1
|
Wang T, Liu C, Li Y, Zhang L, Cheng Z. Preparation of Temperature-Responsive Films Based on PNVCL Microgel with Varying Sizes and Cross-Linking Degrees for Cell Harvesting. Macromol Rapid Commun 2025; 46:e2400156. [PMID: 38683686 DOI: 10.1002/marc.202400156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/25/2024] [Indexed: 05/02/2024]
Abstract
This work reports preparing thermal responsive poly (N-isovinylcaprolactam) (PNVCL) microgel based films for cell growth and detachment. PNVCL microgels of hydrated size ranging from 386 to 815 nm (25 °C) and different crosslinking degree are prepared. The PNVCL microgels can be rapidly and massively deposited on glass by spin coating method. Atomic force microscopy (AFM) and water contact angle (WCA) are used to study the influence of crosslinking degree and particle size on the surface morphology, stability, and hydrophilicity of PNVCL microgel film. The cell activity of the desorbed cells is quantitatively characterized employing human normal lung epithelial cells (BEAS-2B). The results show that BEAS-2B cells can be desorbed quickly from the film in 30 min, and the optical density (OD) value of desorbed cells incubated after 3 d increases by approximately 52% compared to the control group. This study broadens the selection of temperature-sensitive film for cell harvesting, and provides a new tool for the quantitative characterization of desorbed cells.
Collapse
Affiliation(s)
- Tao Wang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Chang Liu
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yu Li
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Lifen Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhenping Cheng
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| |
Collapse
|
2
|
Shidei H, Takagi R, Mitsuboshi S, Sekine H, Shimizu T, Kanzaki M. Allogeneic fibroblast sheets prevent pulmonary air leaks caused by rat pleural defects without adhesion to the thoracic wall. Regen Ther 2025; 28:394-401. [PMID: 39896444 PMCID: PMC11787424 DOI: 10.1016/j.reth.2025.01.012] [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: 09/28/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 02/04/2025] Open
Abstract
Introduction Pulmonary air leak (PAL) is one of a complication of thoracic surgery and an unavoidable problem after lung resection or pleural adhesion detachment. Conventional procedures to close PAL by applying polyglycolic acid (PGA) sheets are prone to pleural adhesion. This study evaluated the ability of allogeneic fibroblast sheet transplantation to prevent PALs. Methods Rat skin fibroblasts were prepared from transgenic rats expressing green fluorescent protein (GFP) and cultured on temperature-responsive culture dishes to harvest fibroblast sheets. Allogeneic fibroblast sheets or PGA sheets were transplanted onto the pleural defects of the left lung in F344/NJcl-rnu/rnu (athymic rat), Slc:SD (SD), and BN/SsNSlc (BN) rats to assess PAL and adhesion prevention. Histological and immunological analyses were conducted to evaluate lung tissue of PALs transplanted with fibroblast or PGA sheets. Results Fibroblast sheets and PGA sheets closed pleural defects with PALs in all rat models. Adhesions were observed in most rat models transplanted with PGA sheets, but no adhesions were observed in rat models transplanted with fibroblast sheets. Immunostaining for HBME-1 indicated the regeneration of pleura by fibroblast sheet transplantation on the defects without adhesions after 2 weeks and 3 months of transplantation. Conclusions Similar to autologous fibroblast sheet transplantation, the transplantation of allogeneic fibroblast sheets prevented PALs and closed pleural defects without adhesion between the visceral and parietal pleura. Therefore, it can be concluded that allogeneic fibroblast sheets can be used as a ready-to-use sealant for preventing postoperative PALs.
Collapse
Affiliation(s)
- Hiroaki Shidei
- The Department of Thoracic Surgery, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Ryo Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Shota Mitsuboshi
- The Department of Thoracic Surgery, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Hidekazu Sekine
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Masato Kanzaki
- The Department of Thoracic Surgery, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
3
|
Wang J, Jian K, Yang Q, Gu C, Sheng J, Zhou Y, Yin H, Zhang Z, Hua K, Zhang C. Retarding human adipose-derived MSCs senescence and promoting tendon repair using cell sheet engineering with a histone methyltransferase inhibitor. Sci Rep 2025; 15:6198. [PMID: 39979391 PMCID: PMC11842574 DOI: 10.1038/s41598-025-89234-3] [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: 10/29/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Mesenchymal stem cell (MSC) holds immense potential as candidates for cell therapy in the treatment of tendon injuries due to their remarkable ability for multiple cell differentiation. However, the proliferative and differentiation capacity of MSCs has been limited by cellular senescence during the process of expanding culture. Therefore, in this study, our aim was to maintain the beneficial properties of MSCs. We found that SETD7, a histone methyltransferase, was upregulated during ex vivo expansion of human adipose-derived mesenchymal stem cells (hAD-MSCs). Pharmacological inhibition of SETD7 with PFI-2 in hAD-MSCs cultures delayed their senescence, as evident by the diminished expression of senescent-associated genes and the maintenance of their proliferation and differentiation capacity. Upon transplantation, cell sheets derived from hAD-MSCs expanded with PFI-2 were better able to accelerate tendon repair. Therefore, the present findings reveal that SETD7 is an important target to improve the expansion of hAD-MSCs by delaying senescence, which is importance for the development of efficient stem cell-based therapeutic approaches.
Collapse
Affiliation(s)
- Junjuan Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Ke Jian
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Qing Yang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China
| | - Chunyi Gu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Jiajun Sheng
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Yan Zhou
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Hantian Yin
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Zhihan Zhang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 310000, China
| | - Kouzhen Hua
- Department of Anatomy, School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311300, China.
| | - Can Zhang
- Department of Biomedical Engineering, College of Biology, Hunan University, Changsha, 410082, China.
| |
Collapse
|
4
|
Samiei M, Harmsen MC, Abdolahinia ED, Barar J, Petridis X. Scaffold-Free Strategies in Dental Pulp/Dentine Tissue Engineering: Current Status and Implications for Regenerative Biological Processes. Bioengineering (Basel) 2025; 12:198. [PMID: 40001717 PMCID: PMC11851408 DOI: 10.3390/bioengineering12020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Conventionally, root canal treatment is performed when the dental pulp is severely damaged or lost due to dental trauma or bacterial endodontic infections. This treatment involves removing the compromised or infected pulp tissue, disinfecting the root canal system, and sealing it with inert, non-degradable materials. However, contemporary endodontic treatment has shifted from merely obturating the root canal system with inert materials to guiding endodontic tissue regeneration through biological approaches. The ultimate goal of regenerative endodontics is to restore dental pulp tissue with structural organization and functional characteristics akin to the native pulp, leveraging advancements in tissue engineering and biomaterial sciences. Dental pulp tissue engineering commonly employs scaffold-based strategies, utilizing biomaterials as initial platforms for cell and growth factor delivery, which subsequently act as scaffolds for cell proliferation, differentiation and maturation. However, cells possess an intrinsic capacity for self-organization into spheroids and can generate their own extracellular matrix, eliminating the need for external scaffolds. This self-assembling property presents a promising alternative for scaffold-free dental pulp engineering, addressing limitations associated with biomaterial-based approaches. This review provides a comprehensive overview of cell-based, self-assembling and scaffold-free approaches in dental pulp tissue engineering, highlighting their potential advantages and challenges in advancing regenerative endodontics.
Collapse
Affiliation(s)
- Mohammad Samiei
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.S.); (M.C.H.)
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Martin Conrad Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.S.); (M.C.H.)
| | - Elaheh Dalir Abdolahinia
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Xenos Petridis
- Department of Endodontics, Section of Dental Pathology & Therapeutics, School of Dentistry, National and Kapodistrian University of Athens, 115 27 Athens, Greece
- Department of Endodontology, Section of Fundamental Dentistry, Center for Dentistry and Oral Hygiene, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
5
|
Jang Y, Lee D, Oh J. Fast Autograft Generation Using Transferable 3D Keratinocyte Cell Sheet on PEDOT:PSS Composite PDMS Membrane for Enhancing Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406529. [PMID: 39588867 DOI: 10.1002/smll.202406529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/20/2024] [Indexed: 11/27/2024]
Abstract
The application of cell sheet technology for wound healing preserves dense cell tissue and the natural extracellular matrix (ECM), contributing to disease prevention. Despite the effectiveness of autologous and allograft cell sheets for wound healing, conventional cell sheets, although stable, may experience necrosis in their middle layers due to a lack of nutrients or oxygen. To address these issues, a novel approach is proposed to create cell sheets using mechanical and electrical stimulation. This method not only facilitates the transfer of cell sheets but also enhances cell bioactivity. The performance of the proposed membrane, with a mechanically controlled microstructure under electrical stimulation, is validated in both in vitro and in vivo models. The micro-structured membrane allows for diverse electrical stimulation compared to flat membranes, which accelerates the detachment of cell sheets and promotes angiogenesis and re-epithelialization. These findings indicate that the innovative cell sheet technology could significantly enhance rapid wound healing in regenerative medicine.
Collapse
Affiliation(s)
- Yeongseok Jang
- Department of Mechanical Design Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Dongwon Lee
- Department of Polymer Nano Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| | - Jonghyun Oh
- Department of Nano-Bio Mechanical System Engineering, Jeonbuk National University, Jeonju-si, Jeollabuk-do, 54896, Republic of Korea
| |
Collapse
|
6
|
Mancuso S, Bhalerao A, Cucullo L. Advances and Challenges of Bioassembly Strategies in Neurovascular In Vitro Modeling: An Overview of Current Technologies with a Focus on Three-Dimensional Bioprinting. Int J Mol Sci 2024; 25:11000. [PMID: 39456783 PMCID: PMC11506837 DOI: 10.3390/ijms252011000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Bioassembly encompasses various techniques such as bioprinting, microfluidics, organoids, and self-assembly, enabling advances in tissue engineering and regenerative medicine. Advancements in bioassembly technologies have enabled the precise arrangement and integration of various cell types to more closely mimic the complexity functionality of the neurovascular unit (NVU) and that of other biodiverse multicellular tissue structures. In this context, bioprinting offers the ability to deposit cells in a spatially controlled manner, facilitating the construction of interconnected networks. Scaffold-based assembly strategies provide structural support and guidance cues for cell growth, enabling the formation of complex bio-constructs. Self-assembly approaches utilize the inherent properties of cells to drive the spontaneous organization and interaction of neuronal and vascular components. However, recreating the intricate microarchitecture and functional characteristics of a tissue/organ poses additional challenges. Advancements in bioassembly techniques and materials hold great promise for addressing these challenges. The further refinement of bioprinting technologies, such as improved resolution and the incorporation of multiple cell types, can enhance the accuracy and complexity of the biological constructs; however, developing bioinks that support the growth of cells, viability, and functionality while maintaining compatibility with the bioassembly process remains an unmet need in the field, and further advancements in the design of bioactive and biodegradable scaffolds will aid in controlling cell adhesion, differentiation, and vascularization within the engineered tissue. Additionally, integrating advanced imaging and analytical techniques can provide real-time monitoring and characterization of bioassembly, aiding in quality control and optimization. While challenges remain, ongoing research and technological advancements propel the field forward, paving the way for transformative developments in neurovascular research and tissue engineering. This work provides an overview of the advancements, challenges, and future perspectives in bioassembly for fabricating neurovascular constructs with an add-on focus on bioprinting technologies.
