn Research; State Key Laboratory of Trauma, Burn and Combined Injury; Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China, ;
| | - Gaoxing Luo
- Institute of Burn Research; State Key Laboratory of Trauma, Burn and Combined Injury; Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China, ;
| |
Collapse
|
47
|
Balqis U, Darmawi, Iskandar CD, Salim MN. Angiogenesis activity of Jatropha curcas L. latex in cream formulation on wound healing in mice. Vet World 2018; 11:939-943. [PMID: 30147263 PMCID: PMC6097565 DOI: 10.14202/vetworld.2018.939-943] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/05/2018] [Indexed: 01/29/2023] Open
Abstract
Aim: The aim of this research was to determine the angiogenesis activity of Jatropha curcas latex in cream formulation on CD34 immune expression during wound healing phase in mice skin. Materials and Methods: Amount of 36 2-month-old male mice were used between 30 and 40 g. To surgical procedures, wound skin incision was performed 2.0 cm in length until subcutaneous on the paravertebral of each animal. The treatment was carried under locally anesthetized with procaine cream. All mice were divided into four groups, namely the base cream as control group (A), sulfadiazine 0.1% cream (B), Jatropha curcas latex cream 10% (C), and J. curcas latex cream 15% (D). All groups were treated entire surface of wound. All experiments were performed twice a day for 10 days. Experiments were terminated on days 3, 7, and 10, respectively. The wound healing was assayed in stained histological section in immunohistochemical of the wounds. The CD34 expression was investigated under a microscope. Results: The results showed that the cream from 10% and 15% latex J. curcas revealed moderate immune reaction to CD34 on days 3 and 7 in wound healing of mice skin. Conclusion: We concluded that the cream from 10% and 15% latex J. curcas has potential as angiogenesis activity in wound healing of mice skin.
Collapse
Affiliation(s)
- Ummu Balqis
- Laboratory of Pathology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Darmawi
- Laboratory of Microbiology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia.,Laboratory of Research, Faculty of Veterinary Medicine, Syiah Kuala University, Darussalam, Banda Aceh 23111, Indonesia
| | - Cut Dahlia Iskandar
- Laboratory of Histology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia
| | - Muhammad Nur Salim
- Laboratory of Pathology, Faculty of Veterinary Medicine, Syiah Kuala University, Banda Aceh 23111, Indonesia.,Study Program of Mathematics and Applied Sciences, Syiah Kuala University, Banda Aceh 23111, Indonesia
| |
Collapse
|
48
|
Lopes L, Setia O, Aurshina A, Liu S, Hu H, Isaji T, Liu H, Wang T, Ono S, Guo X, Yatsula B, Guo J, Gu Y, Navarro T, Dardik A. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res Ther 2018; 9:188. [PMID: 29996912 PMCID: PMC6042254 DOI: 10.1186/s13287-018-0938-6] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Diabetic foot ulcer (DFU) is a severe complication of diabetes, preceding most diabetes-related amputations. DFUs require over US$9 billion for yearly treatment and are now a global public health issue. DFU occurs in the setting of ischemia, infection, neuropathy, and metabolic disorders that result in poor wound healing and poor treatment options. Recently, stem cell therapy has emerged as a new interventional strategy to treat DFU and appears to be safe and effective in both preclinical and clinical trials. However, variability in the stem cell type and origin, route and protocol for administration, and concomitant use of angioplasty confound easy interpretation and generalization of the results. METHODS The PubMed, Google Scholar, and EMBASE databases were searched and 89 preclinical and clinical studies were selected for analysis. RESULTS There was divergence between preclinical and clinical studies regarding stem cell type, origin, and delivery techniques. There was heterogeneous preclinical and clinical study design and few randomized clinical trials. Granulocyte-colony stimulating factor was employed in some studies but with differing protocols. Concomitant performance of angioplasty with stem cell therapy showed increased efficiency compared to either therapy alone. CONCLUSIONS Stem cell therapy is an effective treatment for diabetic foot ulcers and is currently used as an alternative to amputation for some patients without other options for revascularization. Concordance between preclinical and clinical studies may help design future randomized clinical trials.
