1
|
Gillani SMH, Mughal A, Khan RAA, Nawaz MH, Razzaq Z, Ismat MS, Hussain R, Wadood A, Ahmed S, Minhas B, Abbas M, Vayalpurayil T, Rehman MAU. Development of hybrid polyvinylpyrrolidone/carboxymethyl cellulose/collagen incorporated oregano scaffolds via direct ink write printing for potential wound healing applications. Int J Biol Macromol 2024; 278:134528. [PMID: 39111499 DOI: 10.1016/j.ijbiomac.2024.134528] [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: 02/26/2024] [Revised: 07/08/2024] [Accepted: 08/04/2024] [Indexed: 08/20/2024]
Abstract
Additive manufacturing can develop regenerative scaffolds for wound healing. 3D printing offers meticulous porosity, mechanical integrity, cell adhesion and cost-effectiveness. Herein, we prepared ink composed of carboxymethyl cellulose (CMC), polyvinylpyrrolidone (PVP), collagen, and oregano extract for the fabrication of tissue constructs. The blend was optimized to form a homogeneous ink and rheological characterization demonstrated shear thinning behavior. The scaffolds were printed using Direct Ink Write (DIW) at a flow speed of 4 mm3/s and a layer height of 0.18 mm. The fabricated scaffolds demonstrated an ultimate tensile strength (UTS) and toughness of 730 KPa and 2.72 MJ/m3, respectively. Scanning Electron Microscopy (SEM) revealed an average pore size of 300 ± 30 μm. Fourier transform infrared spectroscopy (FTIR) analysis confirmed that all materials were present. The contact angle of the composite scaffold was 68° ± 1°. Moreover, the scaffolds presented 82 % mass loss (degradation) in phosphate buffer saline (PBS) over 14 days. The composite scaffold exhibited inhibition zones of 9 mm and 12 mm against Staphylococcus aureus and Escherichia coli, respectively. The PVP/CMC/collagen/oregano 3D printed scaffolds exhibited excellent biocompatibility with the mesenchymal stem cells and humman dermal fibroblast cells, confirmed by water-soluble tetrazolium - 8 (WST-8) assay (test conducted for 7 days). The enhanced angiogenic potential of said scaffold was assesed by release of vascular endothelial growth factor followed by further validation through in-vivo CAM assay. Thus, confirming suitability for the potential wound healing application.
Collapse
Affiliation(s)
- Syed Muneeb Haider Gillani
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Awab Mughal
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Raja Aqib Akmal Khan
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Muhammad Haseeb Nawaz
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Zohaib Razzaq
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Muhammad Sameet Ismat
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Rabia Hussain
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan
| | - Abdul Wadood
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Sheraz Ahmed
- Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan
| | - Badar Minhas
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan.
| | - Mohamed Abbas
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Thafasalijyas Vayalpurayil
- Central Labs, King Khalid University, AlQura'a, Abha, P.O. Box 960, Saudi Arabia; Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
| | - Muhammad Atiq Ur Rehman
- Center of Excellence in Biomaterials and Tissue Engineering, Materials Science and Engineering Department Government Collage University, 54000 Lahore, Pakistan; Department of Materials Science & Engineering, Institute of Space Technology, 44000 Islamabad, Pakistan.
| |
Collapse
|
2
|
Iqbal MZ, Riaz M, Biedermann T, Klar AS. Breathing new life into tissue engineering: exploring cutting-edge vascularization strategies for skin substitutes. Angiogenesis 2024:10.1007/s10456-024-09928-6. [PMID: 38842751 DOI: 10.1007/s10456-024-09928-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering.
Collapse
Affiliation(s)
- M Zohaib Iqbal
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mahrukh Riaz
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Thomas Biedermann
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Agnes S Klar
- Tissue Biology Research Unit, Department of Surgery, University Children's Hospital Zurich, Wagistrasse 12, CH-8952, Zurich, Switzerland.
- Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Mikimoto D, Mori M, Toyoda A, Yo K, Oda H, Takeuchi S. Culture insert device with perfusable microchannels enhances in vitroskin model development and barrier function assessment. Biofabrication 2024; 16:035006. [PMID: 38569494 DOI: 10.1088/1758-5090/ad3a15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 04/03/2024] [Indexed: 04/05/2024]
Abstract
The ever-stricter regulations on animal experiments in the field of cosmetic testing have prompted a surge in skin-related research with a special focus on recapitulation of thein vivoskin structurein vitro. In vitrohuman skin models are seen as an important tool for skin research, which in recent years attracted a lot of attention and effort, with researchers moving from the simplest 2-layered models (dermis with epidermis) to models that incorporate other vital skin structures such as hypodermis, vascular structures, and skin appendages. In this study, we designed a microfluidic device with a reverse flange-shaped anchor that allows culturing of anin vitroskin model in a conventional 6-well plate and assessing its barrier function without transferring the skin model to another device or using additional contraptions. Perfusion of the skin model through vascular-like channels improved the morphogenesis of the epidermis compared with skin models cultured under static conditions. This also allowed us to assess the percutaneous penetration of the tested caffeine permeation and vascular absorption, which is one of the key metrics for systemic drug exposure evaluation.
Collapse
Affiliation(s)
| | - Masahito Mori
- Research Center for Beauty and Health Care Product Development Department, POLA Chemical Industries, Inc., Kanagawa, Japan
| | - Akemi Toyoda
- Frontier Research Center, POLA Chemical Industries, Inc., Kanagawa, Japan
| | - Kazuyuki Yo
- Frontier Research Center, POLA Chemical Industries, Inc., Kanagawa, Japan
| | | | | |
Collapse
|
4
|
Zhang Z, Xu C, Xu L, Wan J, Cao G, Liu Z, Ji P, Jin Q, Fu Y, Le Y, Ju J, Hou R, Zhang G. Bioprinted dermis with human adipose tissue-derived microvascular fragments promotes wound healing. Biotechnol Bioeng 2024; 121:1407-1421. [PMID: 37876343 DOI: 10.1002/bit.28588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/21/2023] [Accepted: 10/21/2023] [Indexed: 10/26/2023]
Abstract
Tissue-engineered skin is an effective material for treating large skin defects in a clinical setting. However, its use is limited owing to vascular complications. Human adipose tissue-derived microvascular fragments (HaMVFs) are vascularized units that form vascular networks by rapid reassembly. In this study, we designed a vascularized bionic skin tissue using a three-dimensional (3D) bioprinter of HaMVFs and human fibroblasts encapsulated in a hybrid hydrogel composed of GelMA, HAMA, and fibrinogen. Tissues incorporating HaMVFs showed good in vitro vascularization and mechanical properties after UV crosslinking and thrombin exposure. Thus, the tissue could be sutured appropriately to the wound. In vivo, the vascularized 3D bioprinted skin promoted epidermal regeneration, collagen maturation in the dermal tissue, and vascularization of the skin tissue to accelerate wound healing. Overall, vascularized 3D bioprinted skin with HaMVFs is an effective material for treating skin defects and may be clinically applicable to reduce the necrosis rate of skin grafts.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Chi Xu
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Lei Xu
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Jiaming Wan
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
- Department of Orthopaedics, Yangzhou University Medical College, Yangzhou University, Yangzhou, Jiangsu, China
| | - Gaobiao Cao
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Zhe Liu
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Pengxiang Ji
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Qianheng Jin
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Yi Fu
- Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yingying Le
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Shanghai, China
| | - Jihui Ju
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Ruixing Hou
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| | - Guangliang Zhang
- Department of Orthopaedics, Suzhou Medical College of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopaedics, Suzhou Ruihua Orthopaedic Hospital, Suzhou, Jiangsu, China
| |
Collapse
|
5
|
Wei P, Wu L, Xie H, Chen Z, Tan R, Xu Z. Application of a meshed artificial dermal scaffold and negative-pressure wound therapy in the treatment of full-thickness skin defects: a prospective in vivo study. Biomater Sci 2024; 12:1914-1923. [PMID: 38436071 DOI: 10.1039/d3bm01675g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Artificial dermal scaffolds (ADSs) have great value in repairing deep skin defects. However, problems such as unsatisfactory angiogenesis and local dropsy or empyema often occur, resulting in delayed or even failed wound healing. Negative pressure wound therapy (NPWT) is an effective therapy to promote wound healing or shorten wound bed preparation time. Studies on whether it can improve the effects of ADSs have never been interrupted, and no consensus has been reached. In this study, an improved ADS was prepared by mesh technology, physicochemical experiments were conducted, cell adhesion and proliferation were assessed with the meshed ADS, and in vivo experiments were conducted to investigate the effects of meshed ADS or ADS combined with NPWT in repairing full-thickness skin defects. The results showed that the meshed ADS showed through-layer channels arranged in parallel longitudinal and transverse intersections. The cell experiments confirmed the good cytocompatibility. The in vivo experiments showed that there were no differences in the take rate or contraction of grafted skin among all experiment groups. The meshed ADS exhibited good histocompatibility, and there were no differences in tissue inflammation, dermal angiogenesis, or degradation among all groups. In addition, necrosis, dropsy, or empyema of the dermal scaffold were found in all experiment groups except for the meshed ADS + NPWT group, which showed better wound repair results, including fewer scaffold-related complications and satisfactory skin graft survival and wound contraction. In conclusion, this novel meshed ADS, which has a regular through-layer mesh structure and possesses stable physicochemical properties and good biocompatibility, combined with NPWT can ensure adequate subdermal drainage and reduce the risk of scaffold-related complications, thereby improving the quality and efficiency of wound repair, promoting a broader application of biomaterials, and helping physicians and readers implement more effective wound management.
Collapse
Affiliation(s)
- Pei Wei
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Lijiao Wu
- Department of Orthopedic Surgery, Fujian Provincial Hospital South Branch, Fuzhou 350001, China
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou 350001, China
| | - Hongteng Xie
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhaohong Chen
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Rongwei Tan
- GuangDong Engineering Technology Research Center of Implantable Medical Polymer, Shenzhen Lando Biomaterials Co., Ltd., Shenzhen 518107, China
| | - Zhaorong Xu
- Burn and Wound Repair Department, Fujian Medical University Union Hospital, Fuzhou 350001, China.
- Fujian Burn Institute, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Provincial Key Laboratory of Burn and Trauma, Fujian Medical University Union Hospital, Fuzhou 350001, China
| |
Collapse
|
6
|
Oppenheimer FM, Proietti CJ, Ceruti JM, Hagelin K, Leirós GJ, Balañá ME. Dermal papilla cells cultured as spheres improve angiogenesis. Exp Dermatol 2024; 33:e15038. [PMID: 38450780 DOI: 10.1111/exd.15038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/08/2024]
Abstract
Tissue-engineered skin represents a helpful strategy for the treatment of deep skin injuries. Nevertheless, these skin substitutes must promote and encourage proper vascularization for a successful graft take. Previous work showed that dermal papilla cells (DPC) favour an earlier neovascularization process of grafted skin substitute contributing to the rapid maturation of the neovascular network, reducing inflammation and favouring extracellular matrix remodelling in nude mice. Based on these results, we studied the influence of DPC and its culture conditions on the different stages of angiogenesis in in vitro models. Here, we showed that DPC cultured as spheres favour the expression of angiogenic factors such as VEGF, FGF2 and angiogenin compared to their monolayer culture. To study the effects of DPC on the different stages of angiogenesis, an in vitro model has been adapted. DPC cultured as spheres significantly enhanced HUVEC migration and tubule formation, indicating the importance of employing physiological culture systems that provide a closer representation of cell behaviour and interactions occurring in vivo. Overall, these results allow us to speculate that the use of DPC spheres in skin substitutes could promote its grafting, vascularization and vascular network maturation through the secretion of angiogenic factors. This approach has great potential to improve clinical outcomes in regenerative medicine and skin wound repair.
