1
|
Nochalabadi A, Khazaei M, Rezakhani L. Exosomes and tissue engineering: A novel therapeutic strategy for nerve regenerative. Tissue Cell 2025; 93:102676. [PMID: 39693896 DOI: 10.1016/j.tice.2024.102676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/10/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Damage to nerves negatively impacts quality of life and causes considerable morbidity. Self-regeneration is a special characteristic of the nervous system, yet how successful regeneration is accomplished remains unclear. Research on nerve regeneration is advancing and accelerating successful nerve recovery with potential new approaches. Eukaryote cells release extracellular vesicles (EVs), which control intercellular communication in both health and disease. More and more, EVs such as microvesicles and exosomes (EXOs) are being recognized as viable options for cell-free therapies that address complex tissue regeneration. The present study highlights the functional relevance of EVs in regenerative medicine for nerve-related regeneration. A subclass of EVs, EXOs were first identified as a way for cells to expel undesirable cell products. These nanovesicles have a diameter of 30-150 nm and are secreted by a variety of cells in conditions of both health and illness. Their benefits include the ability to promote endothelial cell growth, inhibit inflammation, encourage cell proliferation, and regulate cell differentiation. They are also known to transport functional proteins, metabolites, and nucleic acids to recipient cells, thus playing a significant role in cellular communication. EXOs impact an extensive array of physiological functions, including immunological responses, tissue regeneration, stem cell conservation, communication within the central nervous system, and pathological processes involving cardiovascular disorders, neurodegeneration, cancer, and inflammation. Their biocompatibility and bi-layered lipid structure (which shields the genetic consignment from deterioration and reduces immunogenicity) make them appealing as therapeutic vectors. They can pass through the blood brain barrier and other major biological membranes because of their small size and membrane composition. The creation of modified EXOs is a dynamic area of research that supports the evaluation of diverse therapeutic freights, improvement of target selectivity, and manufacturing optimization.
Collapse
Affiliation(s)
- Azadeh Nochalabadi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
2
|
Daghrery A, Araújo IJDS, Marques JF, Alipour M, Ünsal RBK, Chathoth BM, Sivaramakrishnan G, Delgadillo-Barrera S, Chaurasia A. Role of exosomes in dental and craniofacial regeneration - A review. Tissue Cell 2025; 93:102684. [PMID: 39740273 DOI: 10.1016/j.tice.2024.102684] [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: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The treatment of congenital deformities, traumatic injuries, infectious diseases, and tumors in the craniomaxillofacial (CMF) region is complex due to the intricate nature of the tissues involved. Conventional treatments such as bone grafts and cell transplantation face limitations, including the need for multiple surgeries, complications, and safety concerns. OBJECTIVE This paper aims to provide a comprehensive analysis of the role of exosomes (EXOs) in CMF and dental tissue regeneration and to explore their potential applications in regenerative dental medicine. METHODS An extensive review of advancements in tissue engineering, materials sciences, and nanotechnology was conducted to evaluate the development of delivery systems for EXOs-based therapies. The analysis included how EXOs, as nanovesicles released by cells, can be modified to target specific cells or loaded with functional molecules for drug or gene delivery. RESULTS EXOs have emerged as a promising alternative to cell transplant therapy, offering a safer method for cell communication and epigenetic control. EXOs transport important proteins and genetic materials, facilitating intercellular communication and delivering therapeutics effectively. The potential of EXOs in personalized medicine, particularly in diagnosing, customizing treatment, and predicting patient responses, is highlighted. CONCLUSION EXO-mediated therapy holds significant potential for advancing tissue regeneration, offering targeted, personalized treatment options with reduced side effects. However, challenges in purification, production, and standardized protocols need to be addressed before its clinical application can be fully realized.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | | | - Joana Faria Marques
- Faculdade de Medicina Dentária, Universidade de Lisboa, Cidade Universitária, Lisboa 1600-277, Portugal.
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Iran; Departments of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, USA.
| | - Revan Birke Koca Ünsal
- Department of Periodontology, University of Kyrenia, Faculty of Dentistry, Kyrenia, Cyprus.
| | | | | | - Sara Delgadillo-Barrera
- Grupo de Investigacion Básica y Aplicada en Odontología - IBAPO, Facultad de Odontologia, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, Faculty of Dental Sciences. King George's Medical University, Lucknow, India.
| |
Collapse
|
3
|
Liu L, Liu W, Han Z, Shan Y, Xie Y, Wang J, Qi H, Xu Q. Extracellular Vesicles-in-Hydrogel (EViH) targeting pathophysiology for tissue repair. Bioact Mater 2025; 44:283-318. [PMID: 39507371 PMCID: PMC11539077 DOI: 10.1016/j.bioactmat.2024.10.017] [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: 04/06/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
Regenerative medicine endeavors to restore damaged tissues and organs utilizing biological approaches. Utilizing biomaterials to target and regulate the pathophysiological processes of injured tissues stands as a crucial method in propelling this field forward. The Extracellular Vesicles-in-Hydrogel (EViH) system amalgamates the advantages of extracellular vesicles (EVs) and hydrogels, rendering it a prominent biomaterial in regenerative medicine with substantial potential for clinical translation. This review elucidates the development and benefits of the EViH system in tissue regeneration, emphasizing the interaction and impact of EVs and hydrogels. Furthermore, it succinctly outlines the pathophysiological characteristics of various types of tissue injuries such as wounds, bone and cartilage injuries, cardiovascular diseases, nerve injuries, as well as liver and kidney injuries, underscoring how EViH systems target these processes to address related tissue damage. Lastly, it explores the challenges and prospects in further advancing EViH-based tissue regeneration, aiming to impart a comprehensive understanding of EViH. The objective is to furnish a thorough overview of EViH in enhancing regenerative medicine applications and to inspire researchers to devise innovative tissue engineering materials for regenerative medicine.
Collapse
Affiliation(s)
- Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266003, China
| | - Zeyu Han
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yansheng Shan
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Jialu Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Quanchen Xu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| |
Collapse
|
4
|
Zhou L, Zhang C, Shi T, Wu D, Chen H, Han J, Chen D, Lin J, Liu W. Functionalized 3D-printed GelMA/Laponite hydrogel scaffold promotes BMSCs recruitment through osteoimmunomodulatory enhance osteogenic via AMPK/mTOR signaling pathway. Mater Today Bio 2024; 29:101261. [PMID: 39381262 PMCID: PMC11460517 DOI: 10.1016/j.mtbio.2024.101261] [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: 05/30/2024] [Revised: 09/04/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
The migration and differentiation of bone marrow mesenchymal stem cells (BMSCs) play crucial roles in bone repair processes. However, conventional scaffolds often lack of effectively inducing and recruiting BMSCs. In our study, we present a novel approach by introducing a 3D-bioprinted scaffold composed of hydrogels, with the addition of laponite to the GelMA solution, aimed at enhancing scaffold performance. Both in vivo and in vitro experiments have confirmed the outstanding biocompatibility of the scaffold. Furthermore, for the first time, Apt19s has been chemically modified onto the surface of the hydrogel scaffold, resulting in a remarkable enhancement in the migration and adhesion of BMSCs. Moreover, the scaffold has demonstrated robust osteogenic differentiation capability in both in vivo and in vitro environments. Additionally, the hydrogel scaffold has shown the ability to induce the polarization of macrophages from M1 to M2, thereby facilitating the osteogenic differentiation of BMSCs via the bone immune pathway. Through RNA-seq analysis, it has been revealed that macrophages regulate the osteogenic differentiation of BMSCs through the AMPK/mTOR signaling pathway. In summary, the functionalized GelMA/Laponite scaffold offers a cost-effective approach for tailored in situ bone regeneration.