Collapse
Affiliation(s)
- Salvatore Mancuso
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA; (S.M.); (A.B.)
| | - Aditya Bhalerao
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA; (S.M.); (A.B.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, 586 Pioneer Dr, 460 O’Dowd Hall, Rochester, MI 48309, USA
| |
Collapse
|
7
|
Yang L, Sun L, Sun Y, Qiu G, Fan X, Sun Q, Lu G. Engineering Thermoresponsive Poly( N-isopropylacrylamide)-Based Films with Enhanced Stability and Reusability for Efficient Bone Marrow Mesenchymal Stem Cell Culture and Harvesting. Molecules 2024; 29:4481. [PMID: 39339476 PMCID: PMC11435103 DOI: 10.3390/molecules29184481] [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: 08/12/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Poly(N-isopropylacrylamide) (PNIPAM) offers a promising platform for non-invasive and gentle cell detachment. However, conventional PNIPAM-based substrates often suffer from limitations including limited stability and reduced reusability, which hinder their widespread adoption in biomedical applications. In this study, PNIPAM copolymer films were formed on the surfaces of glass slides or silicon wafers using a two-step film-forming method involving coating and grafting. Subsequently, a comprehensive analysis of the films' surface wettability, topography, and thickness was conducted using a variety of techniques, including contact angle analysis, atomic force microscopy (AFM), and ellipsometric measurements. Bone marrow mesenchymal stem cells (BMMSCs) were then seeded onto PNIPAM copolymer films prepared from different copolymer solution concentrations, ranging from 0.2 to 10 mg·mL-1, to select the optimal culture substrate that allowed for good cell growth at 37 °C and effective cell detachment through temperature reduction. Furthermore, the stability and reusability of the optimal copolymer films were assessed. Finally, AFM and X-ray photoelectron spectroscopy (XPS) were employed to examine the surface morphology and elemental composition of the copolymer films after two rounds of BMMSC adhesion and detachment. The findings revealed that the surface properties and overall characteristics of PNIPAM copolymer films varied significantly with the solution concentration. Based on the selection criteria, the copolymer films derived from 1 mg·mL-1 solution were identified as the optimal culture substrates for BMMSCs. After two rounds of cellular adhesion and detachment, some proteins remained on the film surfaces, acting as a foundation for subsequent cellular re-adhesion and growth, thereby implicitly corroborating the practicability and reusability of the copolymer films. This study not only introduces a stable and efficient platform for stem cell culture and harvesting but also represents a significant advance in the fabrication of smart materials tailored for biomedical applications.
Collapse
Affiliation(s)
- Lei Yang
- School of Petrochemical Engineering, Liaoning Petrochemical University, Fushun 113001, China
| | - Luqiao Sun
- School of Petrochemical Engineering, Liaoning Petrochemical University, Fushun 113001, China
| | - Yuanyuan Sun
- College of Engineering, Shenyang Agricultural University, Shenyang 110866, China
| | - Guangwei Qiu
- School of Petrochemical Engineering, Liaoning Petrochemical University, Fushun 113001, China
| | - Xiaoguang Fan
- College of Engineering, Shenyang Agricultural University, Shenyang 110866, China
| | - Qing Sun
- College of Engineering, Shenyang Agricultural University, Shenyang 110866, China
| | - Guang Lu
- School of Civil Engineering, Liaoning Petrochemical University, Fushun 113001, China
| |
Collapse
|
8
|
Fasciano S, Wheba A, Ddamulira C, Wang S. Recent advances in scaffolding biomaterials for cultivated meat. BIOMATERIALS ADVANCES 2024; 162:213897. [PMID: 38810509 DOI: 10.1016/j.bioadv.2024.213897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/07/2024] [Accepted: 05/15/2024] [Indexed: 05/31/2024]
Abstract
The emergence of cultivated meat provides a sustainable and ethical alternative to traditional animal agriculture, highlighting its increasing importance in the food industry. Biomaterial scaffolds are critical components in cultivated meat production for enabling cell adhesion, proliferation, differentiation, and orientation. While there's extensive research on scaffolding biomaterials, applying them to cultivated meat production poses distinct challenges, with each material offering its own set of advantages and disadvantages. This review summarizes the most recent scaffolding biomaterials used in the last five years for cell-cultured meat, detailing their respective advantages and disadvantages. We suggest future research directions and provide recommendations for scaffolds that support scalable, cost-effective, and safe high-quality meat production. Additionally, we highlight commercial challenges cultivated meat faces, encompassing bioreactor design, cell culture mediums, and regulatory and food safety issues. In summary, this review provides a comprehensive guide and valuable insights for researchers and companies in the field of cultivated meat production.
Collapse
Affiliation(s)
- Samantha Fasciano
- Department of Cellular and Molecular Biology, University of New Haven, West Haven, CT, 06516, USA
| | - Anas Wheba
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, 06516, USA
| | - Christopher Ddamulira
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, 06516, USA
| | - Shue Wang
- Department of Chemistry, Chemical and Biomedical Engineering, University of New Haven, West Haven, CT, 06516, USA.
| |
Collapse
|
9
|
Wang X, Chen Q, Li J, Tian W, Liu Z, Chen T. Recent adavances of functional modules for tooth regeneration. J Mater Chem B 2024; 12:7497-7518. [PMID: 39021127 DOI: 10.1039/d4tb01027b] [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: 07/20/2024]
Abstract
Dental diseases, such as dental caries and periodontal disorders, constitute a major global health challenge, affecting millions worldwide and often resulting in tooth loss. Traditional dental treatments, though beneficial, typically cannot fully restore the natural functions and structures of teeth. This limitation has prompted growing interest in innovative strategies for tooth regeneration methods. Among these, the use of dental stem cells to generate functional tooth modules represents an emerging and promising approach in dental tissue engineering. These modules aim to closely replicate the intricate morphology and essential physiological functions of dental tissues. Recent advancements in regenerative research have not only enhanced the assembly techniques for these modules but also highlighted their therapeutic potential in addressing various dental diseases. In this review, we discuss the latest progress in the construction of functional tooth modules, especially on regenerating dental pulp, periodontal tissue, and tooth roots.
Collapse
Affiliation(s)
- Xuan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Qiuyu Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Jiayi Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Zhi Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Tian Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
10
|
Xu J, Liu X, Liang P, Yuan H, Yang T. In Situ Preparation of Tannic Acid-Modified Poly( N-isopropylacrylamide) Hydrogel Coatings for Boosting Cell Response. Pharmaceutics 2024; 16:538. [PMID: 38675199 PMCID: PMC11054217 DOI: 10.3390/pharmaceutics16040538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
The improvement of the capability of poly(N-isopropylacrylamide) (PNIPAAm) hydrogel coating in cell adhesion and detachment is critical to efficiently prepare cell sheets applied in cellular therapies and tissue engineering. To enhance cell response on the surface, the amine group-modified PNIPAAm (PNIPAAm-APTES) nanohydrogels were synthesized and deposited spontaneously on tannic acid (TA)-modified polyethylene (PE) plates. Subsequently, TA was introduced onto PNIPAAm-APTES nanohydrogels to fabricate coatings composed of TA-modified PNIPAAm-APTES (PNIPAAm-APTES-TA). Characterization techniques, including TEM, SEM, XPS, and UV-Vis spectroscopy, confirmed the effective deposition of hydrogels of PNIPAAm as well as the morphologies, content of chemical bonding-TA, and stability of various coatings. Importantly, the porous hydrogel coatings exhibited superhydrophilicity at 20 °C and thermo-responsive behavior. The fluorescence measurement demonstrated that the coating's stability effectively regulated protein behavior, influencing cell response. Notably, cell response tests revealed that even without precise control over the chain length/thickness of PNIPAAm during synthesis, the coatings enhanced cell adhesion and detachment, facilitating efficient cell culture. This work represented a novel and facile approach to preparing bioactive PNIPAAm for cell culture.
Collapse
Affiliation(s)
- Jufei Xu
- Department of Pharmacy, Air Force Medical Center, PLA, Air Force Medical University, Beijing 100142, China;
- School of Life Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China;
| | - Xiangzhe Liu
- School of Life Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pengpeng Liang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China;
| | - Hailong Yuan
- Department of Pharmacy, Air Force Medical Center, PLA, Air Force Medical University, Beijing 100142, China;
| | - Tianyou Yang
- School of Life Science and Technology, Henan Institute of Science and Technology, Xinxiang 453003, China;
| |
Collapse
|
11
|
Mito K, Lachnish J, Le W, Chan C, Chang YL, Yao J. Scaffold-Free Bone Marrow-Derived Mesenchymal Stem Cell Sheets Enhance Bone Formation in a Weight-Bearing Rat Critical Bone Defect Model. Tissue Eng Part A 2024; 30:107-114. [PMID: 38019087 DOI: 10.1089/ten.tea.2023.0118] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023] Open
Abstract
Researchers have been exploring alternative methods for bone tissue engineering, as current management of critical bone defects may be a significant challenge for both patient and surgeon with conventional surgical treatments associated with several potential complications and drawbacks. Recent studies have shown mesenchymal stem cell sheets may enhance bone regeneration in different animal models. We investigated the efficacy of implanted scaffold-free bone marrow-derived mesenchymal stem cell (BMSC) sheets on bone regeneration of a critical bone defect in a weight-bearing rat model. BMSCs were isolated from the femora of male Sprague-Dawley rats 5-6 weeks of age and cell sheets were produced on temperature-responsive culture dishes. Nine male Sprague-Dawley rats 6-8 weeks of age were utilized. A bilateral femoral critical bone defect was created with a bridge plate serving as internal fixation. One side was randomly selected and BMSC sheets were implanted into the bone defect (BMSC group), with the contralateral side receiving no treatment (control). Rats were anesthetized and radiographs were performed at 2-week intervals. At the 8-week time point, rats were euthanized, femurs harvested, and microcomputed tomography and histological analysis was performed. We found a statistically significant increase in new bone formation and bone volume fraction compared with the control. Histomorphometry analysis revealed a larger percent of newly formed bone and a higher total histological score. Our results suggest that scaffold-free BMSC sheets may be used in the management of large weight-bearing bone defects to complement a different surgical technique or as a standalone approach followed by internal fixation. However, further research is still needed.
Collapse
Affiliation(s)
- Kazuaki Mito
- Robert A. Chase Hand & Upper Limb Center, Department of Orthopaedic Surgery, Stanford University Medical Center, Redwood City, California, USA
| | - Jordan Lachnish
- Robert A. Chase Hand & Upper Limb Center, Department of Orthopaedic Surgery, Stanford University Medical Center, Redwood City, California, USA
| | - Wei Le
- Robert A. Chase Hand & Upper Limb Center, Department of Orthopaedic Surgery, Stanford University Medical Center, Redwood City, California, USA
| | - Calvin Chan
- Robert A. Chase Hand & Upper Limb Center, Department of Orthopaedic Surgery, Stanford University Medical Center, Redwood City, California, USA
| | - Yun-Liang Chang
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Jeffrey Yao
- Robert A. Chase Hand & Upper Limb Center, Department of Orthopaedic Surgery, Stanford University Medical Center, Redwood City, California, USA
| |
Collapse
|
12
|
Li Y, Deng T, Aili D, Chen Y, Zhu W, Liu Q. Cell Sheet Technology: An Emerging Approach for Tendon and Ligament Tissue Engineering. Ann Biomed Eng 2024; 52:141-152. [PMID: 37731091 DOI: 10.1007/s10439-023-03370-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/09/2023] [Indexed: 09/22/2023]
Abstract
Tendon and ligament injuries account for a substantial proportion of disorders in the musculoskeletal system. While non-operative and operative treatment strategies have advanced, the restoration of native tendon and ligament structures after injury is still challenging due to its innate limited regenerative ability. Cell sheet technology is an innovative tool for tissue fabrication and cell transplantation in regenerative medicine. In this review, we first summarize different harvesting procedures and advantages of cell sheet technology, which preserves intact cell-to-cell connections and extracellular matrix. We then describe the recent progress of cell sheet technology from preclinical studies, focusing on the application of stem cell-derived sheets in treating tendon and ligament injuries, as well as highlighting its effects on mitigating inflammation and promoting tendon/graft-bone interface healing. Finally, we discuss several prerequisites for future clinical translation including the selection of appropriate cell source, optimization of preparation process, establishment of suitable animal model, and the fabrication of vascularized complex tissue. We believe this review could potentially provoke new ideas and drive the development of more functional biomimetic tissues using cell sheet technology to meet the needs of clinical patients.