Collapse
Affiliation(s)
- Florencia Maia Oppenheimer
- Instituto de Ciencia y Tecnología Dr. César Milstein, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET - Fundación Pablo Cassará), Ciudad de Buenos Aires, Argentina
| | - Cecilia Jazmín Proietti
- Instituto de Ciencia y Tecnología Dr. César Milstein, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET - Fundación Pablo Cassará), Ciudad de Buenos Aires, Argentina
| | - Julieta María Ceruti
- Instituto de Ciencia y Tecnología Dr. César Milstein, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET - Fundación Pablo Cassará), Ciudad de Buenos Aires, Argentina
| | - Karin Hagelin
- Instituto de Ciencia y Tecnología Dr. César Milstein, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET - Fundación Pablo Cassará), Ciudad de Buenos Aires, Argentina
| | - Gustavo José Leirós
- Instituto de Ciencia y Tecnología Dr. César Milstein, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET - Fundación Pablo Cassará), Ciudad de Buenos Aires, Argentina
- Instituto de Investigación en Medicina y Ciencias de la Salud, Universidad del Salvador, Ciudad de Buenos Aires, Argentina
| | - María Eugenia Balañá
- Instituto de Ciencia y Tecnología Dr. César Milstein, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET - Fundación Pablo Cassará), Ciudad de Buenos Aires, Argentina
| |
Collapse
|
7
|
Li M, Liu Z, Shen Z, Han L, Wang J, Sang S. A heparin-functionalized bioink with sustained delivery of vascular endothelial growth factor for 3D bioprinting of prevascularized dermal constructs. Int J Biol Macromol 2024; 262:130075. [PMID: 38340924 DOI: 10.1016/j.ijbiomac.2024.130075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Skin tissue engineering faces challenges due to the absence of vascular architecture, impeding the development of permanent skin replacements. To address this, a heparin-functionalized 3D-printed bioink (GH/HepMA) was formulated to enable sustained delivery of vascular endothelial growth factor (VEGF), comprising 0.3 % (w/v) hyaluronic acid (HA), 10 % (w/v) gelatin methacrylate (GelMA), and 0.5 % (w/v) heparin methacrylate (HepMA). The bioink was then used to print dermal constructs with angiogenic functions, including fibroblast networks and human umbilical vein endothelial cell (HUVEC) networks. GH/HepMA, with its covalently cross-linked structure, exhibits enhanced mechanical properties and heparin stability, allowing for a 21-day sustained delivery of VEGF. Cytocompatibility experiments showed that the GH/HepMA bioink supported fibroblast proliferation and promoted collagen I production. With VEGF present, the GH/HepMA bioink promoted HUVEC proliferation, migration, as well as the formation of a richer capillary-like network. Furthermore, HA within the GH/HepMA bioink enhanced rheological properties and printability. Additionally, 3D-bioprinted dermal constructs showed significant deposition of collagen I and III and mature stable capillary-like structures along the axial direction. In summary, this study offers a promising approach for constructing biomimetic multicellular skin substitutes with angiogenesis-induced functions.
Collapse
Affiliation(s)
- Meng Li
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Zixian Liu
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Zhizhong Shen
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan 030031, PR China
| | - Lu Han
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan 030031, PR China
| | - Jianming Wang
- General Hospital of TISCO, North Street, Xinghualing District, Taiyuan 030809, PR China
| | - Shengbo Sang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, PR China.
| |
Collapse
|
8
|
Liu Y, Liu X, Guo H, Wang X, Li A, Qiu D, Gu Q. 3D bioprinting bioglass to construct vascularized full-thickness skin substitutes for wound healing. Mater Today Bio 2024; 24:100899. [PMID: 38188644 PMCID: PMC10770530 DOI: 10.1016/j.mtbio.2023.100899] [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: 06/21/2023] [Revised: 08/28/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
Constructing three-dimensional (3D) bioprinted skin tissues that accurately replicate the mechanical properties of native skin and provide adequate oxygen and nutrient support remains a formidable challenge. In this study, we incorporated phosphosilicate calcium bioglasses (PSCs), a type of bioactive glass (BG), into the bioinks used for 3D bioprinting. The resulting bioink exhibited mechanical properties and biocompatibility that closely resembled those of natural skin. Utilizing 3D bioprinting technology, we successfully fabricated full-thickness skin substitutes, which underwent comprehensive evaluation to assess their regenerative potential in treating full-thickness skin injuries in rats. Remarkably, the skin substitutes loaded with PSCs exhibited exceptional angiogenic activity, as evidenced by the upregulation of angiogenesis-related genes in vitro and the observation of enhanced vascularization in wound tissue sections in vivo. These findings conclusively demonstrated the outstanding efficacy of PSCs in promoting angiogenesis and facilitating the repair of full-thickness skin wounds. The insights garnered from this study provide a valuable reference strategy for the development of skin tissue grafts with potent angiogenesis-inducing capabilities.
Collapse
Affiliation(s)
- Yanyan Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- School of Materials Design and Engineering, Beijing Institute of Fashion Technology, Chaoyang District, Beijing, 100029, PR China
| | - Xin Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, PR China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Jilin University, Changchun 130061, PR China
| | - Haitao Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101449, PR China
| | - Xinhuan Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, PR China
| | - Ailing Li
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Haidian District, Beijing, 100190, PR China
| | - Dong Qiu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Haidian District, Beijing, 100190, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101449, PR China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101449, PR China
| |
Collapse
|
9
|
Izumi K, Yortchan W, Aizawa Y, Kobayashi R, Hoshikawa E, Ling Y, Suzuki A. Recent trends and perspectives in reconstruction and regeneration of intra/extra-oral wounds using tissue-engineered oral mucosa equivalents. JAPANESE DENTAL SCIENCE REVIEW 2023; 59:365-374. [PMID: 37954029 PMCID: PMC10632115 DOI: 10.1016/j.jdsr.2023.10.002] [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: 07/25/2023] [Revised: 09/25/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023] Open
Abstract
Many conditions, including cancer, trauma, and congenital anomalies, can damage the oral mucosa. Multiple cultures of oral mucosal cells have been used for biocompatibility tests and oral biology studies. In recent decades, the clinical translation of tissue-engineered products has progressed significantly in developing tangible therapies and inspiring advancements in medical science. However, the reconstruction of an intraoral mucosa defect remains a significant challenge. Despite the drawbacks of donor-site morbidity and limited tissue supply, the use of autologous oral mucosa remains the gold standard for oral mucosa reconstruction and repair. Tissue engineering offers a promising solution for repairing and reconstructing oral mucosa tissues. Cell- and scaffold-based tissue engineering approaches have been employed to treat various soft tissue defects, suggesting the potential clinical use of tissue-engineered oral mucosa (TEOMs). In this review, we first cover the recent trends in the reconstruction and regeneration of extra-/intra-oral wounds using TEOMs. Next, we describe the current status and challenges of TEOMs. Finally, future strategic approaches and potential technologies to support the advancement of TEOMs for clinical use are discussed.
Collapse
Affiliation(s)
- Kenji Izumi
- Division of Biomimetics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Witsanu Yortchan
- Division of Biomimetics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
- Department of Preventive Dentistry, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - Yuka Aizawa
- Division of Biomimetics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Ryota Kobayashi
- Division of Biomimetics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Emi Hoshikawa
- Division of Biomimetics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
- Division of Periodontology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Yiwei Ling
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ayako Suzuki
- Division of Biomimetics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Japan
| |
Collapse
|
10
|
Meßner FC, Metzger W, Marschall JE, Bickelmann C, Menger MD, Laschke MW. Generation of Connective Tissue-Free Microvascular Fragment Isolates from Subcutaneous Fat Tissue of Obese Mice. Tissue Eng Regen Med 2023; 20:1079-1090. [PMID: 37783934 PMCID: PMC10645785 DOI: 10.1007/s13770-023-00571-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/05/2023] [Accepted: 06/27/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Microvascular fragment (MVF) isolates are generated by short-term enzymatic digestion of adipose tissue and contain numerous vessel segments for the vascularization of tissue defects. Recent findings indicate that the functionality of these isolates is determined by the quality of the fat source. Therefore, we compared MVF isolates from subcutaneous adipose tissue of obese and lean mice. METHODS MVF isolates were generated from subcutaneous adipose tissue of donor mice, which received a high fat or control diet for 12 weeks. The isolates were analyzed in vitro and in vivo. RESULTS Feeding of mice with a high fat diet induced obesity with adipocyte hypertrophy, resulting in a significantly lower collagen fraction and microvessel density within the subcutaneous fat depots when compared to lean controls. Accordingly, MVF isolates from obese mice also contained a reduced number of MVF per mL adipose tissue. However, these MVF tended to be longer and, in contrast to MVF from lean mice, were not contaminated with collagen fibers. Hence, they could be freely seeded onto collagen-glycosaminoglycan scaffolds, whereas MVF from lean controls were trapped in between large amounts of collagen fibers that clogged the pores of the scaffolds. In line with these results, scaffolds seeded with MVF isolates from obese mice exhibited a significantly improved in vivo vascularization after implantation into full-thickness skin defects. CONCLUSION Subcutaneous adipose tissue from obese mice facilitates the generation of connective tissue-free MVF isolates. Translated to clinical conditions, these findings suggest that particularly obese patients may benefit from MVF-based vascularization strategies.