Collapse
Affiliation(s)
- Linquan Zhou
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Chengcheng Zhang
- The School of Health, Fujian Medical University, Fuzhou, 350000, China
| | - Tengbin Shi
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Dingwei Wu
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Huina Chen
- The School of Health, Fujian Medical University, Fuzhou, 350000, China
| | - Jiaxin Han
- The School of Health, Fujian Medical University, Fuzhou, 350000, China
| | - Dehui Chen
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Jinxin Lin
- Key Laboratory of Optoelectronic Materials Chemistry and Physics, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350000, China
| | - Wenge Liu
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| |
Collapse
|
5
|
Li W, Li J, Pan C, Lee JS, Kim BS, Gao G. Light-based 3D bioprinting techniques for illuminating the advances of vascular tissue engineering. Mater Today Bio 2024; 29:101286. [PMID: 39435375 PMCID: PMC11492625 DOI: 10.1016/j.mtbio.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Vascular tissue engineering faces significant challenges in creating in vitro vascular disease models, implantable vascular grafts, and vascularized tissue/organ constructs due to limitations in manufacturing precision, structural complexity, replicating the composited architecture, and mimicking the mechanical properties of natural vessels. Light-based 3D bioprinting, leveraging the unique advantages of light including high resolution, rapid curing, multi-material adaptability, and tunable photochemistry, offers transformative solutions to these obstacles. With the emergence of diverse light-based 3D bioprinting techniques and innovative strategies, the advances in vascular tissue engineering have been significantly accelerated. This review provides an overview of the human vascular system and its physiological functions, followed by an in-depth discussion of advancements in light-based 3D bioprinting, including light-dominated and light-assisted techniques. We explore the application of these technologies in vascular tissue engineering for creating in vitro vascular disease models recapitulating key pathological features, implantable blood vessel grafts, and tissue analogs with the integration of capillary-like vasculatures. Finally, we provide readers with insights into the future perspectives of light-based 3D bioprinting to revolutionize vascular tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai 519088, China
| | - Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China
- School of Mechanical and Equipment Engineering, Hebei University of Engineering, Handan, 050024, China
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
6
|
Yu T, Zhao IS, Pan H, Yang J, Wang H, Deng Y, Zhang Y. Extracellular vesicle-functionalized bioactive scaffolds for bone regeneration. Asian J Pharm Sci 2024; 19:100945. [PMID: 39483718 PMCID: PMC11525715 DOI: 10.1016/j.ajps.2024.100945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 11/03/2024] Open
Abstract
The clinical need for effective bone regeneration in compromised conditions continues to drive demand for innovative solutions. Among emerging strategies, extracellular vesicles (EVs) have shown promise as an acellular approach for bone regeneration. However, their efficacy is hindered by rapid sequestration and clearance when administered via bolus injection. To address this challenge, EV-functionalized scaffolds have recently been proposed as an alternative delivery strategy to enhance EV retention and subsequent healing efficacy. This review aims to consolidate recent advancements in the development of EV-functionalized scaffolds for augmenting bone regeneration. It explores various sources of EVs and different strategies for integrating them into biomaterials. Furthermore, the mechanisms underlying their therapeutic effects in bone regeneration are elucidated. Current limitations in clinical translation and perspectives on the design of more efficient EVs for improved therapeutic efficacy are also presented. Overall, this review can provide inspiration for the development of novel EV-assisted grafts with superior bone regeneration potential.
Collapse
Affiliation(s)
- Taozhao Yu
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
| | - Irene Shuping Zhao
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| | - Hongguang Pan
- Department of Otolaryngology, Shenzhen Children Hospital, Shenzhen 518034, China
| | - Jianhua Yang
- Longgang District People's Hospital of Shenzhen & The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Huanan Wang
- Key State Laboratory of Fine Chemicals, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Yongqiang Deng
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University, Shenzhen 518055, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| | - Yang Zhang
- School of Dentistry, Shenzhen University Medical School, Shenzhen 518015, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518015, China
- Institute of Stomatological Research, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
7
|
Doshi RB, Vakil D, Molley TG, Islam MS, Kilian KA, Cunningham C, Sidhu KS. Mesenchymal stem cell-secretome laden photopolymerizable hydrogels for wound healing. J Biomed Mater Res A 2024; 112:1484-1493. [PMID: 38487991 DOI: 10.1002/jbm.a.37697] [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: 07/12/2023] [Revised: 02/17/2024] [Accepted: 02/21/2024] [Indexed: 07/12/2024]
Abstract
Mesenchymal stem cell-derived secretome represents an emerging acellular therapeutic which possess significant opportunity for clinical applications due to its anti-inflammatory, immunomodulatory, and wound healing properties. However, maintaining therapeutic efficacy and ensuring stability of cell-based products is challenging, requiring a robust delivery method. Therefore, we designed a hydrogel-based scaffold loaded with CK Cell Technologies' proprietary Mesenchymal stem cell-secretome for controlled release treatment of acute and chronic wounds. We incorporated both conditioned media (CM) and extracellular vesicles (EVs) into gelatin methacryloyl (GelMA) hydrogels and demonstrated how we can tune the diffusive release of the EVs from them. To demonstrate viability of the approach, we developed a wound healing scratch assay where we see in situ release of CM and EVs promote enhanced migration of human dermal fibroblasts (hDFs). We see the colocalization of these EVs in the fibroblasts using fluorescent microscopy. Finally, as a surrogate for in vivo neovascularization, we conducted an in vitro tube formation assay for the MSC-secretome using matrigel-embedded human microvascular endothelial cells. By adding CM and EVs, we observe an increase in tubulogenesis. Collectively, our data demonstrates by tuning the GelMA properties, we can influence the controlled release of the MSC-secretome for a wound dressing and bandage application for chronic and acute wounds.
Collapse
Affiliation(s)
- Riddhesh B Doshi
- R & D, CK Cell Technologies Pty Ltd, Sydney, New South Wales, Australia
- School of Chemistry, University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Devashree Vakil
- R & D, CK Cell Technologies Pty Ltd, Sydney, New South Wales, Australia
| | - Thomas G Molley
- School of Materials Science and Engineering, University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Md Shariful Islam
- School of Materials Science and Engineering, University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Kristopher A Kilian
- School of Chemistry, University of New South Wales (UNSW), Sydney, New South Wales, Australia
- School of Materials Science and Engineering, University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Corey Cunningham
- R & D, CK Cell Technologies Pty Ltd, Sydney, New South Wales, Australia
| | - Kuldip S Sidhu
- R & D, CK Cell Technologies Pty Ltd, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales (UNSW), Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Hwang HS, Lee CS. Nanoclay-Composite Hydrogels for Bone Tissue Engineering. Gels 2024; 10:513. [PMID: 39195042 DOI: 10.3390/gels10080513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
Nanoclay-composite hydrogels represent a promising avenue for advancing bone tissue engineering. Traditional hydrogels face challenges in providing mechanical strength, biocompatibility, and bioactivity necessary for successful bone regeneration. The incorporation of nanoclay into hydrogel matrices offers a potential unique solution to these challenges. This review provides a comprehensive overview of the fabrication, physico-chemical/biological performance, and applications of nanoclay-composite hydrogels in bone tissue engineering. Various fabrication techniques, including in situ polymerization, physical blending, and 3D printing, are discussed. In vitro and in vivo studies evaluating biocompatibility and bioactivity have demonstrated the potential of these hydrogels for promoting cell adhesion, proliferation, and differentiation. Their applications in bone defect repair, osteochondral tissue engineering and drug delivery are also explored. Despite their potential in bone tissue engineering, nanoclay-composite hydrogels face challenges such as optimal dispersion, scalability, biocompatibility, long-term stability, regulatory approval, and integration with emerging technologies to achieve clinical application. Future research directions need to focus on refining fabrication techniques, enhancing understanding of biological interactions, and advancing towards clinical translation and commercialization. Overall, nanoclay-composite hydrogels offer exciting opportunities for improving bone regeneration strategies.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
9
|
Zhu Y, Yu X, Liu H, Li J, Gholipourmalekabadi M, Lin K, Yuan C, Wang P. Strategies of functionalized GelMA-based bioinks for bone regeneration: Recent advances and future perspectives. Bioact Mater 2024; 38:346-373. [PMID: 38764449 PMCID: PMC11101688 DOI: 10.1016/j.bioactmat.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/07/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024] Open
Abstract
Gelatin methacryloyl (GelMA) hydrogels is a widely used bioink because of its good biological properties and tunable physicochemical properties, which has been widely used in a variety of tissue engineering and tissue regeneration. However, pure GelMA is limited by the weak mechanical strength and the lack of continuous osteogenic induction environment, which is difficult to meet the needs of bone repair. Moreover, GelMA hydrogels are unable to respond to complex stimuli and therefore are unable to adapt to physiological and pathological microenvironments. This review focused on the functionalization strategies of GelMA hydrogel based bioinks for bone regeneration. The synthesis process of GelMA hydrogel was described in details, and various functional methods to meet the requirements of bone regeneration, including mechanical strength, porosity, vascularization, osteogenic differentiation, and immunoregulation for patient specific repair, etc. In addition, the response strategies of smart GelMA-based bioinks to external physical stimulation and internal pathological microenvironment stimulation, as well as the functionalization strategies of GelMA hydrogel to achieve both disease treatment and bone regeneration in the presence of various common diseases (such as inflammation, infection, tumor) are also briefly reviewed. Finally, we emphasized the current challenges and possible exploration directions of GelMA-based bioinks for bone regeneration.