Collapse
Affiliation(s)
- Yexin Li
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ting Deng
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Dilihumaer Aili
- Department of Orthopedic Surgery, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Ürümqi, People's Republic of China
| | - Yang Chen
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Qian Liu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| |
Collapse
|
13
|
Abdolahinia ED, Golestani S, Seif S, Afra N, Aflatoonian K, Jalalian A, Valizadeh N, Abdollahinia ED. A review of the therapeutic potential of dental stem cells as scaffold-free models for tissue engineering application. Tissue Cell 2024; 86:102281. [PMID: 38070384 DOI: 10.1016/j.tice.2023.102281] [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: 08/26/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 01/21/2024]
Abstract
In the realm of regenerative medicine, tissue engineering has introduced innovative approaches to facilitate tissue regeneration. Specifically, in pulp tissue engineering, both scaffold-based and scaffold-free techniques have been applied. Relevant articles were meticulously chosen from PubMed, Scopus, and Google Scholar databases through a comprehensive search spanning from October 2022 to December 2022. Despite the inherent limitations of scaffolding, including inadequate mechanical strength for hard tissues, insufficient vents for vessel penetration, immunogenicity, and suboptimal reproducibility-especially with natural polymeric scaffolds-scaffold-free tissue engineering has garnered significant attention. This methodology employs three-dimensional (3D) cell aggregates such as spheroids and cell sheets with extracellular matrix, facilitating precise regeneration of target tissues. The choice of technique aside, stem cells play a pivotal role in tissue engineering, with dental stem cells emerging as particularly promising resources. Their pluripotent nature, non-invasive extraction process, and unique properties render them highly suitable for scaffold-free tissue engineering. This study delves into the latest advancements in leveraging dental stem cells and scaffold-free techniques for the regeneration of various tissues. This paper offers a comprehensive summary of recent developments in the utilization of dental stem cells and scaffold-free methods for tissue generation. It explores the potential of these approaches to advance tissue engineering and their effectiveness in therapies aimed at tissue regeneration.
Collapse
Affiliation(s)
- Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States.
| | - Shayan Golestani
- Department of Oral and Maxillofacial Surgery, Dental School, Islamic Azad University, Isfahan ( Khorasgan) Branch, Isfahan, Iran
| | - Sepideh Seif
- Faculty of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Afra
- Faculty of Dentistry, Hormozgan University of Medical Sciences, Bandarabbas, Iran
| | - Khotan Aflatoonian
- Department of Restorative Dentistry, Dental School, Shahed University of Medical Sciences, Tehran, Iran
| | - Ali Jalalian
- Faculty of Dentistry, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Nasrin Valizadeh
- Chemistry Department, Sciences Faculty, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Elham Dalir Abdollahinia
- Fellowship of Endocrinology, Endocrinology Department, Tabriz University of Medical Sciences, Iran.
| |
Collapse
|
14
|
Nagase K, Nagaoka M, Nakano Y, Utoh R. bFGF-releasing biodegradable nanoparticles for effectively engrafting transplanted hepatocyte sheet. J Control Release 2024; 366:160-169. [PMID: 38154542 DOI: 10.1016/j.jconrel.2023.12.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/06/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Hepatic tissue engineering has been applied for the treatment of intractable liver diseases, and hepatocyte sheets are promising for this purpose. However, hepatocyte sheets have poor survival after transplantation because of their high metabolic activity. In this study, we aimed to develop basic fibroblast growth factor (bFGF)-releasing nanoparticles to prolong the survival of hepatocyte sheets after transplantation. The nanoparticles were prepared by electrospraying a bFGF-dispersed poly(D,l-lactide-co-glycolide) emulsion. bFGF-loaded PLGA nanoparticles can be developed by optimizing the applied electrospray voltage and the oil:water ratio of the emulsion. The prepared nanoparticles exhibited prompt release at the initial duration and continuous gradual release at the subsequent duration. Hepatocyte sheet engraftment was evaluated by transplanting hepatocyte sheets containing the prepared nanoparticles into rats. The hepatocyte sheets with the prepared nanoparticles exhibited longer survival than those without the bFGF nanoparticles or solution owing to the local and continuous release of bFGF from the nanoparticles and the subsequent enhanced angiogenesis at the transplantation site. These results indicated that the prepared bFGF-releasing nanoparticles can enhance the efficiency of hepatocyte sheet transplantation. The developed bFGF-releasing nanoparticles would be useful for the transplantation of cellular tissue with post-transplantation survival challenges.
Collapse
Affiliation(s)
- Kenichi Nagase
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo 105-8512, Japan.
| | - Marin Nagaoka
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo 105-8512, Japan
| | - Yuto Nakano
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo 105-8512, Japan
| | - Rie Utoh
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
15
|
Huang D, Liang J, Yang J, Yang C, Wang X, Dai T, Steinberg T, Li C, Wang F. Current Status of Tissue Regenerative Engineering for the Treatment of Uterine Infertility. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:558-573. [PMID: 37335062 DOI: 10.1089/ten.teb.2022.0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
With the recent developments in tissue engineering, scientists have attempted to establish seed cells from different sources, create cell sheets through various technologies, implant them on scaffolds with various spatial structures, or load scaffolds with cytokines. These research results are very optimistic, bringing hope to the treatment of patients with uterine infertility. In this article, we reviewed articles related to the treatment of uterine infertility from the aspects of experimental treatment strategy, seed cells, scaffold application, and repair criteria so as to provide a basis for future research.
Collapse
Affiliation(s)
- Di Huang
- Shandong First Medical University, Jinan, China
| | - Junhui Liang
- Departments of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jie Yang
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Chunrun Yang
- Departments of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Ultrasonography, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Tianyu Dai
- Shandong First Medical University, Jinan, China
| | - Thorsten Steinberg
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Changzhong Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Fei Wang
- Departments of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
16
|
Santos LF, Silva AS, Mano JF. Magnetic-Based Strategies for Regenerative Medicine and Tissue Engineering. Adv Healthc Mater 2023; 12:e2300605. [PMID: 37543723 DOI: 10.1002/adhm.202300605] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/03/2023] [Indexed: 08/07/2023]
Abstract
The fabrication of biological substitutes to repair, replace, or enhance tissue- and organ-level functions is a long-sought goal of tissue engineering (TE). However, the clinical translation of TE is hindered by several challenges, including the lack of suitable mechanical, chemical, and biological properties in one biomaterial, and the inability to generate large, vascularized tissues with a complex structure of native tissues. Over the past decade, a new generation of "smart" materials has revolutionized the conventional medical field, transforming TE into a more accurate and sophisticated concept. At the vanguard of scientific development, magnetic nanoparticles (MNPs) have garnered extensive attention owing to their significant potential in various biomedical applications owing to their inherent properties such as biocompatibility and rapid remote response to magnetic fields. Therefore, to develop functional tissue replacements, magnetic force-based TE (Mag-TE) has emerged as an alternative to conventional TE strategies, allowing for the fabrication and real-time monitoring of tissues engineered in vitro. This review addresses the recent studies on the use of MNPs for TE, emphasizing the in vitro, in vivo, and clinical applications. Future perspectives of Mag-TE in the fields of TE and regenerative medicine are also discussed.
Collapse
Affiliation(s)
- Lúcia F Santos
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - Ana S Silva
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, 3810-193, Portugal
| |
Collapse
|
17
|
Jang S, Lee J, Jeong JG, Oh TI, Lee E. Reconstruction of Fibrocartilage with Fibrous Alignment of Type I Collagen in Scaffold-Free Manner. Tissue Eng Part A 2023; 29:529-540. [PMID: 37382424 DOI: 10.1089/ten.tea.2023.0061] [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] [Indexed: 06/30/2023] Open
Abstract
For functional reconstruction of fibrocartilage, it is necessary to reproduce the essential mechanical property exhibited by natural fibrocartilage. The distinctive mechanical property of fibrocartilage is originated from the specific histological features of fibrocartilage composed of highly aligned type I collagen (Col I) and an abundant cartilaginous matrix. While the application of tensile stimulation induces highly aligned Col I, our study reveals that it also exerts an antichondrogenic effect on scaffold-free tissues constructed with meniscal chondrocytes (MCs) and induces downregulation of Sox-9 expression and attenuated glycosaminoglycan production. Modulation of mechanotransduction by blocking nuclear translocation of Yes-associated protein (YAP) ameliorated the antichondrogenic effect in the presence of tensile stimulation. Since MCs subjected to mechanical doses either by surface stiffness or tensile stimulation showed reversibility of YAP status even after a long-term exposure to mechanotransduction, fibrocartilage tissue was constructed by sequentially inducing tissue alignment by tensile stimulation followed by inducing cartilaginous matrix production in a tension-released state. The minimal tensile dose to constitute durable tissue alignment was screened by investigating the alignment of cytoskeleton and Col I after culturing the scaffold-free tissue constructs with various tensile doses (10% static tension for 1, 3, 7, and 10 days) followed by maintaining in a released state for 5 days. Fluorescence-conjugated phalloidin binding and immunofluorescence of Col I indicated that the duration of static tension for more than 7 days resulted in durable tissue alignment for at least 5 days in the tension-released state. The tissues subjected to tensile stimulation for 7 days followed by 14 days in a released state in chondrogenic media resulted in abundant cartilaginous matrix as well as uniaxial anisotropic alignment. Our results show that the optimized tensile dose can facilitate the successful reconstruction of fibrocartilage by modulating the characteristics of matrix production by MCs.
Collapse
Affiliation(s)
- Seoyoung Jang
- Department of Medical Engineering, Graduate School, Kyung Hee University, Seoul, South Korea
- R&D Institute, Akrocell Biosciences, Inc., Seoul, South Korea
| | - Jisoo Lee
- Department of Medical Engineering, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Jin Gil Jeong
- Department of Medical Engineering, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Tong In Oh
- Department of Biomedical Engineering, School of Medicine, Kyung Hee University, Seoul, South Korea
- Impedance Imaging Research Center, Kyung Hee University, Seoul, South Korea
| | - EunAh Lee
- Impedance Imaging Research Center, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
18
|
Xu Y, Zhou A, Chen W, Ning X. Scaffold-Free Multicellular 3D Tissue Constructs Utilizing Bio-orthogonal Click Strategy. NANO LETTERS 2023; 23:8770-8778. [PMID: 37694972 DOI: 10.1021/acs.nanolett.3c02889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Multicellular 3D tissue constructs (MTCs) are important in biomedical research due to their capacity to accurately mimic the structure and variation found in real tissues. This study presents a novel bio-orthogonal engineering strategy (BIEN), a transformative scaffold-free approach, to create advanced MTCs. BIEN harnesses the cellular biosynthetic machinery to incorporate bio-orthogonal azide reporters into cell surface glycoconjugates, followed by a click reaction with multiarm PEG, resulting in rapid assembly of MTCs. The implementation of this cutting-edge strategy culminates in the formation of uniform, heterogeneous spheroids, characterized by a high degree of intercellular junction and pluripotency. Remarkably, MTCs simulate tumor features, ensure cell heterogeneity, and significantly improve the subcutaneous xenograft model after transplantation, thereby bolstering both in vitro and in vivo research models. In conclusion, the utilization of the bio-orthogonal engineering strategy as a scaffold-free method to generate superior MTCs holds promising potential for driving advancements in cancer research.
Collapse
Affiliation(s)
- Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, School of Physics, Nanjing University, Nanjing 210093, People's Republic of China
| | - Weiwei Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, People's Republic of China
| |
Collapse
|
19
|
Song Y, Wang N, Shi H, Zhang D, Wang Q, Guo S, Yang S, Ma J. Biomaterials combined with ADSCs for bone tissue engineering: current advances and applications. Regen Biomater 2023; 10:rbad083. [PMID: 37808955 PMCID: PMC10551240 DOI: 10.1093/rb/rbad083] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
In recent decades, bone tissue engineering, which is supported by scaffold, seed cells and bioactive molecules (BMs), has provided new hope and direction for treating bone defects. In terms of seed cells, compared to bone marrow mesenchymal stem cells, which were widely utilized in previous years, adipose-derived stem cells (ADSCs) are becoming increasingly favored by researchers due to their abundant sources, easy availability and multi-differentiation potentials. However, there is no systematic theoretical basis for selecting appropriate biomaterials loaded with ADSCs. In this review, the regulatory effects of various biomaterials on the behavior of ADSCs are summarized from four perspectives, including biocompatibility, inflammation regulation, angiogenesis and osteogenesis, to illustrate the potential of combining various materials with ADSCs for the treatment of bone defects. In addition, we conclude the influence of additional application of various BMs on the bone repair effect of ADSCs, in order to provide more evidences and support for the selection or preparation of suitable biomaterials and BMs to work with ADSCs. More importantly, the associated clinical case reports and experiments are generalized to provide additional ideas for the clinical transformation and application of bone tissue engineering loaded with ADSCs.