Collapse
Affiliation(s)
- Friederike C Meßner
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Wolfgang Metzger
- Department of Trauma, Hand and Reconstructive Surgery, Saarland University, 66421, Homburg, Germany
| | - Julia E Marschall
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Caroline Bickelmann
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Michael D Menger
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical and Experimental Surgery, Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
11
|
Weng T, Yang M, Zhang W, Jin R, Xia S, Zhang M, Wu P, He X, Han C, Zhao X, Wang X. Dual gene-activated dermal scaffolds regulate angiogenesis and wound healing by mediating the coexpression of VEGF and angiopoietin-1. Bioeng Transl Med 2023; 8:e10562. [PMID: 37693053 PMCID: PMC10487340 DOI: 10.1002/btm2.10562] [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: 06/30/2022] [Revised: 05/22/2023] [Accepted: 06/01/2023] [Indexed: 09/12/2023] Open
Abstract
The vascularization of dermal substitutes is a key challenge in efforts to heal deep skin defects. In this study, dual gene-activated dermal scaffolds (DGADSs-1) were fabricated by loading nanocomposite particles of polyethylenimine (PEI)/multiple plasmid DNAs (pDNAs) encoding vascular endothelial growth factor and angiopoietin-1 at a ratio of 1:1. In a similar manner, DGADSs-2 were loaded with a chimeric plasmid encoding both VEGF and Ang-1. In vitro studies showed that both types of DGADSs released PEI/pDNA nanoparticles in a sustained manner; they demonstrated effective transfection ability, leading to upregulated expression of VEGF and Ang-1. Furthermore, both types of DGADSs promoted fibroblast proliferation and blood vessel formation, although DGADSs-1 showed a more obvious promotion effect. A rat full-thickness skin defect model showed that split-thickness skin transplanted using a one-step method could achieve full survival at the 12th day after surgery in both DGADSs-1 and DGADSs-2 groups, and the vascularization time of dermal substitutes was significantly shortened. Compared with the other three groups of scaffolds, the DGADSs-1 group had significantly greater cell infiltration, collagen deposition, neovascularization, and vascular maturation, all of which promoted wound healing. Thus, compared with single-gene-activated dermal scaffolds, DGADSs show greater potential for enhancing angiogenesis. DGADSs with different loading modes also exhibited differences in terms of angiogenesis; the effect of loading two genes (DGADSs-1) was better than the effect of loading a chimeric gene (DGADSs-2). In summary, DGADSs, which continuously upregulate VEGF and Ang-1 expression, offer a new functional tissue-engineered dermal substitute with the ability to activate vascularization.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Min Yang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Wei Zhang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Ronghua Jin
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Sizhan Xia
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Manjia Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhouChina
| | - Pan Wu
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiaojie He
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Chunmao Han
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| | - Xiong Zhao
- Department of Burn and Plastic SurgeryChildren's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical CenterHangzhouChina
| | - Xingang Wang
- Department of Burns & Wound Care CentreSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Severe Trauma and Burns of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
12
|
Wang Z, Xu H, Yang H, Zhang Y, Wang X, Wang P, Xu Z, Lv D, Rong Y, Dong Y, Tang B, Hu Z, Deng W, Zhu J. Single-stage transplantation combined with epidermal stem cells promotes the survival of tissue-engineered skin by inducing early angiogenesis. Stem Cell Res Ther 2023; 14:51. [PMID: 36959609 PMCID: PMC10035248 DOI: 10.1186/s13287-023-03281-z] [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: 08/13/2022] [Accepted: 03/13/2023] [Indexed: 03/25/2023] Open
Abstract
BACKGROUND The composite transplantation of a split-thickness skin graft (STSG) combined with an acellular dermal matrix (ADM) is a promising repair method for full-thickness skin defects. Due to delayed vascularization of the ADM, no currently available engineered skin tissue is able to permanently cover full-thickness skin defects via a single-stage procedure. Epidermal stem cells (EpSCs) have been found to promote angiogenesis in the wound bed. Whether EpSCs can induce early angiogenesis of dermal substitutes and promote the survival of single-stage tissue-engineered skin transplantation needs to be further studied. METHODS In vitro, rat vascular endothelial cells (RVECs) were treated with the supernatant of EpSCs cultured in ADM and stimulated for 48 h. RVECs were analysed by RNA sequencing and tube formation assays. For the in vivo experiment, 75 rats were randomly divided into five groups: ADM, ADM + EpSCs (AE), STSG, ADM + STSG (AS), and ADM + STSG + EpSCs (ASE) groups. The quality of wound healing was estimated by general observation and H&E and Masson staining. The blood perfusion volume was evaluated using the LDPI system, and the expression of vascular markers was determined by immunohistochemistry (IHC). RESULTS The active substances secreted by EpSCs cultured in ADM promoted angiogenesis, as shown by tube formation experiments and RNA-seq. EpSCs promoted epithelialization of the ADM and vascularization of the ADM implant. The ASE group showed significantly increased skin graft survival, reduced skin contraction, and an improved cosmetic appearance compared with the AS group and the STSG control group. CONCLUSIONS In summary, our findings suggest that EpSCs promote the formation of new blood vessels in dermal substitutes and support one-step transplantation of tissue-engineered skin, and thereby provide new ideas for clinical application.
Collapse
Affiliation(s)
- Zhiyong Wang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hailin Xu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hao Yang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yi Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoyan Wang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peng Wang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhongye Xu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dongming Lv
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yanchao Rong
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yunxian Dong
- Department of Plastic Surgery, Guangdong Second Provincial General Hospital, Southern Medical University, Guangzhou, China
| | - Bing Tang
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhicheng Hu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Wuguo Deng
- Collaborative Innovation Center of Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Jiayuan Zhu
- Department of Burn and Wound Repair Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
13
|
Rincon-Benavides MA, Mendonca NC, Cuellar-Gaviria TZ, Salazar-Puerta AI, Ortega-Pineda L, Blackstone BN, Deng B, McComb DW, Gallego-Perez D, Powell HM, Higuita-Castro N. Engineered Vasculogenic Extracellular Vesicles Drive Nonviral Direct Conversions of Human Dermal Fibroblasts into Induced Endothelial Cells and Improve Wound Closure. ADVANCED THERAPEUTICS 2023; 6:2200197. [PMID: 37577183 PMCID: PMC10416766 DOI: 10.1002/adtp.202200197] [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: 08/22/2022] [Indexed: 08/15/2023]
Abstract
Vasculogenic cell therapies have emerged as a powerful tool to increase vascularization and promote tissue repair/regeneration. Current approaches to cell therapies, however, rely mostly on progenitor cells, which pose significant risks (e.g., uncontrolled differentiation, tumorigenesis, and genetic/epigenetic abnormalities). Moreover, reprogramming methodologies used to generate induced endothelial cells (iECs) from induced pluripotent stem cells rely heavily on viral vectors, which pose additional translational limitations. This work describes the development of engineered human extracellular vesicles (EVs) capable of driving reprogramming-based vasculogenic therapies without the need for progenitor cells and/or viral vectors. The EVs were derived from primary human dermal fibroblasts (HDFs), and were engineered to pack transcription factor genes/transcripts of ETV2, FLI1, and FOXC2 (EFF). Our results indicate that in addition of EFF, the engineered EVs were also loaded with transcripts of angiogenic factors (e.g., VEGF-A, VEGF-KDR, FGF2). In vitro and in vivo studies indicate that such EVs effectively transfected HDFs and drove direct conversions towards iECs within 7-14 days. Finally, wound healing studies in mice indicate that engineered EVs lead to improved wound closure and vascularity. Altogether, our results show the potential of engineered human vasculogenic EVs to drive direct reprogramming processes of somatic cells towards iECs, and facilitate tissue repair/regeneration.
Collapse
Affiliation(s)
- Maria A. Rincon-Benavides
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | | | | | | | | | - Britani N. Blackstone
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
| | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH
| | - David W McComb
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH
| | - Daniel Gallego-Perez
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Surgery, The Ohio State University, Columbus, OH
| | - Heather M. Powell
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
| | - Natalia Higuita-Castro
- Biophysics Graduate Program, The Ohio State University, Columbus, OH
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Surgery, The Ohio State University, Columbus, OH
| |
Collapse
|
14
|
Rosellini E, Cascone MG. Microfluidic Fabrication of Natural Polymer-Based Scaffolds for Tissue Engineering Applications: A Review. Biomimetics (Basel) 2023; 8:biomimetics8010074. [PMID: 36810405 PMCID: PMC9944883 DOI: 10.3390/biomimetics8010074] [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: 12/28/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
Natural polymers, thanks to their intrinsic biocompatibility and biomimicry, have been largely investigated as scaffold materials for tissue engineering applications. Traditional scaffold fabrication methods present several limitations, such as the use of organic solvents, the obtainment of a non-homogeneous structure, the variability in pore size and the lack of pore interconnectivity. These drawbacks can be overcome using innovative and more advanced production techniques based on the use of microfluidic platforms. Droplet microfluidics and microfluidic spinning techniques have recently found applications in the field of tissue engineering to produce microparticles and microfibers that can be used as scaffolds or as building blocks for three-dimensional structures. Compared to standard fabrication technologies, microfluidics-based ones offer several advantages, such as the possibility of obtaining particles and fibers with uniform dimensions. Thus, scaffolds with extremely precise geometry, pore distribution, pore interconnectivity and a uniform pores size can be obtained. Microfluidics can also represent a cheaper manufacturing technique. In this review, the microfluidic fabrication of microparticles, microfibers and three-dimensional scaffolds based on natural polymers will be illustrated. An overview of their applications in different tissue engineering fields will also be provided.
Collapse
|
15
|
The LipoDerm Method for Regeneration and Reconstruction in Plastic Surgery: A Technical Experimental Ex Vivo Note. Med Sci (Basel) 2023; 11:medsci11010016. [PMID: 36810483 PMCID: PMC9944053 DOI: 10.3390/medsci11010016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The combination of adipose-derived stem cells (ASCs) and dermal scaffolds has been shown to be an approach with high potential in soft tissue reconstruction. The addition of dermal templates to skin grafts can increase graft survival through angiogenesis, improve regeneration and healing time, and enhance the overall appearance. However, it remains unknown whether the addition of nanofat-containing ASCs to this construct could effectively facilitate the creation of a multi-layer biological regenerative graft, which could possibly be used for soft tissue reconstruction in the future in a single operation. Initially, microfat was harvested using Coleman's technique, then isolated through the strict protocol using Tonnard's technique. Finally, centrifugation, emulsification, and filtration were conducted to seed the filtered nanofat-containing ASCs onto Matriderm for sterile ex vivo cellular enrichment. After seeding, a resazurin-based reagent was added, and the construct was visualized using two-photon microscopy. Within 1 h of incubation, viable ASCs were detected and attached to the top layer of the scaffold. This experimental ex vivo note opens more dimensions and horizons towards the combination of ASCs and collagen-elastin matrices (i.e., dermal scaffolds) as an effective approach in soft tissue regeneration. The proposed multi-layered structure containing nanofat and dermal template (Lipoderm) may be used, in the future, as a biological regenerative graft for wound defect reconstruction and regeneration in a single operation and can also be combined with skin grafts. Such protocols may optimize the skin graft results by creating a multi-layer soft tissue reconstruction template, leading to more optimal regeneration and aesthetic outcomes.
Collapse
|
16
|
Hou T, Du M, Gao X, An M. Human Vascular Endothelial Cells Promote the Secretion of Vascularization Factors and Migration of Human Skin Fibroblasts under Co-Culture and Its Preliminary Application. Int J Mol Sci 2022; 23:ijms232213995. [PMID: 36430476 PMCID: PMC9697737 DOI: 10.3390/ijms232213995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
The good treatment of skin defects has always been a challenge in the medical field, and the emergence of tissue engineering skin provides a new idea for the treatment of injured skin. However, due to the single seed cells, the tissue engineering skin has the problem of slow vascularization at the premonitory site after implantation into the human body. Cell co-culture technology can better simulate the survival and communication environment of cells in the human body. The study of multicellular co-culture hopes to bring a solution to the problem of tissue engineering. In this paper, human skin fibroblasts (HSFs) and human vascular endothelial cells (HVECs) were co-cultured in Transwell. The Cell Counting Kit 8 (CCK8), Transwell migration chamber, immunofluorescence, Western blot (WB), and real time quantitative PCR (RT-qPCR) were used to study the effects of HVECs on cell activity, migration factor (high mobility group protein 1, HMGB1) and vascularization factor (vascular endothelial growth factor A, VEGFA and fibroblast growth factor 2, FGF2) secretion of HSFs after co-cultured with HVECs in the Transwell. The biological behavior of HSFs co-cultured with HVECs was studied. The experimental results are as follows: (1) The results of cck8 showed that HVECS could promote the activity of HSFs. (2) HVECs could significantly promote the migration of HSFs and promote the secretion of HMGB1. (3) HVECs could promote the secretion of VEGFA and FGF2 of HSFs. (4) The HVECs and HSFs were inoculated on tissue engineering scaffolds at the ratio of 1:4 and were co-cultured and detected for 7 days. The results showed that from the third day, the number of HSFs was significantly higher than that of the control group without HVECs.
Collapse
|
17
|
Ma J, Wu C. Bioactive inorganic particles-based biomaterials for skin tissue engineering. EXPLORATION (BEIJING, CHINA) 2022; 2:20210083. [PMID: 37325498 PMCID: PMC10190985 DOI: 10.1002/exp.20210083] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/09/2022] [Indexed: 06/15/2023]
Abstract
The challenge for treatment of severe cutaneous wound poses an urgent clinical need for the development of biomaterials to promote skin regeneration. In the past few decades, introduction of inorganic components into material system has become a promising strategy for improving performances of biomaterials in the process of tissue repair. In this review, we provide a current overview of the development of bioactive inorganic particles-based biomaterials used for skin tissue engineering. We highlight the three stages in the evolution of the bioactive inorganic biomaterials applied to wound management, including single inorganic materials, inorganic/organic composite materials, and inorganic particles-based cell-encapsulated living systems. At every stage, the primary types of bioactive inorganic biomaterials are described, followed by citation of the related representative studies completed in recent years. Then we offer a brief exposition of typical approaches to construct the composite material systems with incorporation of inorganic components for wound healing. Finally, the conclusions and future directions are suggested for the development of novel bioactive inorganic particles-based biomaterials in the field of skin regeneration.