Collapse
Affiliation(s)
- Yaru Zhu
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
- Quanzhou Women's and Children's Hospital, Quanzhou, China
| | - Xingge Yu
- Department of Oral and Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Hao Liu
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Junjun Li
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Department of Medical Biotechnology, Faculty of Allied Medicine, Tehran, Iran
| | - Kaili Lin
- Department of Oral and Cranio-maxillofacial Science, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Penglai Wang
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
10
|
He Y, Zhang Y, Hu F, Chen M, Wang B, Li Y, Xu H, Dong N, Zhang C, Hu Y, Lin Z, Peng Y, Ye Q, Luo L. Photosensitive Hydrogels Encapsulating DPSCs and AgNPs for Dental Pulp Regeneration. Int Dent J 2024; 74:836-846. [PMID: 38369441 PMCID: PMC11287144 DOI: 10.1016/j.identj.2024.01.017] [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: 11/29/2023] [Revised: 01/12/2024] [Accepted: 01/23/2024] [Indexed: 02/20/2024] Open
Abstract
OBJECTIVE Pulp regeneration with bioactive dentin-pulp complex has been a research hotspot in recent years. Stem cell therapy provided an interest strategy to regenerate the dental-pulp complex. Hence, this study aimed to evaluate the effects of photosensitive gelatin methacrylate (GelMA) hydrogel encapsulating dental pulp stem cells (DPSCs) and silver nanoparticles (AgNPs) for dental pulp regeneration in vitro. METHODS First, the AgNPs@GelMA hydrogels were prepared by lithium phenyl-2,4,6-trimethyl-benzoyl phosphinate (LAP) initiation via blue-light emitting diode light. The physical and chemical properties of AgNPs@GelMA hydrogels were comprehensively analysed via scanning electron microscopy (SEM), and mechanical characterisation, such as swelling ability, degradation properties, and AgNP release profile. Then, AgNPs@GelMA hydrogels encapsulated DPSCs were used to establish an AgNPs@GelMA biomimetic complex, further analysing its biocompatibility, antibacterial properties, and angiogenic capacity in vitro. RESULTS The results indicated that GelMA hydrogels demontrated optimal characteristics with a monomer:LAP ratio of 16:1. The physico-chemical properties of AgNPs@GelMA hydrogels did not change significantly after loading with AgNPs. There was no significant difference in AgNP release rate amongst different concentrations of AgNPs@GelMA hydrogels. Fifty to 200 μg/mL AgNPs@GelMA hydrogels could disperse E faecalis biofilm and reduce its metabolic activity . Furthermore, cell proliferation was arrested in 100 and 200 μg/mL AgNPs@GelMA hydrogels. The inhibition of 50 μg/mL AgNPs@GelMA hydrogels on E faecalis biofilm was above 50%, and the cell viability of the hydrogels was higher than 90%. The angiogenesis assay indicated that AgNPs@GelMA hydrogels encapsulating DPSCs could induce the formation of capillary-like structures and express angiogenic markers CD31, vascular endothelial growth factor , and von willebrand factor (vWF) in vitro. CONCLUSIONS Results of this study indicate that 50 μg/mL AgNPs@GelMA hydrogels encapsulating DPSCs had significant antibacterial properties and angiogenic capacity, which could provide a significant experimental basis for the regeneration of the dentin-pulp complex.
Collapse
Affiliation(s)
- Yan He
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China; Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Yanni Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fengting Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Chen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ben Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yejian Li
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haichao Xu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Na Dong
- Oujiang Laboratory, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chen Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunfan Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiqiang Lin
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Youjian Peng
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingsong Ye
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China; Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Lihua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
11
|
Liu X, Liu C, Lin Q, Shi T, Liu G. Exosome-loaded hydrogels for craniofacial bone tissue regeneration. Biomed Mater 2024; 19:052002. [PMID: 38815606 DOI: 10.1088/1748-605x/ad525c] [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: 01/05/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
It is common for maladies and trauma to cause significant bone deterioration in the craniofacial bone, which can cause patients to experience complications with their appearance and their ability to function. Regarding grafting procedures' complications and disadvantages, the newly emerging field of tissue regeneration has shown promise. Tissue -engineered technologies and their applications in the craniofacial region are increasingly gaining prominence with limited postoperative risk and cost. MSCs-derived exosomes are widely applied in bone tissue engineering to provide cell-free therapies since they not only do not cause immunological rejection in the same way that cells do, but they can also perform a cell-like role. Additionally, the hydrogel system is a family of multipurpose platforms made of cross-linked polymers with considerable water content, outstanding biocompatibility, and tunable physiochemical properties for the efficient delivery of commodities. Therefore, the promising exosome-loaded hydrogels can be designed for craniofacial bone regeneration. This review lists the packaging techniques for exosomes and hydrogel and discusses the development of a biocompatible hydrogel system and its potential for exosome continuous delivery for craniofacial bone healing.
Collapse
Affiliation(s)
- Xiaojie Liu
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Chang Liu
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Qingquan Lin
- Institute of Applied Catalysis, College of Chemistry and Chemical Engineering, Yantai University, Yantai, People's Republic of China
| | - Ting Shi
- Department of Plastic Surgery, Yantaishan Hospital, Yantai, People's Republic of China
| | - Guanying Liu
- Department of Hand and Foot Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, People's Republic of China
| |
Collapse
|
12
|
Sun DS, Chang HH. Extracellular vesicles: Function, resilience, biomarker, bioengineering, and clinical implications. Tzu Chi Med J 2024; 36:251-259. [PMID: 38993825 PMCID: PMC11236075 DOI: 10.4103/tcmj.tcmj_28_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 07/13/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as key players in intercellular communication, disease pathology, and therapeutic innovation. Initially overlooked as cellular debris, EVs are now recognized as vital mediators of cell-to-cell communication, ferrying a cargo of proteins, nucleic acids, and lipids, providing cellular resilience in response to stresses. This review provides a comprehensive overview of EVs, focusing on their role as biomarkers in disease diagnosis, their functional significance in physiological and pathological processes, and the potential of bioengineering for therapeutic applications. EVs offer a promising avenue for noninvasive disease diagnosis and monitoring, reflecting the physiological state of originating cells. Their diagnostic potential spans a spectrum of diseases, including cancer, cardiovascular disorders, neurodegenerative diseases, and infectious diseases. Moreover, their presence in bodily fluids such as blood, urine, and cerebrospinal fluid enhances their diagnostic utility, presenting advantages over traditional methods. Beyond diagnostics, EVs mediate crucial roles in intercellular communication, facilitating the transfer of bioactive molecules between cells. This communication modulates various physiological processes such as tissue regeneration, immune modulation, and neuronal communication. Dysregulation of EV-mediated communication is implicated in diseases such as cancer, immune disorders, and neurodegenerative diseases, highlighting their therapeutic potential. Bioengineering techniques offer avenues for manipulating EVs for therapeutic applications, from isolation and purification to engineering cargo and targeted delivery systems. These approaches hold promise for developing novel therapeutics tailored to specific diseases, revolutionizing personalized medicine. However, challenges such as standardization, scalability, and regulatory approval need addressing for successful clinical translation. Overall, EVs represent a dynamic frontier in biomedical research with vast potential for diagnostics, therapeutics, and personalized medicine.