Collapse
Affiliation(s)
- Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ning Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Dan Zhang
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Qiang Wang
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| | - Jia Ma
- School and Hospital of Stomatology, China Medical University, Shenyang 110001, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang 110001, China
| |
Collapse
|
20
|
Wei X, Wang L, Duan C, Chen K, Li X, Guo X, Chen P, Liu H, Fan Y. Cardiac patches made of brown adipose-derived stem cell sheets and conductive electrospun nanofibers restore infarcted heart for ischemic myocardial infarction. Bioact Mater 2023; 27:271-287. [PMID: 37122901 PMCID: PMC10130885 DOI: 10.1016/j.bioactmat.2023.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Cell sheet engineering has been proven to be a promising strategy for cardiac remodeling post-myocardial infarction. However, insufficient mechanical strength and low cell retention lead to limited therapeutic efficiency. The thickness and area of artificial cardiac patches also affect their therapeutic efficiency. Cardiac patches prepared by combining cell sheets with electrospun nanofibers, which can be transplanted and sutured to the surface of the infarcted heart, promise to solve this problem. Here, we fabricated a novel cardiac patch by stacking brown adipose-derived stem cells (BADSCs) sheet layer by layer, and then they were combined with multi-walled carbon nanotubes (CNTs)-containing electrospun polycaprolactone/silk fibroin nanofibers (CPSN). The results demonstrated that BADSCs tended to generate myocardium-like structures seeded on CPSN. Compared with BADSCs suspension-containing electrospun nanofibers, the transplantation of the CPSN-BADSCs sheets (CNBS) cardiac patches exhibited accelerated angiogenesis and decreased inflammation in a rat myocardial infarction model. In addition, the CNBS cardiac patches could regulate macrophage polarization and promote gap junction remodeling, thus restoring cardiac functions. Overall, the hybrid cardiac patches made of electrospun nanofibers and cell sheets provide a novel solution to cardiac remodeling after ischemic myocardial infarction.
Collapse
Affiliation(s)
- Xinbo Wei
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Li Wang
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Cuimi Duan
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Kai Chen
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Xia Li
- Beijing Citident Stomatology Hospital, Beijing, 100032, PR China
| | - Ximin Guo
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Peng Chen
- Department of Ultrasound, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| |
Collapse
|
21
|
Zarei M, Sayedain SS, Askarinya A, Sabbaghi M, Alizadeh R. Improving physio-mechanical and biological properties of 3D-printed PLA scaffolds via in-situ argon cold plasma treatment. Sci Rep 2023; 13:14120. [PMID: 37644122 PMCID: PMC10465552 DOI: 10.1038/s41598-023-41226-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023] Open
Abstract
As a bone tissue engineering material, polylactic acid (PLA) has received significant attention and interest due to its ease of processing and biocompatibility. However, its insufficient mechanical properties and poor wettability are two major drawbacks that limit its extensive use. For this purpose, the present study uses in-situ cold argon plasma treatment coupled with a fused deposition modeling printer to enhance the physio-mechanical and biological behavior of 3D-printed PLA scaffolds. Following plasma treatment, field emission scanning electron microscopy images indicated that the surface of the modified scaffold became rough, and the interlayer bonding was enhanced. This resulted in an improvement in the tensile properties of samples printed in the X, Y, and Z directions, with the enhancement being more significant in the Z direction. Additionally, the root mean square value of PLA scaffolds increased (up to 70-fold) after plasma treatment. X-ray photoelectron spectroscopy analysis demonstrated that the plasma technique increased the intensity of oxygen-containing bonds, thereby reducing the water contact angle from 92.5° to 42.1°. The in-vitro degradation study also demonstrated that argon plasma treatment resulted in a 77% increase in PLA scaffold degradation rate. Furthermore, the modified scaffold improved the viability, attachment, and proliferation of human adipose-derived stem cells. These findings suggest that in-situ argon plasma treatment may be a facile and effective method for improving the properties of 3D-printed parts for bone tissue engineering and other applications.
Collapse
Affiliation(s)
- Masoud Zarei
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Sayed Shahab Sayedain
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Amirhossein Askarinya
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Mobina Sabbaghi
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran
| | - Reza Alizadeh
- Department of Materials Science and Engineering, Sharif University of Technology, Azadi Ave., Tehran, 11155-9466, Iran.
| |
Collapse
|
22
|
Puricelli C, Gigliotti CL, Stoppa I, Sacchetti S, Pantham D, Scomparin A, Rolla R, Pizzimenti S, Dianzani U, Boggio E, Sutti S. Use of Poly Lactic-co-glycolic Acid Nano and Micro Particles in the Delivery of Drugs Modulating Different Phases of Inflammation. Pharmaceutics 2023; 15:1772. [PMID: 37376219 PMCID: PMC10301392 DOI: 10.3390/pharmaceutics15061772] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic inflammation contributes to the pathogenesis of many diseases, including apparently unrelated conditions such as metabolic disorders, cardiovascular diseases, neurodegenerative diseases, osteoporosis, and tumors, but the use of conventional anti-inflammatory drugs to treat these diseases is generally not very effective given their adverse effects. In addition, some alternative anti-inflammatory medications, such as many natural compounds, have scarce solubility and stability, which are associated with low bioavailability. Therefore, encapsulation within nanoparticles (NPs) may represent an effective strategy to enhance the pharmacological properties of these bioactive molecules, and poly lactic-co-glycolic acid (PLGA) NPs have been widely used because of their high biocompatibility and biodegradability and possibility to finely tune erosion time, hydrophilic/hydrophobic nature, and mechanical properties by acting on the polymer's composition and preparation technique. Many studies have been focused on the use of PLGA-NPs to deliver immunosuppressive treatments for autoimmune and allergic diseases or to elicit protective immune responses, such as in vaccination and cancer immunotherapy. By contrast, this review is focused on the use of PLGA NPs in preclinical in vivo models of other diseases in which a key role is played by chronic inflammation or unbalance between the protective and reparative phases of inflammation, with a particular focus on intestinal bowel disease; cardiovascular, neurodegenerative, osteoarticular, and ocular diseases; and wound healing.
Collapse
Affiliation(s)
- Chiara Puricelli
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Casimiro Luca Gigliotti
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Ian Stoppa
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
| | - Sara Sacchetti
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Deepika Pantham
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Torino, 10125 Turin, Italy;
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Roberta Rolla
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Science, University of Turin, Corso Raffaello 30, 10125 Torino, Italy;
| | - Umberto Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
- Maggiore della Carità University Hospital, Corso Mazzini 18, 28100 Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
- NOVAICOS s.r.l.s, Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Salvatore Sutti
- Department of Health Sciences, Università del Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy; (C.P.); (C.L.G.); (I.S.); (S.S.); (D.P.); (R.R.); (E.B.); (S.S.)
| |
Collapse
|
23
|
Wang M, He M, Xu X, Wu Z, Tao J, Yin F, Luo K, Jiang J. Cementum protein 1 gene-modified adipose-derived mesenchymal stem cell sheets enhance periodontal regeneration in osteoporosis rat. J Periodontal Res 2023. [PMID: 37154214 DOI: 10.1111/jre.13133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/04/2023] [Accepted: 04/27/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND AND OBJECTIVES Osteoporosis (OP) and periodontitis are both diseases with excessive bone resorption, and the number of patients who suffer from these diseases is expected to increase. OP has been identified as a risk factor that accelerates the pathological process of periodontitis. Achieving effective and safe periodontal regeneration in OP patients is a meaningful challenge. This study aimed to assess the efficacy and biosecurity of human cementum protein 1 (hCEMP1) gene-modified cell sheets for periodontal fenestration defect regeneration in an OP rat model. MATERIALS AND METHODS Rat adipose-derived mesenchymal stem cells (rADSCs) were isolated from Sprague-Dawley rats. After primary culture, rADSCs were subjected to cell surface analysis and multi-differentiation assay. And rADSCs were transduced with hCEMP1 by lentiviral vector, and hCEMP1 gene-modified cell sheets were generated. The expression of hCEMP1 was evaluated by reverse transcription polymerase chain reaction and immunocytochemistry staining, and transduced cell proliferation was evaluated by Cell Counting Kit-8. The hCEMP1 gene-modified cell sheet structure was detected by histological analysis and scanning electron microscopy. Osteogenic and cementogenic-associated gene expression was evaluated by real-time quantitative polymerase chain reaction. In addition, an OP rat periodontal fenestration defect model was used to evaluate the regeneration effect of hCEMP1 gene-modified rADSC sheets. The efficacy was assessed with microcomputed tomography and histology, and the biosecurity of gene-modified cell sheets was evaluated by histological analysis of the spleen, liver, kidney and lung. RESULTS The rADSCs showed a phenotype of mesenchymal stem cells and possessed multi-differentiation capacity. The gene and protein expression of hCEMP1 through lentiviral transduction was confirmed, and there was no significant effect on rADSC proliferation. Overexpression of hCEMP1 upregulated osteogenic and cementogenic-related genes such as runt-related transcription factor 2, bone morphogenetic protein 2, secreted phosphoprotein 1 and cementum attachment protein in the gene-modified cell sheets. The fenestration lesions in OP rats treated with hCEMP1 gene-modified cell sheets exhibited complete bone bridging, cementum and periodontal ligament formation. Furthermore, histological sections of the spleen, liver, kidney and lung showed no evident pathological damage. CONCLUSION This pilot study demonstrates that hCEMP1 gene-modified rADSC sheets have a marked ability to enhance periodontal regeneration in OP rats. Thus, this approach may represent an effective and safe strategy for periodontal disease patients with OP.
Collapse
Affiliation(s)
- Meijie Wang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jing Tao
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Fan Yin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jun Jiang
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
24
|
Hu D, Li X, Li J, Tong P, Li Z, Lin G, Sun Y, Wang J. The preclinical and clinical progress of cell sheet engineering in regenerative medicine. Stem Cell Res Ther 2023; 14:112. [PMID: 37106373 PMCID: PMC10136407 DOI: 10.1186/s13287-023-03340-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Cell therapy is an accessible method for curing damaged organs or tissues. Yet, this approach is limited by the delivery efficiency of cell suspension injection. Over recent years, biological scaffolds have emerged as carriers of delivering therapeutic cells to the target sites. Although they can be regarded as revolutionary research output and promote the development of tissue engineering, the defect of biological scaffolds in repairing cell-dense tissues is apparent. Cell sheet engineering (CSE) is a novel technique that supports enzyme-free cell detachment in the shape of a sheet-like structure. Compared with the traditional method of enzymatic digestion, products harvested by this technique retain extracellular matrix (ECM) secreted by cells as well as cell-matrix and intercellular junctions established during in vitro culture. Herein, we discussed the current status and recent progress of CSE in basic research and clinical application by reviewing relevant articles that have been published, hoping to provide a reference for the development of CSE in the field of stem cells and regenerative medicine.
Collapse
Affiliation(s)
- Danping Hu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
- HANGZHOU CHEXMED TECHNOLOGY CO., LTD, Hangzhou, 310000, China
| | - Xinyu Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Jie Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Pei Tong
- Hospital of Hunan Guangxiu, Medical College of Hunan Normal University, Hunan Normal University, Changsha, 410008, China
| | - Zhe Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
- National Engineering and Research Center of Human Stem Cells, Changsha, 410008, China
- Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, 410008, China
| | - Yi Sun
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China.
- National Engineering and Research Center of Human Stem Cells, Changsha, 410008, China.
- Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, 410008, China.
| | - Juan Wang
- Shanghai Biomass Pharmaceutical Product Evaluation Professional Public Service Platform, Center for Pharmacological Evaluation and Research, China State Institute of Pharmaceutical Industry, Shanghai, 200437, China.
| |
Collapse
|
25
|
Mi S, Chang Z, Wang X, Gao J, Liu Y, Liu W, He W, Qi Z. Bioactive Spinal Cord Scaffold Releasing Neurotrophic Exosomes to Promote In Situ Centralis Neuroplasticity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:16355-16368. [PMID: 36958016 DOI: 10.1021/acsami.2c19607] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Spinal cord injury (SCI), one of the most serious injuries of the central nervous system, causes physical functional dysfunction and even paralysis in millions of patients. As a matter of necessity, redressing the neuroleptic pathologic microenvironment to a neurotrophic microenvironment is essential in order to alleviate this dilemma and facilitate the recovery of the spinal cord. Herein, based on cell-sheet technology, two functional cell types─uninduced and neural-induced stem cells from human exfoliated deciduous teeth─were formed into a composite membrane that subsequently self-assembled to form a bioactive scaffold with a spinal-cord-like structure, called a spinal cord assembly (SCA). In a stable extracellular matrix microenvironment, SCA continuously released SCA-derived exosomes containing various neurotrophic factors, which effectively promoted neuronal regeneration, axonal extension, and angiogenesis and inhibited glial scar generation in a rat model of SCI. Neurotrophic exosomes significantly improved the pathological microenvironment and promoted in situ centralis neuroplasticity, ultimately eliciting a strong repair effect in this model. SCA therapy is a promising strategy for the effective treatment of SCI based on neurotrophic exosome delivery.
Collapse
Affiliation(s)
- Sisi Mi
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Zhuo Chang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, SVL, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Xue Wang
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Jiaxin Gao
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Yu Liu
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Wenjia Liu
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Wangxiao He
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
- Department of Medical Oncology and Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| |
Collapse
|
26
|
Mi S, Wang X, Gao J, Liu Y, Qi Z. Implantation with SHED sheet induced with homogenate protein of spinal cord promotes functional recovery from spinal cord injury in rats. Front Bioeng Biotechnol 2023; 11:1119639. [PMID: 36998812 PMCID: PMC10043224 DOI: 10.3389/fbioe.2023.1119639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Introduction: After spinal cord injury (SCI) occurs, the lesion is in a growth inhibitory microenvironment that severely hinders neural regeneration. In this microenvironment, inhibitory factors are predominant and factors that promote nerve regeneration are few. Improving neurotrophic factors in the microenvironment is the key to treating SCI.Methods: Based on cell sheet technology, we designed a bioactive material with a spinal cord‐like structure –SHED sheet induced with homogenate protein of spinal cord (hp–SHED sheet). Hp–SHED sheet was implanted into the spinal cord lesion for treating SCI rats with SHED suspensions as a control to investigate the effects on nerve regeneration.Results: Hp–SHED sheet revealed a highly porous three–dimensional inner structure, which facilitates nerve cell attachment and migration. Hp-SHED sheet in vivo restored sensory and motor functions in SCI rats by promoting nerve regeneration, axonal remyelination, and inhibiting glial scarring.Discussion: Hp–SHED sheet maximally mimics the microenvironment of the natural spinal cord and facilitate cell survival and differentiation. Hp–SHED sheet could release more neurotrophins and the sustained action of neurotrophins improves the pathological microenvironment, which effectively promotes nerve regeneration, axonal extension, and inhibits glial scarring, thereby promoting the in situ centralis neuroplasticity. Hp–SHED sheet therapy is a promising strategy for effective treatment of SCI based on neurotrophins delivery.
Collapse
|
27
|
Schulte A, de Los Santos Pereira A, Pola R, Pop-Georgievski O, Jiang S, Romanenko I, Singh M, Sedláková Z, Schönherr H, Poręba R. On-Demand Cell Sheet Release with Low Density Peptide-Functionalized Non-LCST Polymer Brushes. Macromol Biosci 2023; 23:e2200472. [PMID: 36598869 DOI: 10.1002/mabi.202200472] [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: 11/04/2022] [Revised: 12/16/2022] [Indexed: 01/05/2023]
Abstract
Cell sheet harvesting offers a great potential for the development of new therapies for regenerative medicine. For cells to adhere onto surfaces, proliferate, and to be released on demand, thermoresponsive polymeric coatings are generally considered to be required. Herein, an alternative approach for the cell sheet harvesting and rapid release on demand is reported, circumventing the use of thermoresponsive materials. This approach is based on the end-group biofunctionalization of non-thermoresponsive and antifouling poly(2-hydroxyethyl methacrylate) (p(HEMA)) brushes with cell-adhesive peptide motifs. While the nonfunctionalized p(HEMA) surfaces are cell-repellant, ligation of cell-signaling ligand enables extensive attachment and proliferation of NIH 3T3 fibroblasts until the formation of a confluent cell layer. Remarkably, the formed cell sheets can be released from the surfaces by gentle rinsing with cell-culture medium. The release of the cells is found to be facilitated by low surface density of cell-adhesive peptides, as confirmed by X-ray photoelectron spectroscopy. Additionally, the developed system affords possibility for repeated cell seeding, proliferation, and release on previously used substrates without any additional pretreatment steps. This new approach represents an alternative to thermally triggered cell-sheet harvesting platforms, offering possibility of capture and proliferation of various rare cell lines via appropriate selection of the cell-adhesive ligand.
Collapse
Affiliation(s)
- Anna Schulte
- Physical Chemistry I and Research Center of Micro and Nanochemistry and Engineering (Cµ), Department of Chemistry and Biology University of Siegen, Adolf-Reichwein-Str. 2, 57076, Siegen, Germany
| | - Andres de Los Santos Pereira
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Heyrovsky sq. 2, Prague, 162 06, Czech Republic
| | - Robert Pola
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Heyrovsky sq. 2, Prague, 162 06, Czech Republic
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Heyrovsky sq. 2, Prague, 162 06, Czech Republic
| | - Siyu Jiang
- Physical Chemistry I and Research Center of Micro and Nanochemistry and Engineering (Cµ), Department of Chemistry and Biology University of Siegen, Adolf-Reichwein-Str. 2, 57076, Siegen, Germany
| | - Iryna Romanenko
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Heyrovsky sq. 2, Prague, 162 06, Czech Republic
| | - Manisha Singh
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Heyrovsky sq. 2, Prague, 162 06, Czech Republic
| | - Zdeňka Sedláková
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Heyrovsky sq. 2, Prague, 162 06, Czech Republic
| | - Holger Schönherr
- Physical Chemistry I and Research Center of Micro and Nanochemistry and Engineering (Cµ), Department of Chemistry and Biology University of Siegen, Adolf-Reichwein-Str. 2, 57076, Siegen, Germany
| | - Rafał Poręba
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Heyrovsky sq. 2, Prague, 162 06, Czech Republic
| |
Collapse
|
28
|
Lim J, Lee MS, Jeon J, Yang HS. Fibrinogen-based cell and spheroid sheets manipulating and delivery for mouse hindlimb ischemia. Biofabrication 2023; 15. [PMID: 36630715 DOI: 10.1088/1758-5090/acb233] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/11/2023] [Indexed: 01/12/2023]
Abstract
In this research, we introduced a novel strategy for fabricating cell sheets (CSs) prepared by simply adding a fibrinogen solution to growth medium without using any synthetic polymers or chemical agents. We confirmed that the fibrinogen-based CS could be modified for target tissue regardless of size, shape, and cell types. Also, fibrinogen-based CSs were versatile and could be used to form three-dimensional (3D) CSs such as multi-layered CSs and those mimicking native blood vessels. We also prepared fibrinogen-based spheroid sheets for the treatment of ischemic disease. The fibrinogen-based spheroid sheets had much higherin vitrotubule formation and released more angiogenic factors compared to other types of platform in this research. We transplanted fibrinogen-based spheroid sheets into a mouse hindlimb ischemia model and found that fibrinogen-based spheroid sheets showed significantly improved physiological function and blood perfusion rates compared to the other types of platform in this research.
Collapse
Affiliation(s)
- Juhan Lim
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Min Suk Lee
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea.,Medical Laser Research Center, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin Jeon
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Hee Seok Yang
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea.,Bio-Medical Engineering Research Center, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
29
|
Elomaa L, Lindner M, Leben R, Niesner R, Weinhart M. In vitro vascularization of hydrogel-based tissue constructs via a combined approach of cell sheet engineering and dynamic perfusion cell culture. Biofabrication 2023; 15. [DOI: 10.1088/1758-5090/ac9433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/22/2022] [Indexed: 11/11/2022]
Abstract
Abstract
The bioengineering of artificial tissue constructs requires special attention to their fast vascularization to provide cells with sufficient nutrients and oxygen. We addressed the challenge of in vitro vascularization by employing a combined approach of cell sheet engineering, 3D printing, and cellular self-organization in dynamic maturation culture. A confluent cell sheet of human umbilical vein endothelial cells (HUVECs) was detached from a thermoresponsive cell culture substrate and transferred onto a 3D-printed, perfusable tubular scaffold using a custom-made cell sheet rolling device. Under indirect co-culture conditions with human dermal fibroblasts (HDFs), the cell sheet-covered vessel mimic embedded in a collagen gel together with additional singularized HUVECs started sprouting into the surrounding gel, while the suspended cells around the tube self-organized and formed a dense lumen-containing 3D vascular network throughout the gel. The HDFs cultured below the HUVEC-containing cell culture insert provided angiogenic support to the HUVECs via molecular crosstalk without competing for space with the HUVECs or inducing rapid collagen matrix remodeling. The resulting vascular network remained viable under these conditions throughout the 3 week cell culture period. This static indirect co-culture setup was further transferred to dynamic flow conditions, where the medium perfusion was enabled via two independently addressable perfusion circuits equipped with two different cell culture chambers, one hosting the HDFs and the other hosting the HUVEC-laden collagen gel. Using this system, we successfully connected the collagen-embedded HUVEC culture to a dynamic medium flow, and within 1 week of the dynamic cell culture, we detected angiogenic sprouting and dense microvascular network formation via HUVEC self-organization in the hydrogel. Our approach of combining a 3D-printed and cell sheet-covered vascular precursor that retained its sprouting capacity together with the self-assembling HUVECs in a dynamic perfusion culture resulted in a vascular-like 3D network, which is a critical step toward the long-term vascularization of bioengineered in vitro tissue constructs.
Collapse
|
30
|
Guo R, Zhuang H, Chen X, Ben Y, Fan M, Wang Y, Zheng P. Tissue engineering in growth plate cartilage regeneration: Mechanisms to therapeutic strategies. J Tissue Eng 2023; 14:20417314231187956. [PMID: 37483459 PMCID: PMC10359656 DOI: 10.1177/20417314231187956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/29/2023] [Indexed: 07/25/2023] Open
Abstract
The repair of growth plate injuries is a highly complex process that involves precise spatiotemporal regulation of multiple cell types. While significant progress has been made in understanding the pathological mechanisms underlying growth plate injuries, effectively regulating this process to regenerate the injured growth plate cartilage remains a challenge. Tissue engineering technology has emerged as a promising therapeutic approach for achieving tissue regeneration through the use of functional biological materials, seed cells and biological factors, and it is now widely applied to the regeneration of bone and cartilage. However, due to the unique structure and function of growth plate cartilage, distinct strategies are required for effective regeneration. Thus, this review provides an overview of current research on the application of tissue engineering to promote growth plate regeneration. It aims to elucidates the underlying mechanisms by which tissue engineering promotes growth plate regeneration and to provide novel insights and therapeutic strategies for future research on the regeneration of growth plate.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Pengfei Zheng
- Department of Orthopaedic Surgery, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
You Q, Lu M, Li Z, Zhou Y, Tu C. Cell Sheet Technology as an Engineering-Based Approach to Bone Regeneration. Int J Nanomedicine 2022; 17:6491-6511. [PMID: 36573205 PMCID: PMC9789707 DOI: 10.2147/ijn.s382115] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/12/2022] [Indexed: 12/24/2022] Open
Abstract
Bone defects that are congenital or the result of infection, malignancy, or trauma represent a challenge to the global healthcare system. To address this issue, multiple research groups have been developing novel cell sheet technology (CST)-based approaches to promote bone regeneration. These methods hold promise for use in regenerative medicine because they preserve cell-cell contacts, cell-extracellular matrix interactions, and the protein makeup of cell membranes. This review introduces the concept and preparation system of the cell sheet (CS), explores the application of CST in bone regeneration, highlights the current states of the bone regeneration via CST, and offers perspectives on the challenges and future research direction of translating current knowledge from the lab to the clinic.