Collapse
Affiliation(s)
- Jingge Ma
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of SciencesShanghaiP. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of SciencesShanghaiP. R. China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of SciencesBeijingP. R. China
| |
Collapse
|
18
|
Nawaz A, Zaman Safi S, Sikandar S, Zeeshan R, Zulfiqar S, Mehmood N, Alobaid HM, Rehman F, Imran M, Tariq M, Ali A, Emran TB, Yar M. Heparin-Loaded Alginate Hydrogels: Characterization and Molecular Mechanisms of Their Angiogenic and Anti-Microbial Potential. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15196683. [PMID: 36234025 PMCID: PMC9573464 DOI: 10.3390/ma15196683] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 05/06/2023]
Abstract
Background: Chronic wounds continue to be a global concern that demands substantial resources from the healthcare system. The process of cutaneous wound healing is complex, involving inflammation, blood clotting, angiogenesis, migration and remodeling. In the present study, commercially available alginate wound dressings were loaded with heparin. The purpose of the study was to enhance the angiogenic potential of alginate wound dressings and analyze the antibacterial activity, biocompatibility and other relevant properties. We also aimed to conduct some molecular and gene expression studies to elaborate on the mechanisms through which heparin induces angiogenesis. Methods: The physical properties of the hydrogels were evaluated by Fourier transform infrared spectroscopy (FTIR). Swelling ability was measured by soaking hydrogels in the Phosphate buffer at 37 °C, and cell studies were conducted to evaluate the cytotoxicity and biocompatibility of hydrogels in NIH3T3 (fibroblasts). Real-time PCR was conducted to check the molecular mechanisms of heparin/alginate-induced angiogenesis. The physical properties of the hydrogels were evaluated by Fourier transform infrared spectroscopy (FTIR). Results: FTIR confirmed the formation of heparin-loaded alginate wound dressing and the compatibility of both heparin and alginate. Among all, 10 µg/mL concentration of heparin showed the best antibacterial activity against E. coli. The swelling was considerably increased up to 1500% within 1 h. Alamar Blue assay revealed no cytotoxic effect on NIH3T3. Heparin showed good anti-microbial properties and inhibited the growth of E. coli in zones with a diameter of 18 mm. The expression analysis suggested that heparin probably exerts its pro-angiogenetic effect through VEGF and cPGE. Conclusions: We report that heparin-loaded alginate dressings are not cytotoxic and offer increased angiogenic and anti-bacterial potential. The angiogenesis is apparently taken through the VEGF pathway.
Collapse
Affiliation(s)
- Ayesha Nawaz
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
- Department of Biology, Lahore Garrison University, Lahore 54810, Pakistan
| | - Sher Zaman Safi
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom 42610, Selangor, Malaysia
- Correspondence:
| | - Shomaila Sikandar
- Department of Biology, Lahore Garrison University, Lahore 54810, Pakistan
| | - Rabia Zeeshan
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Saima Zulfiqar
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Nadia Mehmood
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Hussah M. Alobaid
- Department of Zoology, College of Science, King Saud University, Riyadh 11362, Saudi Arabia
| | - Fozia Rehman
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| | - Muhammad Imran
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Pakistan
| | - Muhammad Tariq
- Department of Medical Laboratory Technology, University College of Duba, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Muhammad Yar
- Interdisciplinary Research Center in Biomedical Materials, COMSATS University Islamabad Lahore Campus, Lahore 54000, Pakistan
| |
Collapse
|
19
|
Gao C, Lu C, Qiao H, Zhang Y, Liu H, Jian Z, Guo Z, Liu Y. Strategies for vascularized skin models in vitro. Biomater Sci 2022; 10:4724-4739. [PMID: 35861381 DOI: 10.1039/d2bm00784c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
As the largest organ of the human body, the skin has a complex multi-layered structure. The composition of the skin includes cells, extracellular matrix (ECM), vascular networks, and other appendages. Because of the shortage of donor sites, skin substitutes are of great significance in the field of skin tissue repair. Moreover, skin models for disease research, drug screening, and cosmetic testing fall far short of the demand. Skin tissue engineering has made remarkable progress in developing skin models over the years. However, there are still several problems to be resolved. One of the crucial aspects is the lack of vascular systems for nutrient transport and waste disposal. Here, we will focus on the discussion and analysis of advanced manufacturing strategies for prevascularized skin, such as a scaffold-based method, cell coating technology, cell sheet engineering, skin-on-a-chip, and three-dimensional (3D) bioprinting. These key challenges, which restrict the prevascularized skin and provide perspectives on future directions will also be highlighted.
Collapse
Affiliation(s)
- Chuang Gao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Chunxiang Lu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Hao Qiao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Yi Zhang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Huazhen Liu
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Zhian Jian
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Zilong Guo
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China.
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China. .,Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| |
Collapse
|
20
|
Mousivand Z, Ayazi H, Abdollahi A, Akbari H, Raoufi M, Sharifikolouei E. Hybrid electrospun scaffold loaded with Argireline acetate and Dexpanthenol for skin regeneration. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2090359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
| | | | | | - Hamid Akbari
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Raoufi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Sharifikolouei
- Department of Applied Science and Technology, Politecnico di Torino, Turin, Italy
| |
Collapse
|
21
|
Nour S, Imani R, Sharifi AM. Angiogenic Effect of a Nanoniosomal Deferoxamine-Loaded Poly(vinyl alcohol)-Egg White Film as a Promising Wound Dressing. ACS Biomater Sci Eng 2022; 8:3485-3497. [PMID: 35786844 DOI: 10.1021/acsbiomaterials.2c00046] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Owing to the noticeable increase in the number of patients with impaired wound healing capabilities, developing bioactive wound dressings with supportive physicomechanical and biological properties for clinical wound management has attracted much more attention nowadays. In this regard, engineered dressings with angiogenesis potential are vital for accelerated tissue regeneration. In the current study, nanoniosomal deferoxamine (DFO)-loaded transparent films of egg white-poly(vinyl alcohol) (PVA/EW/ND) were successfully fabricated at three different PVA/EW ratios (1:0, 1:1, and 1:1.5 wt/wt %) through the thin film hydration and solvent casting methods. The developed films' characterizations were carried out using scanning electron microscopy, Fourier transform infrared spectroscopy analysis, uniaxial tensile strength, water uptake, water vapor transmission rate, in vitro degradation, and drug release. The results demonstrated that the various weight ratios of PVA/EW have a significant effect on the microscopic morphology, equilibrium swelling, degradation, and mechanical properties of the films. The drug release profile exhibited a sustained release of DFO with controlled burst-lag phases resembling the Korsmeyer-Peppas pattern. The cytotoxicity and adhesion analysis using human dermal fibroblasts displays the biocompatibility of the developed PVA/EW/ND films and the formation of cellular colonies on the surface. The in vitro angiogenic capability of the developed films evaluated by the scratch wound assay and microbead-assisted tube formation study showed a significant increase in the rate of migration of human umbilical vein endothelial cells and in the number of tube-like structures. Therefore, the achieved results suggest that the presented PVA/EW/ND film has promising potential for effective wound healing applications.
Collapse
Affiliation(s)
- Shirin Nour
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 15916-34311, Iran
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran 15916-34311, Iran
| | - Ali Mohammad Sharifi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran.,Razi Drug Research Center, Department of Pharmacology, Iran University of Medical Sciences, Tehran 14496-14535, Iran.,Tissue Engineering Group (NOCERAL), Department of Orthopedics Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
22
|
Liu J, Zhou Z, Zhang M, Song F, Feng C, Liu H. Simple and robust 3D bioprinting of full-thickness human skin tissue. Bioengineered 2022; 13:10087-10097. [PMID: 35412953 PMCID: PMC9161989 DOI: 10.1080/21655979.2022.2063651] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Artificial skins have been used as skin substitutes for wound healing in the clinic, and as in vitro models for safety assessment in cosmetic and pharmaceutical industries. The three-dimensional (3D) bioprinting technique provides a promising strategy in the fabrication of artificial skins. Despite the technological advances, many challenges remain to be conquered, such as the complicated preparation conditions for bio-printed skin and the unavailability of stability and robustness of skin bioprinting. Here, we formulated a novel bio-ink composed of gelatin, sodium alginate and fibrinogen. By optimizing the ratio of components in the bio-ink, the design of the 3D model and the printing conditions, a fibroblasts-containing dermal layer construct was firstly fabricated, on the top of which laminin and keratinocytes were sequentially placed. Through air-liquid interface (ALI) culture by virtue of sterile wire mesh, a full-thickness skin tissue was thus prepared. HE and immunofluorescence staining showed that the bio-printed skin was not only morphologically representative of the human skin, but also expressed the specific markers related to epidermal differentiation and stratum corneum formation. The presented easy and robust preparation of full-thickness skin constructs provides a powerful tool for the establishment of artificial skins, holding critical academic significance and application value.
Collapse
Affiliation(s)
- Jing Liu
- School of Biology, Food and Environment, Hefei University, Hefei, China
| | - Zhengtong Zhou
- Department of Vascular Surgery, Xuanwu Hospital and Institute of Vascular Surgery, Capital Medical University, Beijing, China
| | - Min Zhang
- School of Biology, Food and Environment, Hefei University, Hefei, China
| | - Feng Song
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou, Shandong, China
| | - Chong Feng
- School of Biology, Food and Environment, Hefei University, Hefei, China
| | - Haochen Liu
- Department of Cardiovascular Surgery, Xi'an Children's Hospital, Xi'an, China
| |
Collapse
|
23
|
Zheng W, Xie R, Liang X, Liang Q. Fabrication of Biomaterials and Biostructures Based On Microfluidic Manipulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2105867. [PMID: 35072338 DOI: 10.1002/smll.202105867] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/22/2021] [Indexed: 06/14/2023]
Abstract
Biofabrication technologies are of importance for the construction of organ models and functional tissue replacements. Microfluidic manipulation, a promising biofabrication technique with micro-scale resolution, can not only help to realize the fabrication of specific microsized structures but also build biomimetic microenvironments for biofabricated tissues. Therefore, microfluidic manipulation has attracted attention from researchers in the manipulation of particles and cells, biochemical analysis, tissue engineering, disease diagnostics, and drug discovery. Herein, biofabrication based on microfluidic manipulation technology is reviewed. The application of microfluidic manipulation technology in the manufacturing of biomaterials and biostructures with different dimensions and the control of the microenvironment is summarized. Finally, current challenges are discussed and a prospect of microfluidic manipulation technology is given. The authors hope this review can provide an overview of microfluidic manipulation technologies used in biofabrication and thus steer the current efforts in this field.