Collapse
Affiliation(s)
- Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
13
|
Mohammad Mehdipour N, Rajeev A, Kumar H, Kim K, Shor RJ, Natale G. Anisotropic hydrogel scaffold by flow-induced stereolithography 3D printing technique. BIOMATERIALS ADVANCES 2024; 161:213885. [PMID: 38743993 DOI: 10.1016/j.bioadv.2024.213885] [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: 02/05/2024] [Revised: 04/07/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024]
Abstract
Essential organs, such as the heart and liver, contain a unique porous network that allows oxygen and nutrients to be exchanged, with distinct random to ordered regions displaying varying degrees of strength. A novel technique, referred to here as flow-induced lithography, was developed. This technique generates tunable anisotropic three-dimensional (3D) structures. The ink for this bioprinting technique was made of titanium dioxide nanorods (Ti) and kaolinite nanoclay (KLT) dispersed in a GelMA/PEGDA polymeric suspension. By controlling the flow rate, aligned particle microstructures were achieved in the suspensions. The application of UV light to trigger the polymerization of the photoactive prepolymer freezes the oriented particles in the polymer network. Because the viability test was successful in shearing suspensions containing cells, the flow-induced lithography technique can be used with both acellular scaffolds and cell-laden structures. Fabricated hydrogels show outstanding mechanical properties resembling human tissues, as well as significant cell viability (> 95 %) over one week. As a result of this technique and the introduction of bio-ink, a novel approach has been pioneered for developing anisotropic tissue implants utilizing low-viscosity biomaterials.
Collapse
Affiliation(s)
- Narges Mohammad Mehdipour
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Ashna Rajeev
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Hitendra Kumar
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada; Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453552, India
| | - Keekyoung Kim
- Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Roman J Shor
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Giovanniantonio Natale
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
14
|
Xu X, Li J, Lu Y, Shan Y, Shen Z, Sun F, Zhu J, Chen W, Shi H. Extracellular Vesicles in the Repair of Bone and Cartilage Injury: From Macro‐Delivery to Micro‐Modification. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202300428] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Indexed: 01/06/2025]
Abstract
AbstractExtracellular vesicles (EVs) are intermediaries in intercellular signal transmission and material exchange and have attracted significant attention from researchers in bone and cartilage repair. These nanoscale vesicles hold immense potential in facilitating bone and cartilage repair and regeneration by regulating the microenvironment at an injury site. However, their in vivo utilization is limited by their self‐clearance and random distribution. Therefore, various delivery platforms have been developed to improve EV targeting and retention rates in target organs while achieving a controlled release of EVs. Additionally, engineering modification of EVs has been proposed to effectively enhance EVs' intrinsic targeting and drug‐loading abilities and further improve their therapeutic effects on bone and cartilage injuries. This review aims to introduce the biogenesis of EVs and their regulatory mechanisms in the microenvironment of bone and cartilage injuries and comprehensively discuss the application of EV‐delivery platforms of different materials and various EV engineering modification methods in treating bone and cartilage injuries. The review's findings can help advance EV research and develop new strategies for improving the therapy of bone and cartilage injuries.
Collapse
Affiliation(s)
- Xiangyu Xu
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Jialu Li
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Yi Lu
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Yibo Shan
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Zhiming Shen
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Fei Sun
- Department of Thoracic Surgery Taizhou People's Hospital Affiliated to Nanjing Medical University Taizhou 225300 China
| | - Jianwei Zhu
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Wenxuan Chen
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| | - Hongcan Shi
- Clinical Medical College Yangzhou University Yangzhou 225001 China
- Institute of Translational Medicine Medical College Yangzhou University Yangzhou 225001 China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases Yangzhou University Yangzhou 225001 China
| |
Collapse
|
15
|
Gharibshahian M, Salehi M, Kamalabadi-Farahani M, Alizadeh M. Magnesium-oxide-enhanced bone regeneration: 3D-printing of gelatin-coated composite scaffolds with sustained Rosuvastatin release. Int J Biol Macromol 2024; 266:130995. [PMID: 38521323 DOI: 10.1016/j.ijbiomac.2024.130995] [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: 10/26/2023] [Revised: 03/11/2024] [Accepted: 03/17/2024] [Indexed: 03/25/2024]
Abstract
Critical-size bone defects are one of the main challenges in bone tissue regeneration that determines the need to use angiogenic and osteogenic agents. Rosuvastatin (RSV) is a class of cholesterol-lowering drugs with osteogenic potential. Magnesium oxide (MgO) is an angiogenesis component affecting apatite formation. This study aims to evaluate 3D-printed Polycaprolactone/β-tricalcium phosphate/nano-hydroxyapatite/ MgO (PCL/β-TCP/nHA/MgO) scaffolds as a carrier for MgO and RSV in bone regeneration. For this purpose, PCL/β-TCP/nHA/MgO scaffolds were fabricated with a 3D-printing method and coated with gelatin and RSV. The biocompatibility and osteogenicity of scaffolds were examined with MTT, ALP, and Alizarin red staining. Finally, the scaffolds were implanted in a bone defect of rat's calvaria, and tissue regeneration was investigated after 3 months. Our results showed that the simultaneous presence of RSV and MgO improved biocompatibility, wettability, degradation rate, and ALP activity but decreased mechanical strength. PCL/β-TCP/nHA/MgO/gelatin-RSV scaffolds produced sustained release of MgO and RSV within 30 days. CT images showed that PCL/β-TCP/nHA/MgO/gelatin-RSV scaffolds filled approximately 86.83 + 4.9 % of the defects within 3 months and improved angiogenesis, woven bone, and osteogenic genes expression. These results indicate the potential of PCL/β-TCP/nHA/MgO/gelatin-RSV scaffolds as a promising tool for bone regeneration and clinical trials.