Collapse
Affiliation(s)
- Qi You
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China,Sichuan Model Worker and Craftsman Talent Innovation Research Studio, Chengdu, Sichuan Province, People’s Republic of China
| | - Minxun Lu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China,Sichuan Model Worker and Craftsman Talent Innovation Research Studio, Chengdu, Sichuan Province, People’s Republic of China
| | - Zhuangzhuang Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China,Sichuan Model Worker and Craftsman Talent Innovation Research Studio, Chengdu, Sichuan Province, People’s Republic of China
| | - Yong Zhou
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China,Sichuan Model Worker and Craftsman Talent Innovation Research Studio, Chengdu, Sichuan Province, People’s Republic of China
| | - Chongqi Tu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China,Sichuan Model Worker and Craftsman Talent Innovation Research Studio, Chengdu, Sichuan Province, People’s Republic of China,Correspondence: Chongqi Tu; Yong Zhou, Department of Orthopedics, West China Hospital, Sichuan University, No. 37, Guoxuexiang, Chengdu, 610041, Sichuan Province, People’s Republic of China, Email ;
| |
Collapse
|
32
|
Nagahara AI, Homma J, Ryu B, Sekine H, Higashi Y, Shimizu T, Kawamata T. Networked lymphatic endothelial cells in a transplanted cell sheet contribute to form functional lymphatic vessels. Sci Rep 2022; 12:21698. [PMID: 36522421 PMCID: PMC9755306 DOI: 10.1038/s41598-022-26041-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
This study evaluated whether cell sheets containing a network of lymphatic endothelial cells (LECs) promoted lymphangiogenesis after transplantation in vivo. Cell sheets with a LEC network were constructed by co-culturing LECs and adipose-derived stem cells (ASCs) on temperature-responsive culture dishes. A cell ratio of 3:2 (vs. 1:4) generated networks with more branches and longer branch lengths. LEC-derived lymphatic vessels were observed 2 weeks after transplantation of a three-layered cell sheet construct onto rat gluteal muscle. Lymphatic vessel number, diameter and depth were greatest for a construct comprising two ASC sheets stacked on a LEC/ASC (3:2 ratio) sheet. Transplantation of this construct in a rat model of femoral lymphangiectomy led to the formation of functional lymphatic vessels containing both transplanted and host LECs. Further development of this technique may lead to a new method of promoting lymphangiogenesis.
Collapse
Affiliation(s)
- Ayumi Inoue Nagahara
- grid.410818.40000 0001 0720 6587Department of Neurosurgery, Graduate School of Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Jun Homma
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Bikei Ryu
- grid.488555.10000 0004 1771 2637Department of Neurosurgery, Tokyo Women’s Medical University Hospital, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Hidekazu Sekine
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Yuhei Higashi
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan ,Tokaihit Co., Ltd., Shizuoka, Japan
| | - Tatsuya Shimizu
- grid.410818.40000 0001 0720 6587Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| | - Takakazu Kawamata
- grid.488555.10000 0004 1771 2637Department of Neurosurgery, Tokyo Women’s Medical University Hospital, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666 Japan
| |
Collapse
|
33
|
Bibliometrics of Functional Polymeric Biomaterials with Bioactive Properties Prepared by Radiation-Induced Graft Copolymerisation: A Review. Polymers (Basel) 2022; 14:polym14224831. [PMID: 36432958 PMCID: PMC9692568 DOI: 10.3390/polym14224831] [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: 09/19/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Functional polymeric biomaterials (FPBMs) with bioactive characteristics obtained by radiation-induced graft copolymerisation (RIGC) have been subjected to intensive research and developed into many commercial products. Various studies have reported the development of a variety of radiation-grafted FPBMs. However, no reports dealing with the quantitative evaluations of these studies from a global bibliographic perspective have been published. Such bibliographic analysis can provide information to overcome the limitations of the databases and identify the main research trends, together with challenges and future directions. This review aims to provide an unprecedented bibliometric analysis of the published literature on the use of RIGC for the preparation of FPBMs and their applications in medical, biomedical, biotechnological, and health care fields. A total of 235 publications obtained from the Web of Science (WoS) in the period of 1985-2021 were retrieved, screened, and evaluated. The records were used to manifest the contributions to each field and underline not only the top authors, journals, citations, years of publication, and countries but also to highlight the core research topics and the hubs for research excellence on these materials. The obtained data overviews are likely to provide guides to early-career scientists and their research institutions and promote the development of new, timely needed radiation-grafted FPBMs, in addition to extending their applications.
Collapse
|
34
|
Smart surface-based cell sheet engineering for regenerative medicine. TRENDS IN CHEMISTRY 2022. [DOI: 10.1016/j.trechm.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Chen C, Shi Q, Li M, Chen Y, Zhang T, Xu Y, Liao Y, Ding S, Wang Z, Li X, Zhao C, Sun L, Hu J, Lu H. Engineering an enthesis-like graft for rotator cuff repair: An approach to fabricate highly biomimetic scaffold capable of zone-specifically releasing stem cell differentiation inducers. Bioact Mater 2022; 16:451-471. [PMID: 35386315 PMCID: PMC8965727 DOI: 10.1016/j.bioactmat.2021.12.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/29/2021] [Accepted: 12/19/2021] [Indexed: 02/09/2023] Open
Abstract
Rotator cuff (RC) attaches to humerus across a triphasic yet continuous tissue zones (bone-fibrocartilage-tendon), termed "enthesis". Regrettably, rapid and functional enthesis regeneration is challenging after RC tear. The existing grafts bioengineered for RC repair are insufficient, as they were engineered by a scaffold that did not mimic normal enthesis in morphology, composition, and tensile property, meanwhile cannot simultaneously stimulate the formation of bone-fibrocartilage-tendon tissues. Herein, an optimized decellularization approach based on a vacuum aspiration device (VAD) was developed to fabricate a book-shaped decellularized enthesis matrix (O-BDEM). Then, three recombinant growth factors (CBP-GFs) capable of binding collagen were synthesized by fusing a collagen-binding peptide (CBP) into the N-terminal of BMP-2, TGF-β3, or GDF-7, and zone-specifically tethered to the collagen of O-BDEM to fabricate a novel scaffold (CBP-GFs/O-BDEM) satisfying the above-mentioned requirements. After ensuring the low immunogenicity of CBP-GFs/O-BDEM by a novel single-cell mass cytometry in a mouse model, we interleaved urine-derived stem cell-sheets into this CBP-GFs/O-BDEM to bioengineer an enthesis-like graft. Its high-performance on regenerating enthesis was determined in a canine model. These findings indicate this CBP-GFs/O-BDEM may be an excellent scaffold for constructing enthesis-like graft to patch large/massive RC tears, and provide breakthroughs in fabricating graded interfacial tissue.
Collapse
Affiliation(s)
- Can Chen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Qiang Shi
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Muzhi Li
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yang Chen
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Tao Zhang
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yan Xu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yunjie Liao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Shulin Ding
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Zhanwen Wang
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xing Li
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Chunfeng Zhao
- Division of Orthopedic Research and Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, 55905, United States
| | - Lunquan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jianzhong Hu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Corresponding author. Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, Hunan, China.
| | - Hongbin Lu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, 410008, China
- Hunan Engineering Research Center of Sports and Health, Changsha, 410008, China
- Xiangya Hospital-International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, 410008, China
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Corresponding author. Xiangya Hospital, Central South University, No. 87, Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
36
|
Freitas-Ribeiro S, Reis RL, Pirraco RP. Long-term and short-term preservation strategies for tissue engineering and regenerative medicine products: state of the art and emerging trends. PNAS NEXUS 2022; 1:pgac212. [PMID: 36714838 PMCID: PMC9802477 DOI: 10.1093/pnasnexus/pgac212] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/09/2022] [Accepted: 09/28/2022] [Indexed: 02/01/2023]
Abstract
There is an ever-growing need of human tissues and organs for transplantation. However, the availability of such tissues and organs is insufficient by a large margin, which is a huge medical and societal problem. Tissue engineering and regenerative medicine (TERM) represent potential solutions to this issue and have therefore been attracting increased interest from researchers and clinicians alike. But the successful large-scale clinical deployment of TERM products critically depends on the development of efficient preservation methodologies. The existing preservation approaches such as slow freezing, vitrification, dry state preservation, and hypothermic and normothermic storage all have issues that somehow limit the biomedical applications of TERM products. In this review, the principles and application of these approaches will be summarized, highlighting their advantages and limitations in the context of TERM products preservation.
Collapse
Affiliation(s)
- Sara Freitas-Ribeiro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal,ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Barco GMR, Portugal
| | - Rui L Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal,ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Barco GMR, Portugal
| | | |
Collapse
|
37
|
Tanaka RI, Sakaguchi K, Yoshida A, Takahashi H, Haraguchi Y, Shimizu T. Production of scaffold-free cell-based meat using cell sheet technology. NPJ Sci Food 2022; 6:41. [PMID: 36057641 PMCID: PMC9440907 DOI: 10.1038/s41538-022-00155-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 08/09/2022] [Indexed: 12/01/2022] Open
Abstract
In the production of cell-based meat, it is desirable to reduce animal-derived materials as much as possible to meet the challenges of sustainability. Here, we demonstrate the “cell sheet-based meat”: scaffold-free cell-based meat using cell sheet technology and characterize its texture and nutrients. Bovine myoblast cell sheets were prepared using temperature-responsive culture dishes (TRCDs) and 10 stacked cell sheets to fabricate three-dimensional tissue of 1.3–2.7 mm thickness. Hardness was increased by incubation on the TRCD and was further increased by boiling as is characteristic of natural meat. The wet weight percentage of total protein in the cell sheet was about half that of beef. In this method, large-sized items of cell sheet-based meat were also created by simply scaling up the TRCD. This method promises an environment-friendly food product.
Collapse
Affiliation(s)
- Ryu-Ichiro Tanaka
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Katsuhisa Sakaguchi
- Department of Integrative Bioscience and Biomedical Engineering, Graduate School of Advanced Science and Engineering, TWIns, Waseda University, Tokyo, Japan.
| | - Azumi Yoshida
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Hironobu Takahashi
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Yuji Haraguchi
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
38
|
Guerron A, Phan HT, Peñaloza-Arias C, Brambilla D, Roullin VG, Giasson S. Selectively triggered cell detachment from poly(N-isopropylacrylamide) microgel functionalized substrates. Colloids Surf B Biointerfaces 2022. [DOI: 10.1016/j.colsurfb.2022.112699] [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]
|
39
|
Stimuli-responsive polyelectrolyte multilayer films and microcapsules. Adv Colloid Interface Sci 2022; 310:102773. [DOI: 10.1016/j.cis.2022.102773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 08/20/2022] [Accepted: 09/05/2022] [Indexed: 12/28/2022]
|
40
|
The Potential of Cell Sheet Technology for Beta Cell Replacement Therapy. CURRENT TRANSPLANTATION REPORTS 2022. [DOI: 10.1007/s40472-022-00371-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Abstract
Purpose of Review
Here, we review the use of cell sheet technology using different cell types and its potential for restoring the extracellular matrix microenvironment, perfusion, and immunomodulatory action on islets and beta cells.
Recent Findings
Cell sheets can be produced with different fabrication techniques ranging from the widely used temperature responsive system to the magnetic system. A variety of cells have been used to produce cell sheets including skin fibroblasts, smooth muscle cells, human umbilical vein endothelial cells, and mesenchymal stem cells.