Collapse
Affiliation(s)
- Wenchen Zheng
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Ruoxiao Xie
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xiaoping Liang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangdong, 510006, China
| | - Qionglin Liang
- Department of Chemistry, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
24
|
Hosseini M, Brown J, Shafiee A. Strategies to Induce Blood Vessel Ingrowth into Skin Grafts and Tissue-Engineered Substitutes. Tissue Eng Part C Methods 2022; 28:113-126. [PMID: 35172639 DOI: 10.1089/ten.tec.2021.0213] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Skin is a multilayer organ consisting of several tissues and appendages residing in a complex niche. Adequate and physiologically regulated vascularization is an absolute requirement for skin homeostasis, regeneration, and wound healing. The lack of vascular networks and ischemia results in delayed wound closure. In addition, vascularization is critical for the prolonged function and survival of skin grafts and tissue-engineered skin substitutes. This study highlights the clinical challenges associated with the limited vascularization in the cutaneous wounds. Then, we highlight the novel approaches for the development of vascular networks in the skin autografts, allografts, and artificial substitutes. Also, the future directions to overcome the existing vascularization complications in skin grafting and synthetic skin substitutes are presented. Statement of Significance Delayed closure of large dermal wounds, such as burn injuries, results from the lack of vascular networks and ischemia. The amount of blood supply in the skin graft is the primary factor determining the quality of the transplanted grafts. The current skin grafts and their fabrication methods lack the appropriate features that contribute to the vascularization and integration of the wound bed and graft and adherence to the skin layers. Therefore, the new generation of skin grafts should consider advanced technologies to induce vascularization and overcome current challenges.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
| | - Jason Brown
- Herston Biofabrication Institute and Metro North Hospital and Health Service, Brisbane, Australia.,Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, Australia
| | - Abbas Shafiee
- Herston Biofabrication Institute and Metro North Hospital and Health Service, Brisbane, Australia.,Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Brisbane, Australia.,UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Australia
| |
Collapse
|
25
|
Aavani F, Biazar E, Kheilnezhad B, Amjad F. 3D Bio-printing For Skin Tissue Regeneration: Hopes and Hurdles. Curr Stem Cell Res Ther 2022; 17:415-439. [DOI: 10.2174/1574888x17666220204144544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/10/2021] [Accepted: 12/03/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
For many years, discovering the appropriate methods for the treatment of skin irritation has been challenging for specialists and researchers. Bio-printing can be extensively applied to address the demand for proper skin substitutes to improve skin damage. Nowadays, to make more effective bio-mimicking of natural skin, many research teams have developed cell-seeded bio-inks for bioprinting of skin substitutes. These loaded cells can be single or co-cultured in these structures. The present review gives a comprehensive overview of the methods, substantial parameters of skin bioprinting, examples of in vitro and in vivo studies, and current advances and challenges for skin tissue engineering.
Collapse
Affiliation(s)
- Farzaneh. Aavani
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Esmaeil Biazar
- Tissue Engineering Group, Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Bahareh Kheilnezhad
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Fatemeh Amjad
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
26
|
Wong J, Murphy M, Wu YF, Murphy R, Frueh FS, Farnebo S. Basic science approaches to common hand surgery problems. J Hand Surg Eur Vol 2022; 47:117-126. [PMID: 34472390 DOI: 10.1177/17531934211042697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The field of hand surgery is constantly evolving to meet challenges of populations with increasing age and higher demands for active living. While our surgical care has improved over the last decades, it seems that future major improvement in outcomes of clinical treatment will come through advances in biologics and the translation of major discoveries in basic science. This article aims to provide an update on where basic science solutions may answer some of the most critical issues in hand surgery, with a focus on augmentation of tissue repair.
Collapse
Affiliation(s)
- Jason Wong
- Blond McIndoe Laboratories, Manchester, UK.,Department of Plastic Surgery, University of Manchester and Manchester University Foundation Trust, Manchester, UK
| | - Matthew Murphy
- Blond McIndoe Laboratories, Manchester, UK.,Department of Plastic Surgery, University of Manchester and Manchester University Foundation Trust, Manchester, UK
| | - Ya Fang Wu
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Ralph Murphy
- Blond McIndoe Laboratories, Manchester, UK.,Department of Plastic Surgery, University of Manchester and Manchester University Foundation Trust, Manchester, UK
| | - Florian S Frueh
- Department of Plastic Surgery and Hand Surgery, University of Zurich, Zurich, Switzerland
| | - Simon Farnebo
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Plastic Surgery, Hand Surgery, and Burns, Linköping University, Linköping, Sweden
| |
Collapse
|
27
|
Weng T, Wang J, Yang M, Zhang W, Wu P, You C, Han C, Wang X. Nanomaterials for the delivery of bioactive factors to enhance angiogenesis of dermal substitutes during wound healing. BURNS & TRAUMA 2022; 10:tkab049. [PMID: 36960274 PMCID: PMC8944711 DOI: 10.1093/burnst/tkab049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/14/2021] [Indexed: 11/14/2022]
Abstract
Dermal substitutes provide a template for dermal regeneration and reconstruction. They constitutes an ideal clinical treatment for deep skin defects. However, rapid vascularization remains as a major hurdle to the development and application of dermal substitutes. Several bioactive factors play an important regulatory role in the process of angiogenesis and an understanding of the mechanism of achieving their effective delivery and sustained function is vital. Nanomaterials have great potential for tissue engineering. Effective delivery of bioactive factors (including growth factors, peptides and nucleic acids) by nanomaterials is of increasing research interest. This paper discusses the process of dermal substitute angiogenesis and the roles of related bioactive factors in this process. The application of nanomaterials for the delivery of bioactive factors to enhance angiogenesis and accelerate wound healing is also reviewed. We focus on new systems and approaches for delivering bioactive factors for enhancing angiogenesis in dermal substitutes.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Jialiang Wang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Min Yang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Wei Zhang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Pan Wu
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Chuangang You
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Chunmao Han
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | | |
Collapse
|
28
|
Peng Y, Li J, Lin H, Tian S, Liu S, Pu F, Zhao L, Ma K, Qing X, Shao Z. Endogenous repair theory enriches construction strategies for orthopaedic biomaterials: a narrative review. BIOMATERIALS TRANSLATIONAL 2021; 2:343-360. [PMID: 35837417 PMCID: PMC9255795 DOI: 10.12336/biomatertransl.2021.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/19/2021] [Indexed: 02/06/2023]
Abstract
The development of tissue engineering has led to new strategies for mitigating clinical problems; however, the design of the tissue engineering materials remains a challenge. The limited sources and inadequate function, potential risk of microbial or pathogen contamination, and high cost of cell expansion impair the efficacy and limit the application of exogenous cells in tissue engineering. However, endogenous cells in native tissues have been reported to be capable of spontaneous repair of the damaged tissue. These cells exhibit remarkable plasticity, and thus can differentiate or be reprogrammed to alter their phenotype and function after stimulation. After a comprehensive review, we found that the plasticity of these cells plays a major role in establishing the cell source in the mechanism involved in tissue regeneration. Tissue engineering materials that focus on assisting and promoting the natural self-repair function of endogenous cells may break through the limitations of exogenous seed cells and further expand the applications of tissue engineering materials in tissue repair. This review discusses the effects of endogenous cells, especially stem cells, on injured tissue repairing, and highlights the potential utilisation of endogenous repair in orthopaedic biomaterial constructions for bone, cartilage, and intervertebral disc regeneration.
Collapse
Affiliation(s)
- Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jinye Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shuo Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Feifei Pu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lei Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaige Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
29
|
Brown SJ, Surti F, Sibbons P, Hook L. Wound healing properties of a fibrin-based dermal replacement scaffold. Biomed Phys Eng Express 2021; 8. [PMID: 34883468 DOI: 10.1088/2057-1976/ac4176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/09/2021] [Indexed: 11/11/2022]
Abstract
When serious cutaneous injury occurs, the innate wound healing process attempts to restore the skin's appearance and function. Wound healing outcome is affected by factors such as contraction, revascularisation, regeneration versus fibrosis and re-epithelialisation and is also strongly influenced by the pattern and extent of damage to the dermal layer. Dermal replacement scaffolds have been designed to substitute for lost tissue, provide a structure to promote dermal regeneration, and aid skin grafting, resulting in a superior healing outcome. In this study the wound healing properties of a novel fibrin-alginate dermal scaffold were assessed in the porcine wound healing model and also compared to two widely used dermal scaffolds and grafting alone. The fibrin-alginate scaffold, unlike the other scaffolds tested, is not used in combination with an overlying skin graft. Fibrin scaffold treated wounds showed increased, sustained superficial blood flow and reduced contraction during early healing while showing comparable wound closure, re-epithelialisation and final wound outcome to other treatments. The increase in early wound vascularisation coupled with a decrease in contraction and no requirement for a skin graft suggest that the fibrin-based scaffold could provide an effective, distinctive treatment option to improve healing outcomes in human patients.
Collapse
Affiliation(s)
- Stuart J Brown
- Centre for Stem Cells and Regenerative Medicine, 28th Floor Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom.,RAFT Institute, 475 Salisbury House, London Wall, London EC2M 5QQ, United Kingdom
| | - Farhana Surti
- The Griffin Institute , Northwick Park and St Mark's Hospital, Y Block, Watford Road, Harrow, Middlesex, HA1 3UJ, United Kingdom
| | - Paul Sibbons
- The Griffin Institute , Northwick Park and St Mark's Hospital, Y Block, Watford Road, Harrow, Middlesex, HA1 3UJ, United Kingdom
| | - Lilian Hook
- Smart Matrix Ltd, 3rd Floor, 207 Regent Street , London W1B 3HH, United Kingdom
| |
Collapse
|
30
|
Cui M, Wiraja C, Zheng M, Singh G, Yong K, Xu C. Recent Progress in Skin‐on‐a‐Chip Platforms. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Mingyue Cui
- School of Chemical and Biomedical Engineering Nanyang Technological University 62 Nanyang Drive Singapore 637459 Singapore
- Continental‐NTU Corporate Lab Nanyang Technological University 50 Nanyang Avenue Singapore 639798 Singapore
| | - Christian Wiraja
- School of Chemical and Biomedical Engineering Nanyang Technological University 62 Nanyang Drive Singapore 637459 Singapore
| | - Mengjia Zheng
- Department of Biomedical Engineering City University of Hong Kong 83 Tat Chee Avenue Kowloon Hong Kong SAR 00000 China
| | - Gurvinder Singh
- School of Biomedical Engineering The University of Sydney Sydney New South Wales 2006 Australia
- The University of Sydney Nano Institute The University of Sydney Sydney New South Wales 2006 Australia
- The Biophotonics and MechanoBioengineering Lab The University of Sydney Sydney New South Wales 2006 Australia
| | - Ken‐Tye Yong
- School of Biomedical Engineering The University of Sydney Sydney New South Wales 2006 Australia
- The University of Sydney Nano Institute The University of Sydney Sydney New South Wales 2006 Australia
- The Biophotonics and MechanoBioengineering Lab The University of Sydney Sydney New South Wales 2006 Australia
| | - Chenjie Xu
- Department of Biomedical Engineering City University of Hong Kong 83 Tat Chee Avenue Kowloon Hong Kong SAR 00000 China
| |
Collapse
|
31
|
Miguel SP, Ribeiro MP, Otero A, Coutinho P. Application of microalgae and microalgal bioactive compounds in skin regeneration. ALGAL RES 2021. [DOI: 10.1016/j.algal.2021.102395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
32
|
Blanco-Elices C, Chato-Astrain J, Oyonarte S, Bermejo-Casares F, España-López A, Fernández-Valadés R, Sánchez-Quevedo MDC, Alaminos M, Martín-Piedra MA, Garzón I. Generation of a novel model of bioengineered human oral mucosa with increased vascularization potential. J Periodontal Res 2021; 56:1116-1131. [PMID: 34510438 PMCID: PMC9293188 DOI: 10.1111/jre.12927] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/29/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022]
Abstract
Objective The aim of this study was to generate novel models of bioartificial human oral mucosa with increased vascularization potential for future use as an advanced therapies medicinal product, by using different vascular and mesenchymal stem cell sources. Background Oral mucosa substitutes could contribute to the clinical treatment of complex diseases affecting the oral cavity. Although several models of artificial oral mucosa have been described, biointegration is a major issue that could be favored by the generation of novel substitutes with increased vascularization potential once grafted in vivo. Methods Three types of mesenchymal stem cells (MSCs) were obtained from adipose tissue, bone marrow, and dental pulp, and their in vitro potential was evaluated by inducing differentiation to the endothelial lineage using conditioning media. Then, 3D models of human artificial oral mucosa were generated using biocompatible fibrin‐agarose biomaterials combined with human oral mucosa fibroblasts and each type of MSC before and after induction to the endothelial lineage, using human umbilical vein endothelial cells (HUVEC) as controls. The vascularization potential of each oral mucosa substitute was assessed in vitro and in vivo in nude mice. Results In vitro induction of MSCs kept in culture was able to increase the expression of VEGF, CD31, and vWF endothelial markers, especially in bone marrow and dental pulp‐MSCs, and numerous proteins with a role in vasculogenesis become overexpressed. Then, in vivo grafting resulted in a significant increase in blood vessels formation at the interface area between the graft and the host tissues, with significantly positive expression of VEGF, CD31, vWF, and CD34 as compared to negative controls, especially when pre‐differentiated MSCs derived from bone marrow and dental pulp were used. In addition, a significantly higher number of cells committed to the endothelial lineage expressing the same endothelial markers were found within the bioartificial tissue. Conclusion Our results suggest that the use of pre‐differentiated MSCs could contribute to a rapid generation of a vascular network that may favor in vivo biointegration of bioengineered human oral mucosa substitutes.