Collapse
Affiliation(s)
- Maliheh Gharibshahian
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Majid Salehi
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Kamalabadi-Farahani
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
16
|
Mamidi N, Ijadi F, Norahan MH. Leveraging the Recent Advancements in GelMA Scaffolds for Bone Tissue Engineering: An Assessment of Challenges and Opportunities. Biomacromolecules 2024; 25:2075-2113. [PMID: 37406611 DOI: 10.1021/acs.biomac.3c00279] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
The field of bone tissue engineering has seen significant advancements in recent years. Each year, over two million bone transplants are performed globally, and conventional treatments, such as bone grafts and metallic implants, have their limitations. Tissue engineering offers a new level of treatment, allowing for the creation of living tissue within a biomaterial framework. Recent advances in biomaterials have provided innovative approaches to rebuilding bone tissue function after damage. Among them, gelatin methacryloyl (GelMA) hydrogel is emerging as a promising biomaterial for supporting cell proliferation and tissue regeneration, and GelMA has exhibited exceptional physicochemical and biological properties, making it a viable option for clinical translation. Various methods and classes of additives have been used in the application of GelMA for bone regeneration, with the incorporation of nanofillers or other polymers enhancing its resilience and functional performance. Despite promising results, the fabrication of complex structures that mimic the bone architecture and the provision of balanced physical properties for both cell and vasculature growth and proper stiffness for load bearing remain as challenges. In terms of utilizing osteogenic additives, the priority should be on versatile components that promote angiogenesis and osteogenesis while reinforcing the structure for bone tissue engineering applications. This review focuses on recent efforts and advantages of GelMA-based composite biomaterials for bone tissue engineering, covering the literature from the last five years.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Fatemeh Ijadi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| | - Mohammad Hadi Norahan
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| |
Collapse
|
17
|
Hashemi A, Ezati M, Nasr MP, Zumberg I, Provaznik V. Extracellular Vesicles and Hydrogels: An Innovative Approach to Tissue Regeneration. ACS OMEGA 2024; 9:6184-6218. [PMID: 38371801 PMCID: PMC10870307 DOI: 10.1021/acsomega.3c08280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/27/2023] [Accepted: 12/19/2023] [Indexed: 02/20/2024]
Abstract
Extracellular vesicles have emerged as promising tools in regenerative medicine due to their inherent ability to facilitate intercellular communication and modulate cellular functions. These nanosized vesicles transport bioactive molecules, such as proteins, lipids, and nucleic acids, which can affect the behavior of recipient cells and promote tissue regeneration. However, the therapeutic application of these vesicles is frequently constrained by their rapid clearance from the body and inability to maintain a sustained presence at the injury site. In order to overcome these obstacles, hydrogels have been used as extracellular vesicle delivery vehicles, providing a localized and controlled release system that improves their therapeutic efficacy. This Review will examine the role of extracellular vesicle-loaded hydrogels in tissue regeneration, discussing potential applications, current challenges, and future directions. We will investigate the origins, composition, and characterization techniques of extracellular vesicles, focusing on recent advances in exosome profiling and the role of machine learning in this field. In addition, we will investigate the properties of hydrogels that make them ideal extracellular vesicle carriers. Recent studies utilizing this combination for tissue regeneration will be highlighted, providing a comprehensive overview of the current research landscape and potential future directions.
Collapse
Affiliation(s)
- Amir Hashemi
- Department
of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3082/12, 61600 Brno, Czech Republic
| | - Masoumeh Ezati
- Department
of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3082/12, 61600 Brno, Czech Republic
| | - Minoo Partovi Nasr
- Department
of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3082/12, 61600 Brno, Czech Republic
| | - Inna Zumberg
- Department
of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3082/12, 61600 Brno, Czech Republic
| | - Valentine Provaznik
- Department
of Biomedical Engineering, Faculty of Electrical Engineering and Communication, Brno University of Technology, Technicka 3082/12, 61600 Brno, Czech Republic
| |
Collapse
|
18
|
Rodrigo MJ, Cardiel MJ, Fraile JM, Mayoral JA, Pablo LE, Garcia-Martin E. Laponite for biomedical applications: An ophthalmological perspective. Mater Today Bio 2024; 24:100935. [PMID: 38239894 PMCID: PMC10794930 DOI: 10.1016/j.mtbio.2023.100935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024] Open
Abstract
Clay minerals have been applied in biomedicine for thousands of years. Laponite is a nanostructured synthetic clay with the capacity to retain and progressively release drugs. In recent years there has been a resurgence of interest in Laponite application in various biomedical areas. This is the first paper to review the potential biomedical applications of Laponite in ophthalmology. The introduction briefly covers the physical, chemical, rheological, and biocompatibility features of different routes of administration. After that, emphasis is placed on 1) drug delivery for antibiotics, anti-inflammatories, growth factors, other proteins, and cancer treatment; 2) bleeding prevention or treatment; and 3) tissue engineering through regenerative medicine using scaffolds in intraocular and extraocular tissue. Although most scientific research is not performed on the eye, both the findings and the new treatments resulting from that research are potentially applicable in ophthalmology since many of the drugs used are the same, the tissue evaluated in vitro or in vivo is also present in the eye, and the pathologies treated also occur in the eye. Finally, future prospects for this emerging field are discussed.
Collapse
Affiliation(s)
- Maria J. Rodrigo
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
| | - Maria J. Cardiel
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
- Department of Pathology, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Jose M. Fraile
- Institute for Chemical Synthesis and Homogeneous Catalysis (ISQCH), Faculty of Sciences, University of Zaragoza–CSIC, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Jose A. Mayoral
- Institute for Chemical Synthesis and Homogeneous Catalysis (ISQCH), Faculty of Sciences, University of Zaragoza–CSIC, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Luis E. Pablo
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
- Biotech Vision SLP (spin-off Company), University of Zaragoza, Spain
| | - Elena Garcia-Martin
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
| |
Collapse
|
19
|
Shi H, Yang Y, Xing H, Jia J, Xiong W, Guo S, Yang S. Exosomal non-coding RNAs: Emerging insights into therapeutic potential and mechanisms in bone healing. J Tissue Eng 2024; 15:20417314241286606. [PMID: 39371940 PMCID: PMC11456177 DOI: 10.1177/20417314241286606] [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: 08/11/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Exosomes are nano-sized extracellular vesicles (EVs) released by diverse types of cells, which affect the functions of targeted cells by transporting bioactive substances. As the main component of exosomes, non-coding RNA (ncRNA) is demonstrated to impact multiple pathways participating in bone healing. Herein, this review first introduces the biogenesis and secretion of exosomes, and elucidates the role of the main cargo in exosomes, ncRNAs, in mediating intercellular communication. Subsequently, the potential molecular mechanism of exosomes accelerating bone healing is elucidated from the following four aspects: macrophage polarization, vascularization, osteogenesis and osteoclastogenesis. Then, we systematically introduce construction strategies based on modified exosomes in bone regeneration field. Finally, the clinical trials of exosomes for bone healing and the challenges of exosome-based therapies in the biomedical field are briefly introduced, providing solid theoretical frameworks and optimization methods for the clinical application of exosomes in orthopedics.
Collapse
Affiliation(s)
- Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yang Yang
- Department of Rehabilitation, The First Hospital of China Medical University, Shenyang, China
| | - Hao Xing
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jialin Jia
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Li Z, Zhang B, Shang J, Wang Y, Jia L, She X, Xu X, Zhang D, Guo J, Zhang F. Diabetic and nondiabetic BMSC-derived exosomes affect bone regeneration via regulating miR-17-5p/SMAD7 axis. Int Immunopharmacol 2023; 125:111190. [PMID: 37976598 DOI: 10.1016/j.intimp.2023.111190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Diabetic bone disease (DBD) is a complication of diabetes mellitus (DM) and is characterized by impaired osteocyte function and delayed bone remodeling due to high blood glucose levels and sustained release of inflammatory factors. Recent studies show that the regulation of osteoblasts (OBs) by bone marrow stromal cells (BMSCs) is an important mechanism in alleviating DBD and that exosomes are recognized as the key medium. Mesenchymal stem cell-derived exosome (MSC-Exos) therapy is a promising approach to facilitate tissue repair. However, the influence of exosomes from diabetic conditioned BMSCs on OBs and bone regeneration, as well as the underlying mechanism, are still elusive. Here, we used high-glucose medium to mimic diabetic conditions and normal-glucose medium as control to mimic nondiabetic conditions in vitro and found that microRNA-17 (miR-17) was downregulated in diabetic-conditioned BMSC-derived exosomes (HG-Exos), HG-Exo-co-cultured osteoblasts, and the skull of rats with type 2 diabetes mellitus (T2DM). Further experiment concluded that nondiabetic conditioned BMSC-Exos (NG-Exos) promoted the osteogenesis of OBs and bone regeneration of rats with T2DM via upregulation of miR-17. Compare with NG-Exos, HG-Exos impeded osteogenesis of OBs in vitro and bone regeneration of rats in vivo by downregulation of miR-17. Moreover, miR-17 promoted bone regeneration by targeting SMAD7, which was further proved to have a negative effect on osteogenesis. Taken together, nondiabetic BMSC-derived exosomes greatly foster bone regeneration, whereas diabetic BMSC-derived exosomes undermine the promotion effect of MSC-Exos by regulating the miR-17/SMAD7 axis. These findings provide support for the miR-17-5p/SMAD7 axis as a promising therapeutic target to treat DBD.