Summary
CST would allow to recreate the ECM of islets which would provide cues to support islet survival and improvement of islet function. Depending on the used cell type, different additional supporting properties like immunoprotection or cues for better revascularization could be provided. Furthermore, CST offers the possibility to use other implantation sites than inside the liver. Further research should focus on cell sheet thickness and size to generate a potential translational therapy.
Collapse
|
41
|
Yuan SM, Yang XT, Zhang SY, Tian WD, Yang B. Therapeutic potential of dental pulp stem cells and their derivatives: Insights from basic research toward clinical applications. World J Stem Cells 2022; 14:435-452. [PMID: 36157522 PMCID: PMC9350620 DOI: 10.4252/wjsc.v14.i7.435] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/25/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
For more than 20 years, researchers have isolated and identified postnatal dental pulp stem cells (DPSCs) from different teeth, including natal teeth, exfoliated deciduous teeth, healthy teeth, and diseased teeth. Their mesenchymal stem cell (MSC)-like immunophenotypic characteristics, high proliferation rate, potential for multidirectional differentiation and biological features were demonstrated to be superior to those of bone marrow MSCs. In addition, several main application forms of DPSCs and their derivatives have been investigated, including stem cell injections, modified stem cells, stem cell sheets and stem cell spheroids. In vitro and in vivo administration of DPSCs and their derivatives exhibited beneficial effects in various disease models of different tissues and organs. Therefore, DPSCs and their derivatives are regarded as excellent candidates for stem cell-based tissue regeneration. In this review, we aim to provide an overview of the potential application of DPSCs and their derivatives in the field of regenerative medicine. We describe the similarities and differences of DPSCs isolated from donors of different ages and health conditions. The methodologies for therapeutic administration of DPSCs and their derivatives are introduced, including single injections and the transplantation of the cells with a support, as cell sheets, or as cell spheroids. We also summarize the underlying mechanisms of the regenerative potential of DPSCs.
Collapse
Affiliation(s)
- Sheng-Meng Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xue-Ting Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Si-Yuan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wei-Dong Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bo Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
42
|
Munderere R, Kim SH, Kim C, Park SH. The Progress of Stem Cell Therapy in Myocardial-Infarcted Heart Regeneration: Cell Sheet Technology. Tissue Eng Regen Med 2022; 19:969-986. [PMID: 35857259 DOI: 10.1007/s13770-022-00467-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/30/2022] Open
Abstract
Various tissues, including the heart, cornea, bone, esophagus, bladder and liver, have been vascularized using the cell sheet technique. It overcomes the limitations of existing techniques by allowing small layers of the cell sheet to generate capillaries on their own, and it can also be used to vascularize tissue-engineered transplants. Cell sheets eliminate the need for traditional tissue engineering procedures such as isolated cell injections and scaffold-based technologies, which have limited applicability. While cell sheet engineering can eliminate many of the drawbacks, there are still a few challenges that need to be addressed. The number of cell sheets that can be layered without triggering core ischemia or hypoxia is limited. Even when scaffold-based technologies are disregarded, strategies to tackle this problem remain a substantial impediment to the efficient regeneration of thick, living three-dimensional cell sheets. In this review, we summarize the cell sheet technology in myocardial infarcted tissue regeneration.
Collapse
Affiliation(s)
- Raissa Munderere
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea.,The Center for Marine Integrated Biomedical Technology (BK21 PLUS), Pukyong National University, Busan, Republic of Korea
| | - Seon-Hwa Kim
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea.,The Center for Marine Integrated Biomedical Technology (BK21 PLUS), Pukyong National University, Busan, Republic of Korea
| | - Changsu Kim
- Department of Orthopedics Surgery, Kosin University Gospel Hospital, Busan, Republic of Korea
| | - Sang-Hyug Park
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, Republic of Korea. .,The Center for Marine Integrated Biomedical Technology (BK21 PLUS), Pukyong National University, Busan, Republic of Korea. .,Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan, 48513, Republic of Korea.
| |
Collapse
|
43
|
Açarı İK, Sel E, Özcan İ, Ateş B, Köytepe S, Thakur VK. Chemistry and engineering of brush type polymers: Perspective towards tissue engineering. Adv Colloid Interface Sci 2022; 305:102694. [PMID: 35597039 DOI: 10.1016/j.cis.2022.102694] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/21/2022] [Accepted: 05/06/2022] [Indexed: 11/01/2022]
Abstract
In tissue engineering, it is imperative to control the behaviour of cells/stem cells, such as adhesion, proliferation, propagation, motility, and differentiation for tissue regeneration. Surfaces that allow cells to behave in this way are critical as support materials in tissue engineering. Among these surfaces, brush-type polymers have an important potential for tissue engineering and biomedical applications. Brush structure and length, end groups, bonding densities, hydrophilicity, surface energy, structural flexibility, thermal stability, surface chemical reactivity, rheological and tribological properties, electron and energy transfer ability, cell binding and absorption abilities for various biological molecules of brush-type polymers were increased its importance in tissue engineering applications. In addition, thanks to these functional properties and adjustable surface properties, brush type polymers are used in different high-tech applications such as electronics, sensors, anti-fouling, catalysis, purification and energy etc. This review comprehensively highlights the use of brush-type polymers in tissue engineering applications. Considering the superior properties of brush-type polymer structures, it is believed that in the future, it will be an effective tool in structure designs containing many different biomolecules (enzymes, proteins, etc.) in the field of tissue engineering.
Collapse
|
44
|
Karasu T, Erkoc-Biradli FZ, Öztürk-Öncel MÖ, Armutcu C, Uzun L, Garipcan B, Çorman ME. Synthesis and characterization of stimuli-responsive hydrogels: evaluation of external stimuli influence on L929 fibroblast viability. Biomed Phys Eng Express 2022; 8. [PMID: 35738237 DOI: 10.1088/2057-1976/ac7baa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/23/2022] [Indexed: 11/11/2022]
Abstract
In this study, poly(2-hydroxyethyl methacrylate) [p(HEMA)] based hydrogels responsive to the pH, temperature and magnetic field were synthesized. The surface properties of p(HEMA) were improved by designing the stimuli-responsive hydrogels made of MAGA, NIPAAm and methacrylate-decorated magnetite nanoparticles as a function of pH-, thermo- and magnetic responsive cell culture surfaces. These materials were then modified an abundant extracellular matrix component, type I collagen, which has been considered as a biorecognition element to increase the applicability of hydrogels to cell viability. Based on results from scanning electron microscopy (SEM) and thermal gravimetric analysis (TGA), stimuli-responsive hydrogel demonstrated improved non-porous structures and thermal stability with a high degree of cross-linking. Mechanical analyses of the hydrogels also showed that stimuli-responsive hydrogels are more elastomeric due to the polymeric chains and heterogeneous amorphous segments compared to plain hydrogels. Furthermore, surface modification of hydrogels with collagen provided better biocompatibility, which was confirmed with L929 fibroblast cell adhesion. Produced stimuli-responsive hydrogels modulated cellular viability by changing pH and magnetic field.
Collapse
Affiliation(s)
- Tunca Karasu
- Department of Chemistry, Hacettepe University, Hacettepe Üniversitesi, Beytepe-Ankara, Ankara, 06800, TURKEY
| | - Fatma Zehra Erkoc-Biradli
- Boğaziçi Üniversitesi Biomedikal Mühendisliği Enstitüsü, Boğaziçi University, Kandilli-İstanbul, Istanbul, 34684, TURKEY
| | - M Özgen Öztürk-Öncel
- Boğaziçi Üniversitesi Biomedikal Mühendisliği Enstitüsü, Boğaziçi Üniversitesi, Kandilli, Istanbul, 34684, TURKEY
| | - Canan Armutcu
- Department of Chemistry, Hacettepe University, Hacettepe Ünviersitesi, Beytepe-Ankara, Ankara, 06800, TURKEY
| | - Lokman Uzun
- Department of Chemistry, Hacettepe University, Hacettepe Üniversitesi, Beytepe-Ankara, Ankara, 06800, TURKEY
| | - Bora Garipcan
- Boğaziçi Üniversitesi Biomedikal Mühendisliği Enstitüsü, Boğaziçi Üniversites, Kandilli, Istanbul, 34684, TURKEY
| | - Mehmet Emin Çorman
- Department of Biochemistry, University of Health Sciences, Sağlık Bilimleri Üniversitesi, Etlik, Istanbul, Ankara, 34668, TURKEY
| |
Collapse
|
45
|
Wiersma LE, Avramut MC, Lievers E, Rabelink TJ, van den Berg CW. Large-scale engineering of hiPSC-derived nephron sheets and cryopreservation of their progenitors. Stem Cell Res Ther 2022; 13:208. [PMID: 35578313 PMCID: PMC9109372 DOI: 10.1186/s13287-022-02881-5] [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: 11/26/2021] [Accepted: 04/30/2022] [Indexed: 11/17/2022] Open
Abstract
Background The generation of human induced pluripotent stem cells (hiPSCs) has opened a world of opportunities for stem cell-based therapies in regenerative medicine. Currently, several human kidney organoid protocols are available that generate organoids containing kidney structures. However, these kidney organoids are relatively small ranging up to 0.13 cm2 and therefore contain a small number of nephrons compared to an adult kidney, thus defying the exploration of future use for therapy. Method We have developed a scalable, easily accessible, and reproducible protocol to increase the size of the organoid up to a nephron sheet of 2.5 cm2 up to a maximum of 12.6 cm2 containing a magnitude of nephrons. Results Confocal microscopy showed that the subunits of the nephrons remain evenly distributed throughout the entire sheet and that these tissue sheets can attain ~ 30,000–40,000 glomerular structures. Upon transplantation in immunodeficient mice, such nephron sheets became vascularized and matured. They also show reuptake of injected low-molecular mass dextran molecules in the tubular structures, indicative of glomerular filtration. Furthermore, we developed a protocol for the cryopreservation of intermediate mesoderm cells during the differentiation and demonstrate that these cells can be successfully thawed and recovered to create such tissue sheets. Conclusion The scalability of the procedures, and the ability to cryopreserve the cells during differentiation are important steps forward in the translation of these differentiation protocols to future clinical applications such as transplantable auxiliary kidney tissue. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02881-5.
Collapse
Affiliation(s)
- Loes E Wiersma
- Department of Internal Medicine - Nephrology, Leiden University Medical Center, Postal Zone C7-Q, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - M Cristina Avramut
- Department of Cell and Chemical Biology - Electron Microscopy, Leiden University Medical Center, Postal zone S-1-P, Einthovenweg 20, 2333 ZC, Leiden, The Netherlands
| | - Ellen Lievers
- Department of Internal Medicine - Nephrology, Leiden University Medical Center, Postal Zone C7-Q, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine - Nephrology, Leiden University Medical Center, Postal Zone C7-Q, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Cathelijne W van den Berg
- Department of Internal Medicine - Nephrology, Leiden University Medical Center, Postal Zone C7-Q, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands. .,Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
46
|
Hung LT, Poon SHL, Yan WH, Lace R, Zhou L, Wong JKW, Williams RL, Shih KC, Shum HC, Chan YK. Scaffold-Free Strategy Using a PEG-Dextran Aqueous Two-Phase-System for Corneal Tissue Repair. ACS Biomater Sci Eng 2022; 8:1987-1999. [PMID: 35362956 DOI: 10.1021/acsbiomaterials.1c01500] [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] [Indexed: 11/29/2022]
Abstract
Forming thin tissue constructs with minimal extracellular matrix surrounding them is important for tissue engineering applications. Here, we explore and optimize a strategy that enables rapid fabrication of scaffold-free corneal tissue constructs using the liquid-liquid interface of an aqueous two-phase system (ATPS) that is based on biocompatible polymers, dextran and polyethylene glycol. Intact tissue-like constructs, made of corneal epithelial or endothelial cells, can be formed on the interface between the two liquid phases of ATPS within hours and subsequently collected simply by removing the liquid phases. The formed corneal cell constructs express essential physiological markers and have preserved viability and proliferative ability in vitro. The corneal epithelial cell constructs are also able to re-epithelialize the corneal epithelial wound in vitro. The results suggest the promise of our reported strategy in corneal repair.