Collapse
Affiliation(s)
- Cristina Blanco-Elices
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.,Doctoral Programme in Biomedicine, University of Granada, Granada, Spain
| | - Jesús Chato-Astrain
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Salvador Oyonarte
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.,Andalusian Network for Transfusional Medicine, Cells and Tissues and Blood and Tissue Bank of Granada, Granada, Spain
| | | | - Antonio España-López
- Craniofacial Malformations and Cleft Lip and Palate Management Unit, University Hospital Virgen de las Nieves, Granada, Spain
| | - Ricardo Fernández-Valadés
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.,Division of Pediatric Surgery, University Hospital Virgen de las Nieves, Granada, Spain
| | - Maria Del Carmen Sánchez-Quevedo
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Miguel Alaminos
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Miguel Angel Martín-Piedra
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Ingrid Garzón
- Department of Histology (Tissue Engineering Group), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| |
Collapse
|
33
|
Gao C, Lu C, Jian Z, Zhang T, Chen Z, Zhu Q, Tai Z, Liu Y. 3D bioprinting for fabricating artificial skin tissue. Colloids Surf B Biointerfaces 2021; 208:112041. [PMID: 34425531 DOI: 10.1016/j.colsurfb.2021.112041] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 01/17/2023]
Abstract
As an organ in direct contact with the external environment, the skin is the first line of defense against external stimuli, so it is the most vulnerable to damage. In addition, there is an increasing demand for artificial skin in the fields of drug testing, disease research and cosmetic testing. Traditional skin tissue engineering has made encouraging progress after years of development. However, due to the complexity of the skin structures, there is still a big gap between existing artificial skin and natural skin in terms of function. Three-dimensional (3D) bioprinting is an advanced biological manufacturing method. It accurately deposits bioinks into pre-designed three-dimensional shapes to create complex biological tissues. This technology aims to print artificial tissues and organs with biological activities and complete physiological functions, thereby alleviating the problem of tissues and organs in short supply. Here, based on the introduction to skin structure and function, we systematically elaborate and analyze skin manufacturing methods, 3D bioprinting biomaterials and strategies, etc. Finally, the challenges and perspectives in 3D bioprinting skin field are summarized.
Collapse
Affiliation(s)
- Chuang Gao
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Chunxiang Lu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Zhian Jian
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China
| | - Tingrui Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China; Shanghai Engineering Research Center for External Chinese Medicine, Shanghai, 200443, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Shanghai Engineering Research Center for External Chinese Medicine, Shanghai, 200443, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Shanghai Engineering Research Center for External Chinese Medicine, Shanghai, 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China; Shanghai Engineering Research Center for External Chinese Medicine, Shanghai, 200443, China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
34
|
Ma J, Qin C, Wu J, Zhang H, Zhuang H, Zhang M, Zhang Z, Ma L, Wang X, Ma B, Chang J, Wu C. 3D Printing of Strontium Silicate Microcylinder-Containing Multicellular Biomaterial Inks for Vascularized Skin Regeneration. Adv Healthc Mater 2021; 10:e2100523. [PMID: 33963672 DOI: 10.1002/adhm.202100523] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/27/2021] [Indexed: 12/12/2022]
Abstract
The reconstruction of dermal blood vessels is essential for skin regeneration process. However, the lack of vascular structure, insufficient angiogenesis induction, and ineffective graft-host anastomosis of the existing skin substitutes are major bottle-necks for permanent skin replacement in tissue engineering. In this study, the uniform strontium silicate (SS) microcylinders are successfully synthesized and integrated into the biomaterial ink to serve as stable cell-induced factors for angiogenesis, and then a functional skin substitute based on a vascularization-induced biomimetic multicellular system is prepared via a "cell-writing" bioprinting technology. With an unprecedented combination of vascularized skin-mimicking structure and vascularization-induced function, the SS-containing multicellular system exhibits outstanding angiogenic activity both in vitro and in vivo. As a result, the bioprinted skin substitutes significantly accelerate the healing of both acute and chronic wounds by promoting the graft-host integration and vascularized skin regeneration in three animal models. Therefore, the study provides a referable strategy to fabricate biomimetic multicellular constructs with angiogenesis-induced function for regeneration of vascularized complex and hierarchical tissues.
Collapse
Affiliation(s)
- Jingge Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Chen Qin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jinfu Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Hongjian Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Hui Zhuang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Meng Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Zhaowenbin Zhang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Lingling Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Xin Wang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Bing Ma
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| |
Collapse
|
35
|
Ramos-Rodriguez DH, MacNeil S, Claeyssens F, Ortega Asencio I. Delivery of Bioactive Compounds to Improve Skin Cell Responses on Microfabricated Electrospun Microenvironments. Bioengineering (Basel) 2021; 8:105. [PMID: 34436108 PMCID: PMC8389211 DOI: 10.3390/bioengineering8080105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 12/05/2022] Open
Abstract
The introduction of microtopographies within biomaterial devices is a promising approach that allows one to replicate to a degree the complex native environment in which human cells reside. Previously, our group showed that by combining electrospun fibers and additive manufacturing it is possible to replicate to an extent the stem cell microenvironment (rete ridges) located between the epidermal and dermal layers. Our group has also explored the use of novel proangiogenic compounds to improve the vascularization of skin constructs. Here, we combine our previous approaches to fabricate innovative polycaprolactone fibrous microtopographical scaffolds loaded with bioactive compounds (2-deoxy-D-ribose, 17β-estradiol, and aloe vera). Metabolic activity assay showed that microstructured scaffolds can be used to deliver bioactive agents and that the chemical relation between the working compound and the electrospinning solution is critical to replicate as much as possible the targeted morphologies. We also reported that human skin cell lines have a dose-dependent response to the bioactive compounds and that their inclusion has the potential to improve cell activity, induce blood vessel formation and alter the expression of relevant epithelial markers (collagen IV and integrin β1). In summary, we have developed fibrous matrixes containing synthetic rete-ridge-like structures that can deliver key bioactive compounds that can enhance skin regeneration and ultimately aid in the development of a complex wound healing device.
Collapse
Affiliation(s)
- David H. Ramos-Rodriguez
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK;
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK; (S.M.); (F.C.)
| | - Sheila MacNeil
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK; (S.M.); (F.C.)
| | - Frederik Claeyssens
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK; (S.M.); (F.C.)
| | - Ilida Ortega Asencio
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK;
| |
Collapse
|
36
|
Kemp Bohan PM, Cooper LE, Fletcher JL, Corkins CJ, Natesan S, Aden JK, Carlsson A, Chan RK. Impact of dermal matrix thickness on split-thickness skin graft survival and wound contraction in a single-stage procedure. Int Wound J 2021; 19:370-379. [PMID: 34240793 PMCID: PMC8762550 DOI: 10.1111/iwj.13637] [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] [Received: 04/21/2021] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 12/21/2022] Open
Abstract
Optimal treatment of full‐thickness skin injuries requires dermal and epidermal replacement. To spare donor dermis, dermal substitutes can be used ahead of split‐thickness skin graft (STSG) application. However, this two‐stage procedure requires an additional general anaesthetic, often prolongs hospitalisation, and increases outpatient services. Although a few case series have described successful single‐stage reconstructions, with application of both STSG and dermal substitute at the index operation, we have little understanding of how the physical characteristics of dermal substitutes affects the success of a single‐stage procedure. Here, we evaluated several dermal substitutes to optimise single‐stage skin replacement in a preclinical porcine model. A porcine full‐thickness excisional wound model was used to evaluate the following dermal substitutes: autologous dermal graft (ADG; thicknesses 0.15‐0.60 mm), Integra (0.4‐0.8 mm), Alloderm (0.9‐1.6 mm), and chitosan‐based hydrogel (0.1‐0.2 mm). After excision, each wound was treated with either a dermal substitute followed by STSG or STSG alone (control). Endpoints included graft take at postoperative days (PODs) 7 and 14, wound closure at POD 28, and wound contracture from POD 28‐120. Graft take was highest in the STSG alone and hydrogel groups at POD 14 (86.9% ± 19.5% and 81.3% ± 12.3%, respectively; P < .001). There were no differences in graft take at POD 7 or in wound closure at POD 28, though highest rates of wound closure were seen in the STSG alone and hydrogel groups (93.6% ± 9.1% and 99.8% ± 0.5%, respectively). ADG‐treated wounds demonstrated the least amount of wound contracture at each time point. Increase dermal substitute thickness was associated with worse percent graft take at PODs 14 and 28 (Spearman ρ of −0.50 and −0.45, respectively; P < .001). In this preclinical single‐stage skin reconstruction model, thinner ADG and hydrogel dermal substitutes outperformed thicker dermal substitutes. Both substitute thickness and composition affect treatment success. Further preclinical and clinical studies to optimise this treatment modality are warranted.
Collapse
Affiliation(s)
| | - Laura E Cooper
- United States Army Institute of Surgical Research, San Antonio, Texas, USA
| | - John L Fletcher
- Department of Surgery, Brooke Army Medical Center, San Antonio, Texas, USA
| | | | | | - James K Aden
- Department of Graduate Medical Education, Brooke Army Medical Center, San Antonio, Texas, USA
| | - Anders Carlsson
- United States Army Institute of Surgical Research, San Antonio, Texas, USA
| | - Rodney K Chan
- Department of Surgery, Brooke Army Medical Center, San Antonio, Texas, USA.,United States Army Institute of Surgical Research, San Antonio, Texas, USA
| |
Collapse
|
37
|
Matzkeit N, Schulz L, Schleusser S, Jensen JO, Stang FH, Mailaender P, Krämer R, Kisch T. Cold atmospheric plasma improves cutaneous microcirculation in standardized acute wounds: Results of a controlled, prospective cohort study. Microvasc Res 2021; 138:104211. [PMID: 34144075 DOI: 10.1016/j.mvr.2021.104211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Given the high prevalence of wounds and their challenging treatment, the research of therapies to improve wound healing is of great clinical interest. In addition, the general consequences of developing chronic wounds constitute a large health economic aspect, which underscores the interest in the development of efficient treatment strategies. Direct cold atmospheric plasma (di_CAP) has been shown to have beneficial effects on microcirculation of human tissue (Kisch et al., 2016a). It also affects microbial settlements, which may have supportive effects on wound healing processes (Balzer et al., 2015). To treat these adequately, in our view, the positive effects on wound healing should be objectified by application on standardized wounds. However, wound healing is a complex process, depending on nutrient and oxygen supply by cutaneous blood circulation. In spite of microcirculation has been shown to improve in healthy skin by CAP, a quantification of the effect in a standardized wound model has never been evaluated (Kisch et al., 2016a). Based on this, we hypothesize that CAP also influences the microcirculation in standardized acute wounds in a prospective cohort study. METHODS Microcirculatory data of 20 healthy subjects (14 males, 6 females; mean age 40.85 ± 15.84 years; BMI 26.83 ± 7.27 kg/m2) were recorded continuously at a standardized acute wound after skin transplantation (donor site) at the thigh. Under standardized conditions, microcirculatory measurements were performed using a combined laser Doppler and photospectrometry system. After baseline measurement, CAP was applied by a dielectric barrier discharge (DBD) plasma device for 90 s to the acute wound area. Immediately after the application, cutaneous microcirculation was assessed for 30 min (min) at the same site. RESULTS After CAP application, tissue oxygen saturation immediately increased by 5% (92,66 ± 4,76% vs. Baseline 88,21 ± 6,52%, p < 0,01) in the first 60 s and remained significantly elevated for 4 min. Capillary blood flow increased by 19.3% within the first minute of CAP therapy (220.14 ± 65.91 AU vs. Baseline 184.52 ± 56.77 AU, p < 0.001). The statistically highly significant increase in blood flow continued over the entire measurement time. A maximum value was shown in the blood flow in the 15th minute (232.15 ± 58.90 AU, p < 0.001) according to CAP application. With regard to the output measurement, it represents a percentage increase of 25.8%. The measurement of post-capillary venous filling pressure at a tissue depth of 6-8 mm was 59.39 ± AU 12.94 at baseline measurement. After application, there were no significant changes. CONCLUSION CAP increases cutaneous tissue oxygen saturation and capillary blood flow at the standardized acute wound healing model. These results support recently published data on wound healing after CAP treatment. However, further studies are needed to determine if this treatment can improve the reduced microcirculation in chronic wounds. Moreover, repetitive application protocols have to be compared with a single session treatment approach.