Collapse
Affiliation(s)
- Zechuan Li
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Bing Zhang
- Department of Health Care, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Jiaming Shang
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Yanan Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Linglu Jia
- Department of Prosthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Xiao She
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China
| | - Jing Guo
- Engineering Laboratory for Biomaterials and Tissue Regeneration, Ningbo Stomatology Hospital, No. 435 Xinxing Road, Ningbo 315042, Zhejiang, China; Savaid Stomatology School of Hangzhou Medical College, No. 1 Pinghai Road, Hangzhou 310051, Zhejiang, China
| | - Fan Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, 250012 Jinan, Shandong, China.
| |
Collapse
|
21
|
Simon L, Lapinte V, Morille M. Exploring the role of polymers to overcome ongoing challenges in the field of extracellular vesicles. J Extracell Vesicles 2023; 12:e12386. [PMID: 38050832 PMCID: PMC10696644 DOI: 10.1002/jev2.12386] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/03/2023] [Accepted: 11/09/2023] [Indexed: 12/07/2023] Open
Abstract
Extracellular vesicles (EVs) are naturally occurring nanoparticles released from all eucaryotic and procaryotic cells. While their role was formerly largely underestimated, EVs are now clearly established as key mediators of intercellular communication. Therefore, these vesicles constitute an attractive topic of study for both basic and applied research with great potential, for example, as a new class of biomarkers, as cell-free therapeutics or as drug delivery systems. However, the complexity and biological origin of EVs sometimes complicate their identification and therapeutic use. Thus, this rapidly expanding research field requires new methods and tools for the production, enrichment, detection, and therapeutic application of EVs. In this review, we have sought to explain how polymer materials actively contributed to overcome some of the limitations associated to EVs. Indeed, thanks to their infinite diversity of composition and properties, polymers can act through a variety of strategies and at different stages of EVs development. Overall, we would like to emphasize the importance of multidisciplinary research involving polymers to address persistent limitations in the field of EVs.
Collapse
Affiliation(s)
| | | | - Marie Morille
- ICGM, Univ Montpellier, CNRS, ENSCMMontpellierFrance
- Institut universitaire de France (IUF)ParisFrance
| |
Collapse
|
22
|
Li W, Hu J, Chen C, Li X, Zhang H, Xin Y, Tian Q, Wang S. Emerging advances in hydrogel-based therapeutic strategies for tissue regeneration. Regen Ther 2023; 24:459-471. [PMID: 37772128 PMCID: PMC10523184 DOI: 10.1016/j.reth.2023.09.007] [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: 04/17/2023] [Revised: 08/14/2023] [Accepted: 09/07/2023] [Indexed: 09/30/2023] Open
Abstract
Significant developments in cell therapy and biomaterial science have broadened the therapeutic landscape of tissue regeneration. Tissue damage is a complex biological process in which different types of cells play a specific role in repairing damaged tissues and growth factors strictly regulate the activity of these cells. Hydrogels have become promising biomaterials for tissue regeneration if appropriate materials are selected and the hydrogel properties are well-regulated. Importantly, they can be used as carriers for living cells and growth factors due to the high water-holding capacity, high permeability, and good biocompatibility of hydrogels. Cell-loaded hydrogels can play an essential role in treating damaged tissues and open new avenues for cell therapy. There is ample evidence substantiating the ability of hydrogels to facilitate the delivery of cells (stem cell, macrophage, chondrocyte, and osteoblast) and growth factors (bone morphogenetic protein, transforming growth factor, vascular endothelial growth factor and fibroblast growth factor). This paper reviewed the latest advances in hydrogels loaded with cells or growth factors to promote the reconstruction of tissues. Furthermore, we discussed the shortcomings of the application of hydrogels in tissue engineering to promote their further development.
Collapse
Affiliation(s)
- Wenqi Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jing Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Cheng Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xinyue Li
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Honghua Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yanru Xin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qingchang Tian
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shuling Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
23
|
Cedillo-Servin G, Louro AF, Gamelas B, Meliciano A, Zijl A, Alves PM, Malda J, Serra M, Castilho M. Microfiber-reinforced hydrogels prolong the release of human induced pluripotent stem cell-derived extracellular vesicles to promote endothelial migration. BIOMATERIALS ADVANCES 2023; 155:213692. [PMID: 37952463 DOI: 10.1016/j.bioadv.2023.213692] [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: 02/28/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
Extracellular vesicle (EV)-based approaches for promoting angiogenesis have shown promising results. Yet, further development is needed in vehicles that prolong EV exposure to target organs. Here, we hypothesized that microfiber-reinforced gelatin methacryloyl (GelMA) hydrogels could serve as sustained delivery platforms for human induced pluripotent stem cell (hiPSC)-derived EV. EV with 50-200 nm size and typical morphology were isolated from hiPSC-conditioned culture media and tested negative for common co-isolated contaminants. hiPSC-EV were then incorporated into GelMA hydrogels with or without a melt electrowritten reinforcing mesh. EV release was found to increase with GelMA concentration, as 12 % (w/v) GelMA hydrogels provided higher release rate and total release over 14 days in vitro, compared to lower hydrogel concentrations. Release profile modelling identified diffusion as a predominant release mechanism based on a Peppas-Sahlin model. To study the effect of reinforcement-dependent hydrogel mechanics on EV release, stress relaxation was assessed. Reinforcement with highly porous microfiber meshes delayed EV release by prolonging hydrogel stress relaxation and reducing the swelling ratio, thus decreasing the initial burst and overall extent of release. After release from photocrosslinked reinforced hydrogels, EV remained internalizable by human umbilical vein endothelial cells (HUVEC) over 14 days, and increased migration was observed in the first 4 h. EV and RNA cargo stability was investigated at physiological temperature in vitro, showing a sharp decrease in total RNA levels, but a stable level of endothelial migration-associated small noncoding RNAs over 14 days. Our data show that hydrogel formulation and microfiber reinforcement are superimposable approaches to modulate EV release from hydrogels, thus depicting fiber-reinforced GelMA hydrogels as tunable hiPSC-EV vehicles for controlled release systems that promote endothelial cell migration.
Collapse
Affiliation(s)
- Gerardo Cedillo-Servin
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ana Filipa Louro
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Beatriz Gamelas
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana Meliciano
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Anne Zijl
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands; Faculty of Medicine, Utrecht University, Utrecht, the Netherlands
| | - Paula M Alves
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Jos Malda
- Regenerative Medicine Centre Utrecht, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Margarida Serra
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
24
|
Harun-Or-Rashid M, Aktar MN, Hossain MS, Sarkar N, Islam MR, Arafat ME, Bhowmik S, Yusa SI. Recent Advances in Micro- and Nano-Drug Delivery Systems Based on Natural and Synthetic Biomaterials. Polymers (Basel) 2023; 15:4563. [PMID: 38231996 PMCID: PMC10708661 DOI: 10.3390/polym15234563] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 01/19/2024] Open
Abstract
Polymeric drug delivery technology, which allows for medicinal ingredients to enter a cell more easily, has advanced considerably in recent decades. Innovative medication delivery strategies use biodegradable and bio-reducible polymers, and progress in the field has been accelerated by future possible research applications. Natural polymers utilized in polymeric drug delivery systems include arginine, chitosan, dextrin, polysaccharides, poly(glycolic acid), poly(lactic acid), and hyaluronic acid. Additionally, poly(2-hydroxyethyl methacrylate), poly(N-isopropyl acrylamide), poly(ethylenimine), dendritic polymers, biodegradable polymers, and bioabsorbable polymers as well as biomimetic and bio-related polymeric systems and drug-free macromolecular therapies have been employed in polymeric drug delivery. Different synthetic and natural biomaterials are in the clinical phase to mitigate different diseases. Drug delivery methods using natural and synthetic polymers are becoming increasingly common in the pharmaceutical industry, with biocompatible and bio-related copolymers and dendrimers having helped cure cancer as drug delivery systems. This review discusses all the above components and how, by combining synthetic and biological approaches, micro- and nano-drug delivery systems can result in revolutionary polymeric drug and gene delivery devices.