Collapse
Affiliation(s)
- Lara Lopes
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
- Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ocean Setia
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Afsha Aurshina
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Shirley Liu
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Haidi Hu
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Haiyang Liu
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Tun Wang
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Shun Ono
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Xiangjiang Guo
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
| | - Jianming Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tulio Navarro
- Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery , Yale School of Medicine, Yale University, New Haven, CT USA
- VA Connecticut Healthcare System, West Haven, CT USA
| |
Collapse
|
49
|
Zhang W, Bai X, Zhao B, Li Y, Zhang Y, Li Z, Wang X, Luo L, Han F, Zhang J, Han S, Cai W, Su L, Tao K, Shi J, Hu D. Cell-free therapy based on adipose tissue stem cell-derived exosomes promotes wound healing via the PI3K/Akt signaling pathway. Exp Cell Res 2018; 370:333-342. [PMID: 29964051 DOI: 10.1016/j.yexcr.2018.06.035] [Citation(s) in RCA: 252] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Adipose tissue-derived stem cells (ADSCs) have been shown to enhance wound healing via their paracrine function. Exosomes, as one of the most important paracrine factors, play an essential role in this process. However, the concrete mechanisms that underlie this effect are poorly understood. In this study, we aim to explore the potential roles and molecular mechanisms of exosomes derived from ADSCs in cutaneous wound healing. METHODS Normal human skin fibroblasts and ADSCs were isolated from patient skin and adipose tissues. ADSCs were characterized by using flow cytometric analysis and adipogenic and osteogenic differentiation assays. Exosomes were purified from human ADSCs by differential ultracentrifugation and identified by electron microscopy, nanoparticle tracking, fluorescence confocal microscopy and western blotting. Fibroblasts were treated with different concentrations of exosomes, and the synthesis of collagen was analyzed by western blotting; the levels of growth factors were analyzed by real-time quantitative PCR (RT-PCR) and ELISA; and the proliferation and migration abilities of fibroblasts were analyzed by real-time cell analysis, CCK-8 assays and scratch assays. A mouse model with a full-thickness incision wound was used to evaluate the effect of ADSC-derived exosomes on wound healing. The level of p-Akt/Akt was analyzed by western blotting. Ly294002, a phosphatidylinositol 3-kinases (PI3K) inhibitor, was used to identify the underlying mechanisms by which ADSC-derived exosomes promote wound healing. RESULTS ADSC-derived exosomes were taken up by the fibroblasts, which showed significant, dose-dependent increases in cell proliferation and migration compared to the behavior of cells without exosome treatment. More importantly, both the mRNA and protein levels of type I collagen (Col 1), type III collagen (Col 3), MMP1, bFGF, and TGF-β1 were increased in fibroblasts after stimulation with exosomes. Furthermore, exosomes significantly accelerated wound healing in vivo and increased the level of p-Akt/Akt in vitro. However, Ly294002 alleviated these exosome-induced changes, suggesting that exosomes from ADSCs could promote and optimize collagen deposition in vitro and in vivo and further promote wound healing via the PI3K/Akt signaling pathway. CONCLUSIONS This study demonstrates that ADSC-derived exosomes can promote fibroblast proliferation and migration and optimize collagen deposition via the PI3K/Akt signaling pathway to further accelerate wound healing. Our results suggest that ADSCs likely facilitate wound healing via the release of exosomes, and the PI3K/Akt pathway may play a role in this process. Our data also suggest that the clinical application of ADSC-derived exosomes may shed new light on the use of cell-free therapy to accelerate full-thickness skin wound healing and attenuate scar formation.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China; Department of Plastics and Aesthetic Surgery, The First Affiliated Hospital of Xi'an Medical University, No. 48 West Fenghao Road, Xi'an 710077, Shaanxi, China
| | - Xiaozhi Bai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Bin Zhao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Yan Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Yijie Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Zhenzhen Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Xujie Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Fu Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Julei Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Shichao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Weixia Cai
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Linlin Su
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Ke Tao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Jihong Shi
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
50
|
Cabral J, Ryan AE, Griffin MD, Ritter T. Extracellular vesicles as modulators of wound healing. Adv Drug Deliv Rev 2018; 129:394-406. [PMID: 29408181 DOI: 10.1016/j.addr.2018.01.018] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 01/17/2018] [Accepted: 01/29/2018] [Indexed: 02/07/2023]
Abstract
Impaired healing of cutaneous wounds and ulcers continues to have a major impact on the quality of life of millions of people. In recent years, the capacity for stem and progenitor cells to promote wound repair has been investigated with evidence that secreted factors are responsible for the observed therapeutic benefits. This review addresses current evidence in support of stem/progenitor cell-derived extracellular vesicles (EVs) as a regenerative therapy for acceleration of wound healing. Encouraging results for local or systemic administration of EVs have been reported in a range of clinically-relevant animal models of cutaneous wounds. Furthermore, a number of plausible mechanisms involving EV-mediated transfer of proteins and RNAs that trigger pro-repair pathways in target cells have been demonstrated experimentally. However, for successful clinical translation in the coming years, further emphasis on standardized experimental protocols, detailed methodological reporting and clear definition of EV-based therapeutic products will be required.
Collapse
Affiliation(s)
- Joana Cabral
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland.
| | - Aideen E Ryan
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; Discipline of Pharmacology and Therapeutics, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland.
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland.
| | - Thomas Ritter
- Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine, Nursing and Health Sciences, National University of Ireland, Galway, Ireland; CÚRAM Centre for Research in Medical Devices, National University of Ireland, Galway, Ireland.
| |
Collapse
|