Collapse
Affiliation(s)
- Nico Matzkeit
- Department of Plastic Surgery, Hand Surgery, Burn Unit, University Hospital of Schleswig-Holstein, Campus Lübeck, University of Lübeck, Lübeck, Germany.
| | - Lysann Schulz
- Division of Interdisciplinary internal ICU, Medical Department I, University Hospital Leipzig, Germany
| | - Sophie Schleusser
- Department of Plastic Surgery, Hand Surgery, Burn Unit, University Hospital of Schleswig-Holstein, Campus Lübeck, University of Lübeck, Lübeck, Germany
| | - Jan-Oluf Jensen
- Department of Plastic Surgery, Hand Surgery, Burn Unit, University Hospital of Schleswig-Holstein, Campus Lübeck, University of Lübeck, Lübeck, Germany
| | - Felix Hagen Stang
- Department of Plastic Surgery, Hand Surgery, Burn Unit, University Hospital of Schleswig-Holstein, Campus Lübeck, University of Lübeck, Lübeck, Germany
| | - Peter Mailaender
- Department of Plastic Surgery, Hand Surgery, Burn Unit, University Hospital of Schleswig-Holstein, Campus Lübeck, University of Lübeck, Lübeck, Germany
| | - Robert Krämer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Klinikum Westfalen, Dortmund, Germany
| | - Tobias Kisch
- Department of Plastic Surgery, Hand Surgery, Burn Unit, University Hospital of Schleswig-Holstein, Campus Lübeck, University of Lübeck, Lübeck, Germany
| |
Collapse
|
38
|
Engineered Skin Substitute Regenerates the Skin with Hair Follicle Formation. Biomedicines 2021; 9:biomedicines9040400. [PMID: 33917746 PMCID: PMC8068101 DOI: 10.3390/biomedicines9040400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
Currently, engineered skin substitutes (ESS) are unable to regenerate cutaneous appendages. Recent studies have shown that skin-derived precursors (SKPs), which are extensively available, have the potential to induce hair follicle neogenesis. Here, we demonstrate that ESS consisting of culture-expanded SKPs and epidermal stem cells (Epi-SCs) reconstitute the skin with hair follicle regeneration after grafting into nude mice. SKPs seeded in a C-GAG matrix proliferated and expressed higher levels of hair induction signature genes—such as Akp2, Sox2, CD133 and Bmp6—compared to dermal fibroblasts. Moreover, when ESS prepared by seeding a mixture of culture-expanded murine SKPs and human adult Epi-SCs into a C-GAG matrix was grafted into full-thickness skin wounds in nude mice, black hairs were generated within 3 weeks. Immunofluorescence analysis showed that the SKPs were localized to the dermal papillae of the newly-formed hair follicle. Our results indicate that SKPs can serve as the hair-inductive cells in ESS to furnish it with hair genesis potential
Collapse
|
39
|
The triad of nanotechnology, cell signalling, and scaffold implantation for the successful repair of damaged organs: An overview on soft-tissue engineering. J Control Release 2021; 332:460-492. [DOI: 10.1016/j.jconrel.2021.02.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 12/11/2022]
|
40
|
Duan W, Jin Y, Cui Y, Xi F, Liu X, Wo F, Wu J. A co-delivery platform for synergistic promotion of angiogenesis based on biodegradable, therapeutic and self-reporting luminescent porous silicon microparticles. Biomaterials 2021; 272:120772. [PMID: 33838529 DOI: 10.1016/j.biomaterials.2021.120772] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 01/21/2023]
Abstract
Insufficient angiogenesis happened in body defects such as ulceration, coronary heart disease, and chronic wounds constitutes a major challenge in tissue regeneration engineering. Owing to the poor bioactivity and maintenance of pro-angiogenic cells and factors during transplantation, new bioactive materials to tackle the barrier are highly desirable. Herein, we demonstrate a co-delivery platform for synergistic promotion of angiogenesis based on biodegradable, therapeutic, and self-reporting luminescent porous silicon (PSi) microparticles. The biodegradable and biocompatible PSi microparticles could quickly release therapeutic Si ions, which is bioactive to promote cell migration, tube formation, and angiogenic gene expression in vitro. To construct a highly efficient angiogenesis treatment platform, vascular endothelial growth factor (VEGF) was electrostatically adsorbed by PSi microparticles for effective drug loading and delivery. The dual therapeutic components (Si ions and VEGF) could release with the dissolution of Si skeleton, accompanying by the decay of photoluminescence (PL) intensity and blue shift of the maximum PL wavelength. Therefore, real-time drug release could be self-reported and assessed with the two-dimensional PL signal. The co-delivery of Si ions and VEGF displayed synergistic effect and highly efficient angiogenesis, which was evidenced by the enhancement of endothelial cell migration and tube formation in vitro with approximately 1.5-5 times higher than control. The blood vessel formation in vivo was also significantly improved as shown by the chick chorioallantoic membrane (CAM) model, in which the total length, size and junctions exhibited 2.1 ± 0.4, 4 ± 0.4, and 3.9 ± 0.3 times in comparison to control, respectively. The PSi and VEGF co-delivery system display great potential in tissue engineering as a biodegradable and self-reporting theranostic platform to promote angiogenesis.
Collapse
Affiliation(s)
- Wei Duan
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Yao Jin
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Yaoxuan Cui
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Fengna Xi
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China; Department of Chemistry, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingyue Liu
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Fangjie Wo
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China
| | - Jianmin Wu
- Institute of Analytical Chemistry, Department of Chemistry, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
41
|
Agarwal T, Kazemi S, Costantini M, Perfeito F, Correia CR, Gaspar V, Montazeri L, De Maria C, Mano JF, Vosough M, Makvandi P, Maiti TK. Oxygen releasing materials: Towards addressing the hypoxia-related issues in tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111896. [PMID: 33641899 DOI: 10.1016/j.msec.2021.111896] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
Manufacturing macroscale cell-laden architectures is one of the biggest challenges faced nowadays in the domain of tissue engineering. Such living constructs, in fact, pose strict requirements for nutrients and oxygen supply that can hardly be addressed through simple diffusion in vitro or without a functional vasculature in vivo. In this context, in the last two decades, a substantial amount of work has been carried out to develop smart materials that could actively provide oxygen-release to contrast local hypoxia in large-size constructs. This review provides an overview of the currently available oxygen-releasing materials and their synthesis and mechanism of action, highlighting their capacities under in vitro tissue cultures and in vivo contexts. Additionally, we also showcase an emerging concept, herein termed as "living materials as releasing systems", which relies on the combination of biomaterials with photosynthetic microorganisms, namely algae, in an "unconventional" attempt to supply the damaged or re-growing tissue with the necessary supply of oxygen. We envision that future advances focusing on tissue microenvironment regulated oxygen-supplying materials would unlock an untapped potential for generating a repertoire of anatomic scale, living constructs with improved cell survival, guided differentiation, and tissue-specific biofunctionality.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Sara Kazemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marco Costantini
- Institute of Physical Chemistry - Polish Academy of Sciences, Warsaw, Poland
| | - Francisca Perfeito
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Clara R Correia
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - Vítor Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Carmelo De Maria
- Research Center "E. Piaggio", Department of Information Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Pooyan Makvandi
- Center for MicroBioRobotics (CMBR), Istituto Italiano di Tecnologia, Pisa, Italy
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
42
|
Rother S, Ruiz-Gómez G, Balamurugan K, Koehler L, Fiebig KM, Galiazzo VD, Hempel U, Moeller S, Schnabelrauch M, Waltenberger J, Pisabarro MT, Scharnweber D, Hintze V. Hyaluronan/Collagen Hydrogels with Sulfated Glycosaminoglycans Maintain VEGF165 Activity and Fine-Tune Endothelial Cell Response. ACS APPLIED BIO MATERIALS 2020; 4:494-506. [DOI: 10.1021/acsabm.0c01001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sandra Rother
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Gloria Ruiz-Gómez
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, Dresden 01307, Germany
| | | | - Linda Koehler
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Karen M. Fiebig
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Vanessa D. Galiazzo
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Ute Hempel
- Institute of Physiological Chemistry, Carl Gustav Carus Faculty of Medicine, TU Dresden, Fiedlerstraße 42, 01307 Dresden, Germany
| | - Stephanie Moeller
- Biomaterials Department, INNOVENT e.V., Prüssingstr. 27B, 07745 Jena, Germany
| | | | - Johannes Waltenberger
- Department of Cardiovascular Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - M. Teresa Pisabarro
- Structural Bioinformatics, BIOTEC TU Dresden, Tatzberg 47-51, Dresden 01307, Germany
| | - Dieter Scharnweber
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| | - Vera Hintze
- Institute of Materials Science, Max Bergmann Center of Biomaterials, TU Dresden, Budapester Str. 27, 01069 Dresden, Germany
| |
Collapse
|
43
|
Morrison KA, Weinreb RH, Dong X, Toyoda Y, Jin JL, Bender R, Mukherjee S, Spector JA. Facilitated self-assembly of a prevascularized dermal/epidermal collagen scaffold. Regen Med 2020; 15:2273-2283. [PMID: 33325258 DOI: 10.2217/rme-2020-0070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Introduction: Resurfacing complex full thickness wounds requires free tissue transfer which creates donor site morbidity. We describe a method to fabricate a skin flap equivalent with a hierarchical microvascular network. Materials & methods: We fabricated a flap of skin-like tissue containing a hierarchical vascular network by sacrificing Pluronic® F127 macrofibers and interwoven microfibers within collagen encapsulating human pericytes and fibroblasts. Channels were seeded with smooth muscle and endothelial cells. Constructs were topically seeded with keratinocytes. Results: After 28 days in culture, multiphoton microscopy revealed a hierarchical interconnected network of macro- and micro-vessels; larger vessels (>100 μm) were lined with a monolayer endothelial neointima and a subendothelial smooth muscle neomedia. Neoangiogenic sprouts formed in the collagen protodermis and pericytes self-assembled around both fabricated vessels and neoangiogenic sprouts. Conclusion: We fabricated a prevascularized scaffold containing a hierarchical 3D network of interconnected macro- and microchannels within a collagen protodermis subjacent to an overlying protoepidermis with the potential for recipient microvascular anastomosis.