Collapse
Affiliation(s)
- Md. Harun-Or-Rashid
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| | - Most. Nazmin Aktar
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| | - Md. Sabbir Hossain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Nadia Sarkar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Md. Easin Arafat
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.S.H.); (N.S.); (M.R.I.); (M.E.A.)
| | - Shukanta Bhowmik
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| | - Shin-ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji 671-2280, Hyogo, Japan; (M.H.-O.-R.); (M.N.A.); (S.B.)
| |
Collapse
|
25
|
Zhang C, Zhou X, Wang D, Hao L, Zeng Z, Su L. Hydrogel-Loaded Exosomes: A Promising Therapeutic Strategy for Musculoskeletal Disorders. J Clin Pharm Ther 2023; 2023:1-36. [DOI: 10.1155/2023/1105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Clinical treatment strategies for musculoskeletal disorders have been a hot research topic. Accumulating evidence suggests that hydrogels loaded with MSC-derived EVs show great potential in improving musculoskeletal injuries. The ideal hydrogels should be capable of promoting the development of new tissues and simulating the characteristics of target tissues, with the properties matching the cell-matrix constituents of autologous tissues. Although there have been numerous reports of hydrogels loaded with MSC-derived EVs for the repair of musculoskeletal injuries, such as intervertebral disc injury, tendinopathy, bone fractures, and cartilage injuries, there are still many hurdles to overcome before the clinical application of modified hydrogels. In this review, we focus on the advantages of the isolation technique of EVs in combination with different types of hydrogels. In this context, the efficacy of hydrogels loaded with MSC-derived EVs in different musculoskeletal injuries is discussed in detail to provide a reference for the future application of hydrogels loaded with MSC-derived EVs in the clinical treatment of musculoskeletal injuries.
Collapse
Affiliation(s)
- Chunyu Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Li Hao
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Lei Su
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| |
Collapse
|
26
|
Ma C, Kim YK, Lee MH, Jang YS. Development of Gelatin Methacryloyl/Sodium Alginate Interpenetrating Polymer Network Hydrogels for Bone Regeneration by Activating the Wnt/β-Catenin Signaling Pathway via Lithium Release. Int J Mol Sci 2023; 24:13613. [PMID: 37686419 PMCID: PMC10487821 DOI: 10.3390/ijms241713613] [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: 07/13/2023] [Revised: 08/07/2023] [Accepted: 08/12/2023] [Indexed: 09/10/2023] Open
Abstract
Hydrogels have gained significant attention as biomaterials due to their remarkable properties resembling those of the extracellular matrix (ECM). In the present investigation, we successfully synthesized interpenetrating polymer network (IPN) hydrogels using gelatin methacryloyl (GelMA) and sodium alginate (SA), incorporating various concentrations of lithium chloride (LiCl; 0, 5, and 10 mM), aiming to develop a hydrogel scaffold for bone regeneration. Notably, the compressive modulus of the IPN hydrogels remained largely unaffected upon the inclusion of LiCl. However, the hydrogel with the high concentration of LiCl exhibited reduced fragmentation after compression testing. Intriguingly, we observed a significant improvement in cellular biocompatibility, primarily attributed to activation of the Wnt/β-catenin signaling pathway induced by LiCl. Subsequently, we evaluated the efficacy of the newly developed IPN-Li hydrogels in a rat cranial defect model and found that they substantially enhanced bone regeneration. Nevertheless, it is important to note that the introduction of high concentrations of LiCl did not significantly promote osteogenesis. This outcome can be attributed to the excessive release of Li+ ions into the extracellular matrix, hindering the desired effect. Overall, the IPN-Li hydrogel developed in this study holds great promise as a biodegradable material for bone regeneration applications.
Collapse
Affiliation(s)
| | | | | | - Yong-Seok Jang
- Department of Dental Biomaterials, Institute of Biodegradable Materials, School of Dentistry, Jeonbuk National University, Jeon-Ju 54896, Republic of Korea; (C.M.); (Y.-K.K.); (M.-H.L.)
| |
Collapse
|
27
|
Wei J, Ou Z, Tong B, Liao Z, Yang C. Engineered extracellular vesicles as therapeutics of degenerative orthopedic diseases. Front Bioeng Biotechnol 2023; 11:1162263. [PMID: 37362216 PMCID: PMC10289007 DOI: 10.3389/fbioe.2023.1162263] [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: 02/09/2023] [Accepted: 05/19/2023] [Indexed: 06/28/2023] Open
Abstract
Degenerative orthopedic diseases, as a global public health problem, have made serious negative impact on patients' quality of life and socio-economic burden. Traditional treatments, including chemical drugs and surgical treatments, have obvious side effects and unsatisfactory efficacy. Therefore, biological therapy has become the focus of researches on degenerative orthopedic diseases. Extracellular vesicles (EVs), with superior properties of immunoregulatory, growth support, and drug delivery capabilities, have emerged as a new cell-free strategy for the treatment of many diseases, including degenerative orthopedic diseases. An increasing number of studies have shown that EVs can be engineered through cargo loading, surface modification, and chemical synthesis to improve efficiency, specificity, and safety. Herein, a comprehensive overview of recent advances in engineering strategies and applications of engineered EVs as well as related researches in degenerative orthopedic diseases, including osteoarthritis (OA), osteoporosis (OP), intervertebral disc degeneration (IDD) and osteonecrosis of the femoral head (ONFH), is provided. In addition, we analyze the potential and challenges of applying engineered EVs to clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Cao Yang
- *Correspondence: Zhiwei Liao, ; Cao Yang,
| |
Collapse
|
28
|
Davies OG. Extracellular vesicles: From bone development to regenerative orthopedics. Mol Ther 2023; 31:1251-1274. [PMID: 36869588 PMCID: PMC10188641 DOI: 10.1016/j.ymthe.2023.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Regenerative medicine aims to promote the replacement of tissues lost to damage or disease. While positive outcomes have been observed experimentally, challenges remain in their clinical translation. This has led to growing interest in applying extracellular vesicles (EVs) to augment or even replace existing approaches. Through the engineering of culture environments or direct/indirect manipulation of EVs themselves, multiple avenues have emerged to modulate EV production, targeting, and therapeutic potency. Drives to modulate release using material systems or functionalize implants for improved osseointegration have also led to outcomes that could have real-world impact. The purpose of this review is to highlight advantages in applying EVs for the treatment of skeletal defects, outlining the current state of the art in the field and emphasizing avenues for further investigation. Notably, the review identifies inconsistencies in EV nomenclature and outstanding challenges in defining a reproducible therapeutic dose. Challenges also remain in the scalable manufacture of a therapeutically potent and pure EV product, with a need to address scalable cell sources and optimal culture environments. Addressing these issues will be critical if we are to develop regenerative EV therapies that meet the demands of regulators and can be translated from bench to bedside.