Collapse
Affiliation(s)
- Lap Tak Hung
- Department of Mechanical Engineering, Faculty of Engineering, University of Hong Kong, Rm 7-25, Haking Wong Building, Pokfulam Road, Hong Kong SAR 999077, China
| | - Stephanie Hiu Ling Poon
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, 301B Cyberport 4, 100 Cyberport Road, Pokfulam, Hong Kong SAR 999077, China
| | - Wing Huen Yan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, 301B Cyberport 4, 100 Cyberport Road, Pokfulam, Hong Kong SAR 999077, China
| | - Rebecca Lace
- Department of Eye and Vision Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, U.K
| | - Liangyu Zhou
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, 301B Cyberport 4, 100 Cyberport Road, Pokfulam, Hong Kong SAR 999077, China
| | - Jasper Ka Wai Wong
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, 301B Cyberport 4, 100 Cyberport Road, Pokfulam, Hong Kong SAR 999077, China
| | - Rachel L Williams
- Department of Eye and Vision Sciences, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, U.K
| | - Kendrick Co Shih
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, 301B Cyberport 4, 100 Cyberport Road, Pokfulam, Hong Kong SAR 999077, China
| | - Ho Cheung Shum
- Department of Mechanical Engineering, Faculty of Engineering, University of Hong Kong, Rm 7-25, Haking Wong Building, Pokfulam Road, Hong Kong SAR 999077, China
| | - Yau Kei Chan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, University of Hong Kong, 301B Cyberport 4, 100 Cyberport Road, Pokfulam, Hong Kong SAR 999077, China
| |
Collapse
|
47
|
Xu W, Wang T, Wang Y, Wu X, Chen Y, Song D, Ci Z, Cao Y, Hua Y, Zhou G, Liu Y. An Injectable Platform of Engineered Cartilage Gel and Gelatin Methacrylate to Promote Cartilage Regeneration. Front Bioeng Biotechnol 2022; 10:884036. [PMID: 35528206 PMCID: PMC9074996 DOI: 10.3389/fbioe.2022.884036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/30/2022] [Indexed: 12/04/2022] Open
Abstract
Cell–hydrogel constructs are frequently used as injectable platforms for irregular cartilage regeneration. However, cell–hydrogel constructs have obvious disadvantages, such as long culture times, high probability of infection, and poor cartilage formation capacity, significantly limiting their clinical translation. In this study, we aimed to develop a novel injectable platform comprising engineered cartilage gel (ECG) and gelatin methacrylate (GelMA) to improve cartilage regeneration. We first prepared an ECG by cutting the in vitro engineered cartilage sheet into pieces. The chondrocytes and ECG were evenly encapsulated into GelMA to form Cell-GelMA and ECG-GelMA constructs. The ECG-GelMA construct exhibited preferred gel characteristics and superior biocompatibility compared with the Cell-GelMA construct counterpart. After subcutaneous implantation in nude mice and goat, both gross views and histological evaluations showed that the ECG-GelMA construct achieved more homogenous, stable, and mature cartilage regeneration than the Cell-GelMA construct. Immunological evaluations showed that ECG-GelMA had a mitigatory immunologic reaction than the Cell-GelMA construct. Overall, the results suggest that the ECG-GelMA is a promising injectable platform for cartilage regeneration that may advance clinical translation.
Collapse
Affiliation(s)
- Wei Xu
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, China
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Wang
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, China
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yahui Wang
- National Tissue Engineering Center of China, Shanghai, China
| | - Xiaodi Wu
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, China
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujie Chen
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Daiying Song
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, China
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Ci
- Shanghai Resthetic Bio CO., LTD, Shanghai, China
| | - Yilin Cao
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujie Hua
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yujie Hua, ; Guangdong Zhou, Yu Liu,
| | - Guangdong Zhou
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, China
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yujie Hua, ; Guangdong Zhou, Yu Liu,
| | - Yu Liu
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, China
- National Tissue Engineering Center of China, Shanghai, China
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Stem Cell Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Resthetic Bio CO., LTD, Shanghai, China
- *Correspondence: Yujie Hua, ; Guangdong Zhou, Yu Liu,
| |
Collapse
|
48
|
Berntsen L, Forghani A, Hayes DJ. Mesenchymal Stem Cell Sheets for Engineering of the Tendon-Bone Interface. Tissue Eng Part A 2022; 28:341-352. [PMID: 34476994 PMCID: PMC9057909 DOI: 10.1089/ten.tea.2021.0072] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/25/2021] [Indexed: 11/12/2022] Open
Abstract
Failure to regenerate the gradient tendon-bone interface of the enthesis results in poor clinical outcomes for surgical repair. The goal of this study was to evaluate the potential of composite cell sheets for engineering of the tendon-bone interface to improve regeneration of the functionally graded tissue. We hypothesize that stacking cell sheets at early stages of differentiation into tenogenic and osteogenic progenitors will create a composite structure with integrated layers. Cell sheets were fabricated on methyl cellulose and poly(N-isopropylacrylamide) thermally reversible polymers with human adipose-derived stem cells and differentiated into progenitors of tendon and bone with chemical induction media. Tenogenic and osteogenic cell sheets were stacked, and the engineered tendon-bone interface (TM-OM) was characterized in vitro in comparison to stacked cell sheet controls cultured in basal growth medium (GM-GM), osteogenic medium (OM-OM), and tenogenic medium (TM-TM). Samples were characterized by histology, quantitative real-time polymerase chain reaction, and immunofluorescent staining for markers of tendon, fibrocartilage, and bone including mineralization, scleraxis, tenomodulin, COL2, COLX, RUNX2, osteonectin, and osterix. After 1 week co-culture in basal growth medium, TM-OM cell sheets formed a tissue construct with integrated layers expressing markers of tendon, mineralized fibrocartilage, and bone with a spatial gradient in RUNX2 expression. Tenogenic cell sheets had increased expression of scleraxis and tenomodulin. Osteogenic cell sheets exhibited mineralization 1 week after stacking and upregulation of osterix and osteonectin. Additionally, in the engineered interface, there was significantly increased gene expression of IHH and COLX, indicative of endochondral ossification. These results highlight the potential for composite cell sheets fabricated with adipose-derived stem cells for engineering of the tendon-bone interface. Impact statement This study presents a method for fabrication of the tendon-bone interface using stacked cell sheets of tenogenic and osteogenic progenitors differentiated from human adipose-derived mesenchymal stem cells, resulting in a composite structure expressing markers of tendon, mineralized fibrocartilage, and bone. This work is an important step toward regeneration of the biological gradient of the enthesis and demonstrates the potential for engineering complex tissue interfaces from a single autologous cell source to facilitate clinical translation.
Collapse
Affiliation(s)
- Lisa Berntsen
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Anoosha Forghani
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
49
|
Agarwal A, Rao GK, Majumder S, Shandilya M, Rawat V, Purwar R, Verma M, Srivastava CM. Natural protein-based electrospun nanofibers for advanced healthcare applications: progress and challenges. 3 Biotech 2022; 12:92. [PMID: 35342680 PMCID: PMC8921418 DOI: 10.1007/s13205-022-03152-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
Electrospinning is an electrostatic fiber fabrication technique that operates by the application of a strong electric field on polymer solution or melts. It is used to fabricate fibers whose size lies in the range of few microns to the nanometer range. Historic development of electrospinning has evinced attention due to its outstanding attributes such as small diameter, excellent pore inter-connectivity, high porosity, and high surface-to-volume ratio. This review aims to highlight the theory behind electrospinning and the machine setup with a detailed discussion about the processing parameters. It discusses the latest innovations in natural protein-based electrospun nanofibers for health care applications. Various plant- and animal-based proteins have been discussed with detailed sample preparation and corresponding processing parameters. The usage of these electrospun nanofibers in regenerative medicine and drug delivery has also been discussed. Some technical innovations in electrospinning techniques such as emulsion electrospinning and coaxial electrospinning have been highlighted. Coaxial electrospun core-shell nanofibers have the potential to be utilized as an advanced nano-architecture for sustained release targeted delivery as well as for regenerative medicine. Healthcare applications of nanofibers formed via emulsion and coaxial electrospinning have been discussed briefly. Electrospun nanofibers have still much scope for commercialization on large scale. Some of the available wound-dressing materials have been discussed in brief.
Collapse
Affiliation(s)
- Anushka Agarwal
- Department of Chemistry, Biochemistry and Forensic Science, Amity School of Applied Sciences, Amity University Haryana, Gurugram, 122413 India
| | - Gyaneshwar K. Rao
- Department of Chemistry, Biochemistry and Forensic Science, Amity School of Applied Sciences, Amity University Haryana, Gurugram, 122413 India
| | - Sudip Majumder
- Department of Chemistry, Biochemistry and Forensic Science, Amity School of Applied Sciences, Amity University Haryana, Gurugram, 122413 India
| | - Manish Shandilya
- Department of Chemistry, Biochemistry and Forensic Science, Amity School of Applied Sciences, Amity University Haryana, Gurugram, 122413 India
| | - Varun Rawat
- Department of Chemistry, Biochemistry and Forensic Science, Amity School of Applied Sciences, Amity University Haryana, Gurugram, 122413 India
| | - Roli Purwar
- Department of Applied Chemistry, Delhi Technological University, New Delhi, Delhi 110042 India
| | - Monu Verma
- Department of Environmental Engineering, University of Seoul, Seoul, 130743 South Korea
| | - Chandra Mohan Srivastava
- Department of Chemistry, Biochemistry and Forensic Science, Amity School of Applied Sciences, Amity University Haryana, Gurugram, 122413 India
- Centre for Polymer Technology, Amity School of Applied Sciences, Amity University Haryana, Gurugram, 122413 India
| |
Collapse
|
50
|
Santos LF, Patrício SG, Silva AS, Mano JF. Freestanding Magnetic Microtissues for Tissue Engineering Applications. Adv Healthc Mater 2022; 11:e2101532. [PMID: 34921719 DOI: 10.1002/adhm.202101532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/10/2021] [Indexed: 02/06/2023]
Abstract
A long-sought goal in tissue engineering (TE) is the development of tissues able to recapitulate the complex architecture of the native counterpart. Microtissues, by resembling the functional units of living structures, can be used to recreate tissues' architecture. Howbeit, microfabrication methodologies fail to reproduce cell-based tissues with uniform shape. At the macroscale, complex tissues are already produced by magnetic-TE using solely magnetized cells as building materials. The enhanced extracellular matrix (ECM) deposition guaranties the conservation of tissues' architecture, leading to a successful cellular engraftment. Following the same rational, now the combination of a versatile microfabrication-platform is proposed with magnetic-TE to generate robust micro-tissues with complex architecture for TE purposes. Small tissue units with circle, square, and fiber-like shapes are designed with high fidelity acting as building blocks for engineering complex tissues. Notably, freestanding microtissues maintain their geometry after 7 days post-culturing, overcoming the challenges of microtissues fabrication. Lastly, the ability of microtissues in invading distinct tissue models while releasing trophic factors is substantiated in methacryloyl laminarin (LAM) and platelet lysates (PLMA) hydrogels. By simply using cells as building units and such microfabrication-platform, the fabrication of complex multiscale and multifunctional tissues with clinical relevance is envisaged, including for therapies or disease models.
Collapse
Affiliation(s)
- Lúcia F. Santos
- Department of Chemistry CICECO–Aveiro Institute of Materials University of Aveiro Aveiro 3810‐193 Portugal
| | - Sónia G. Patrício
- Department of Chemistry CICECO–Aveiro Institute of Materials University of Aveiro Aveiro 3810‐193 Portugal
| | - Ana Sofia Silva
- Department of Chemistry CICECO–Aveiro Institute of Materials University of Aveiro Aveiro 3810‐193 Portugal
| | - João F. Mano
- Department of Chemistry CICECO–Aveiro Institute of Materials University of Aveiro Aveiro 3810‐193 Portugal
| |
Collapse
|