Collapse
Affiliation(s)
- Kerry A Morrison
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA.,Plastic Surgery Resident Physician affiliated with the Hansjorg Wyss Department of Plastic Surgery, New York University Langone Medical Center, New York, NY 10016, USA
| | - Ross H Weinreb
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Xue Dong
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Yoshiko Toyoda
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA.,Plastic Surgery Resident Physician affiliated with the Division of Plastic Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julia L Jin
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Ryan Bender
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Sushmita Mukherjee
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 14850, USA
| | - Jason A Spector
- Department of Surgery, Laboratory of Bioregenerative Medicine & Surgery, Division of Plastic Surgery, Weill Cornell Medical College, New York, NY 10021, USA.,Nancy E. & Peter C. Meinig School of Bioengineering, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
44
|
Ahmadi M, Mehdikhani M, Varshosaz J, Farsaei S, Torabi H. Pharmaceutical evaluation of atorvastatin-loaded nanostructured lipid carriers incorporated into the gelatin/hyaluronic acid/polycaprolactone scaffold for the skin tissue engineering. J Biomater Appl 2020; 35:958-977. [PMID: 33148109 DOI: 10.1177/0885328220970760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In this study, gelatin/hyaluronic acid (HA) scaffolds containing different amounts of atorvastatin-loaded nanostructured lipid carriers (NLCs) coated entirely with polycaprolactone (PCL) film were fabricated for skin regeneration. 12 atorvastatin-loaded NLCs formulations were synthesized, and particle size, zeta potential, drug entrapment efficiency (EE), and drug release of the formulations were determined. The optimum freeze-dried atorvastatin-loaded NLCs were added in 3 different weight percentages to the gelatin and HA membranous scaffolds. Thereafter, the membranes were coated entirely by a thin layer of the PCL. They were characterized, and then mechanical properties, in vitro degradation and in vitro drug release were assessed. Moreover, human dermal fibroblasts (HDF) were cultured on the prepared nanocomposite scaffolds in order to investigate the cytotoxicity by the MTT assay after the first day, third day, and fifth day. Results revealed that the most favorable atorvastatin-loaded NLCs had 99.54 nm average particle size, -24.30 mV zeta potential, 97.98% EE, and 75.24% drug release within 237 hrs. Mechanical tests indicated that all the three scaffolds had approximately a 90 MPa elastic modulus which was more than two-fold of tensile modulus of normal human skin. The in vitro degradation test demonstrated that the membranes were degraded up to 98% after 5 days, and the scaffolds drug release efficiency (DRE) was in a range of 75-79% during those 5 days. The MTT assay results confirmed the cytocompatibility of the scaffolds. The scaffold containing 54.1 wt% NCLs was the optimum sample (S3). Scanning Electron Microscopy (SEM) images of the latter one showed the uniform distribution of the NLCs with an average size of 150 nm, and the images of cultured HDF illustrated the good cell attachment. In conclusion, suitable physicochemical and biological properties of the novel gelatin/HA/PCL nanocomposite scaffold containing 54.1 wt% atorvastatin-loaded NLCs (S3) can be a good candidate for skin regeneration.
Collapse
Affiliation(s)
- Mahsa Ahmadi
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Mehdi Mehdikhani
- Department of Biomedical Engineering, Faculty of Engineering, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Shadi Farsaei
- Department of Clinical Pharmacy and Pharmacy Practice, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Islamic Republic of Iran
| | - Hadis Torabi
- University of Isfahan, Isfahan, Islamic Republic of Iran
| |
Collapse
|
45
|
Abstract
Microvasculature functions at the tissue and cell level, regulating local mass exchange of oxygen and nutrient-rich blood. While there has been considerable success in the biofabrication of large- and small-vessel replacements, functional microvasculature has been particularly challenging to engineer due to its size and complexity. Recently, three-dimensional bioprinting has expanded the possibilities of fabricating sophisticated microvascular systems by enabling precise spatiotemporal placement of cells and biomaterials based on computer-aided design. However, there are still significant challenges facing the development of printable biomaterials that promote robust formation and controlled 3D organization of microvascular networks. This review provides a thorough examination and critical evaluation of contemporary biomaterials and their specific roles in bioprinting microvasculature. We first provide an overview of bioprinting methods and techniques that enable the fabrication of microvessels. We then offer an in-depth critical analysis on the use of hydrogel bioinks for printing microvascularized constructs within the framework of current bioprinting modalities. We end with a review of recent applications of bioprinted microvasculature for disease modeling, drug testing, and tissue engineering, and conclude with an outlook on the challenges facing the evolution of biomaterials design for bioprinting microvasculature with physiological complexity.
Collapse
Affiliation(s)
- Ryan W. Barrs
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Jia Jia
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sophia E. Silver
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael Yost
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ying Mei
- Bioengineering Department, Clemson University, Clemson, SC 29634, USA
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
46
|
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: 44] [Impact Index Per Article: 11.0] [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
|
47
|
Sutterby E, Thurgood P, Baratchi S, Khoshmanesh K, Pirogova E. Microfluidic Skin-on-a-Chip Models: Toward Biomimetic Artificial Skin. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002515. [PMID: 33460277 DOI: 10.1002/smll.202002515] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/29/2020] [Indexed: 06/12/2023]
Abstract
The role of skin in the human body is indispensable, serving as a barrier, moderating homeostatic balance, and representing a pronounced endpoint for cosmetics and pharmaceuticals. Despite the extensive achievements of in vitro skin models, they do not recapitulate the complexity of human skin; thus, there remains a dependence on animal models during preclinical drug trials, resulting in expensive drug development with high failure rates. By imparting a fine control over the microenvironment and inducing relevant mechanical cues, skin-on-a-chip (SoC) models have circumvented the limitations of conventional cell studies. Enhanced barrier properties, vascularization, and improved phenotypic differentiation have been achieved by SoC models; however, the successful inclusion of appendages such as hair follicles and sweat glands and pigmentation relevance have yet to be realized. The present Review collates the progress of SoC platforms with a focus on their fabrication and the incorporation of mechanical cues, sensors, and blood vessels.
Collapse
Affiliation(s)
- Emily Sutterby
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
| | - Sara Baratchi
- School of Health and Medical Science, RMIT University, Bundoora, Victoria, 3083, Australia
| | | | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, Victoria, 3001, Australia
| |
Collapse
|
48
|
Shahin H, Elmasry M, Steinvall I, Söberg F, El-Serafi A. Vascularization is the next challenge for skin tissue engineering as a solution for burn management. BURNS & TRAUMA 2020; 8:tkaa022. [PMID: 32766342 PMCID: PMC7396265 DOI: 10.1093/burnst/tkaa022] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/23/2020] [Indexed: 12/19/2022]
Abstract
Skin regeneration represents a promising line of management for patients with skin loss, including burn victims. The current approach of spraying single cells over the defective areas results in variable success rates in different centers. The modern approach is to synthesize a multilayer skin construct that is based on autologous stem cells. One of the main complications with different types of transplants is sloughing due to the absence of proper vascularization. Ensuring proper vascularization will be crucial for the integration of skin constructs with the surrounding tissues. Combination of the right cells with scaffolds of proper physico-chemical properties, vascularization can be markedly enhanced. The material effect, pore size and adsorption of certain proteins, as well as the application of appropriate growth factors, such as vascular endothelial growth factors, can have an additive effect. A selection of the most effective protocols is discussed in this review.
Collapse
Affiliation(s)
- Hady Shahin
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
- Faculty of Biotechnology, MSA University, 26 July Mehwar Road, 125 85, 6th October City. Egypt
| | - Moustafa Elmasry
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| | - Ingrid Steinvall
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| | - Folke Söberg
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| | - Ahmed El-Serafi
- Department of Hand Surgery and Plastic Surgery and Burns, Linköping University Hospital, 581 85, Linköping, Östergötland, Sweden
- The Department of Biomedical and Clinical Sciences, Linköping University, Linköping University Hospital, 581 83, Linköping, Östergötland, Sweden
| |
Collapse
|
49
|
Wright ME, Yu JK, Jain D, Maeda A, Yeh SCA, DaCosta RS, Lin CP, Santerre JP. Engineering functional microvessels in synthetic polyurethane random-pore scaffolds by harnessing perfusion flow. Biomaterials 2020; 256:120183. [PMID: 32622017 DOI: 10.1016/j.biomaterials.2020.120183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/24/2022]
Abstract
Recently reported biomaterial-based approaches toward prevascularizing tissue constructs rely on biologically or structurally complex scaffolds that are complicated to manufacture and sterilize, and challenging to customize for clinical applications. In the current work, a prevascularization method for soft tissue engineering that uses a non-patterned and non-biological scaffold is proposed. Human fibroblasts and HUVECs were seeded on an ionomeric polyurethane-based hydrogel and cultured for 14 days under medium perfusion. A flow rate of 0.05 mL/min resulted in a greater lumen density in the constructs relative to 0.005 and 0.5 mL/min, indicating the critical importance of flow magnitude in establishing microvessels. Constructs generated at 0.05 mL/min perfusion flow were implanted in a mouse subcutaneous model and intravital imaging was used to characterize host blood perfusion through the construct after 2 weeks. Engineered microvessels were functional (i.e. perfused with host blood and non-leaky) and neovascularization of the construct by host vessels was enhanced relative to non-prevascularized constructs. We report on the first strategy toward engineering functional microvessels in a tissue construct using non-bioactive, non-patterned synthetic polyurethane materials.
Collapse
Affiliation(s)
- Meghan Ee Wright
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Jonathan K Yu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Devika Jain
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Azusa Maeda
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Shu-Chi A Yeh
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph S DaCosta
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
50
|
Han G, Xia X, Pan Z, Lin Y, Li L, Jiao Y, Zhou C, Ding S. Different influence of sulfated chitosan with different sulfonic acid group sites on HUVECs behaviors. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 31:1237-1253. [PMID: 32493148 DOI: 10.1080/09205063.2019.1702764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The vascularization within the scaffold is still a significant challenge in tissue engineering applications. Sulfated chitosan (SCS) as an amazing substance have been used in tissue engineering to stimulate angiogenesis. However, it is not clear whether they have difference in the ability to promote vascularization of SCS with different sulfonic acid group sites. The aim of this study was to evaluate human umbilical vein endothelial cells (HUVECs) viability and differentiation in vitro, affected by three types of sulfated chitosan' i.e. 2-N-6-O-sulfated chitosan (2,6-SCS), 3'6-O-sulfated chitosan (3,6-SCS) and 6-O-sulfated chitosan (6-SCS). The results are showed that all the SCS possesses excellent biological properties to promote HUVECs viability and proliferation. Especially, 2,6-SCS promotes desirable intracellular nitric oxide secretion and capillary tube formation. Meanwhile, 2,6-SCS up-regulate the related gene and protein expression compared with other sulfonic acid group sites SCS and heparin. Therefore, 2,6-SCS is a promising substitute material for angiogenesis and as aqueous formulation can be employed to fabrication functionalization scaffold surface with promoted angiogenesis.
Collapse
Affiliation(s)
- Guijuan Han
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| | - Xiaohui Xia
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| | - Zhicheng Pan
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| | - Yucheng Lin
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| | - Lihua Li
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| | - Yanpeng Jiao
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| | - Changren Zhou
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| | - Shan Ding
- Department of Materials Science and Engineering, Jinan University, Guangzhou, P. R. China.,Engineering Research Centre of Artificial Organs & Materials, Jinan University, Guangzhou, P. R. China
| |
Collapse
|