Collapse
Affiliation(s)
- Owen G Davies
- School of Sport, Exercise, and Health Sciences, Loughborough University, Epinal Way, Loughborough, Leicestershire LE11 3TU, UK.
| |
Collapse
|
29
|
Man K, Eisenstein NM, Hoey DA, Cox SC. Bioengineering extracellular vesicles: smart nanomaterials for bone regeneration. J Nanobiotechnology 2023; 21:137. [PMID: 37106449 PMCID: PMC10134574 DOI: 10.1186/s12951-023-01895-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
In the past decade, extracellular vesicles (EVs) have emerged as key regulators of bone development, homeostasis and repair. EV-based therapies have the potential to circumnavigate key issues hindering the translation of cell-based therapies including functional tissue engraftment, uncontrolled differentiation and immunogenicity issues. Due to EVs' innate biocompatibility, low immunogenicity, and high physiochemical stability, these naturally-derived nanoparticles have garnered growing interest as potential acellular nanoscale therapeutics for a variety of diseases. Our increasing knowledge of the roles these cell-derived nanoparticles play, has made them an exciting focus in the development of novel pro-regenerative therapies for bone repair. Although these nano-sized vesicles have shown promise, their clinical translation is hindered due to several challenges in the EV supply chain, ultimately impacting therapeutic efficacy and yield. From the biochemical and biophysical stimulation of parental cells to the transition to scalable manufacture or maximising vesicles therapeutic response in vivo, a multitude of techniques have been employed to improve the clinical efficacy of EVs. This review explores state of the art bioengineering strategies to promote the therapeutic utility of vesicles beyond their native capacity, thus maximising the clinical potential of these pro-regenerative nanoscale therapeutics for bone repair.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Neil M Eisenstein
- Research and Clinical Innovation, Royal Centre for Defence Medicine, ICT Centre, Vincent Drive, Birmingham, B15 2SQ, UK
- Institute of Translational Medicine, University of Birmingham, Heritage Building, Mindelsohn Way, Birmingham, B15 2TH, UK
| | - David A Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, D02 R590, Ireland
- Dept. of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College, Dublin 2, D02 DK07, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2, D02 VN51, Dublin, Ireland
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
30
|
Man K, Brunet MY, Lees R, Peacock B, Cox SC. Epigenetic Reprogramming via Synergistic Hypomethylation and Hypoxia Enhances the Therapeutic Efficacy of Mesenchymal Stem Cell Extracellular Vesicles for Bone Repair. Int J Mol Sci 2023; 24:ijms24087564. [PMID: 37108726 PMCID: PMC10142722 DOI: 10.3390/ijms24087564] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a promising cell population for regenerative medicine applications, where paracrine signalling through the extracellular vesicles (EVs) regulates bone tissue homeostasis and development. MSCs are known to reside in low oxygen tension, which promotes osteogenic differentiation via hypoxia-inducible factor-1α activation. Epigenetic reprogramming has emerged as a promising bioengineering strategy to enhance MSC differentiation. Particularly, the process of hypomethylation may enhance osteogenesis through gene activation. Therefore, this study aimed to investigate the synergistic effects of inducing hypomethylation and hypoxia on improving the therapeutic efficacy of EVs derived from human bone marrow MSCs (hBMSCs). The effects of the hypoxia mimetic agent deferoxamine (DFO) and the DNA methyltransferase inhibitor 5-azacytidine (AZT) on hBMSC viability was assessed by quantifying the DNA content. The epigenetic functionality was evaluated by assessing histone acetylation and histone methylation. hBMSC mineralisation was determined by quantifying alkaline phosphate activity, collagen production and calcium deposition. EVs were procured from AZT, DFO or AZT/DFO-treated hBMSCs over a two-week period, with EV size and concentration defined using transmission electron microscopy, nanoflow cytometry and dynamic light scattering. The effects of AZT-EVs, DFO-EVs or AZT/DFO-EVs on the epigenetic functionality and mineralisation of hBMSCs were evaluated. Moreover, the effects of hBMSC-EVs on human umbilical cord vein endothelial cells (HUVECs) angiogenesis was assessed by quantifying pro-angiogenic cytokine release. DFO and AZT caused a time-dose dependent reduction in hBMSC viability. Pre-treatment with AZT, DFO or AZT/DFO augmented the epigenetic functionality of the MSCs through increases in histone acetylation and hypomethylation. AZT, DFO and AZT/DFO pre-treatment significantly enhanced extracellular matrix collagen production and mineralisation in hBMSCs. EVs derived from AZT/DFO-preconditioned hBMSCs (AZT/DFO-EVs) enhanced the hBMSC proliferation, histone acetylation and hypomethylation when compared to EVs derived from AZT-treated, DFO-treated and untreated hBMSCs. Importantly, AZT/DFO-EVs significantly increased osteogenic differentiation and mineralisation of a secondary hBMSC population. Furthermore, AZT/DFO-EVs enhanced the pro-angiogenic cytokine release of HUVECs. Taken together, our findings demonstrate the considerable utility of synergistically inducing hypomethylation and hypoxia to improve the therapeutic efficacy of the MSC-EVs as a cell-free approach for bone regeneration.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | - Mathieu Y Brunet
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK
| | | | | | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
31
|
Song X, Xu L, Zhang W. Biomimetic synthesis and optimization of extracellular vesicles for bone regeneration. J Control Release 2023; 355:18-41. [PMID: 36706840 DOI: 10.1016/j.jconrel.2023.01.057] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/29/2023]
Abstract
Critical-size bone defect repair is in high demand but is difficult to treat. Modern therapies, such as autograft and cell-based treatments, face limitations, including potential immunological rejection and tumorigenesis. Therefore, extracellular vesicle (EV)-based strategies have been proposed as a novel approach for tissue regeneration owing to EVs' complex composition of lipids, proteins, and nucleic acids, as well as their low immunogenicity and congenital cell-targeting features. Despite these remarkable features of EVs, biomimetic synthesis and optimization of natural EVs can lead to enhanced bioactivity, increased cellular uptake, and specific cell targeting, aiming to achieve optimal therapeutic efficacy. To maximize their function, these nanoparticles can be integrated into bone graft biomaterials for superior bone regeneration. Herein, we summarize the role of naturally occurring EVs from distinct cell types in bone regeneration, the current strategies for optimizing biomimetic synthetic EVs in bone regeneration, and discuss the recent advances in applying bone graft biomaterials for the delivery of EVs to bone defect repair. We focused on distinct strategies for optimizing EVs with different functions and the most recent research on achieving time-controlled release of nanoparticles from EV-loaded biomaterials. Furthermore, we thoroughly discuss several current challenges and proposed solutions, aiming to provide insight into current progress, inspiration for future development directions, and incentives for clinical application in this field.
Collapse
Affiliation(s)
- Xinyu Song
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China
| | - Ling Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai, China.
| |
Collapse
|
32
|
Beheshtizadeh N, Farzin A, Rezvantalab S, Pazhouhnia Z, Lotfibakhshaiesh N, Ai J, Noori A, Azami M. 3D printing of complicated GelMA-coated Alginate/Tri-calcium silicate scaffold for accelerated bone regeneration. Int J Biol Macromol 2023; 229:636-653. [PMID: 36586652 DOI: 10.1016/j.ijbiomac.2022.12.267] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Polymer-based composite scaffolds are an attractive class of biomaterials due to their suitable physical and mechanical performance as well as appropriate biological properties. When such composites contain osteoinductive ceramic nanopowders, it is possible, in principle, to stimulate the seeded cells to differentiate into osteoblasts. However, reproducibly fabricating and developing an appropriate niche for cells' activities in three-dimensional (3D) scaffolds remains a challenge using conventional fabrication techniques. Additive manufacturing provides a new strategy for the fabrication of complex 3D structures. Here, an extrusion-based 3D printing method was used to fabricate the Alginate (Alg)/Tri-calcium silicate (C3S) bone scaffolds. To improve physical and biological attributes, scaffolds were coated with gelatin methacryloyl (GelMA), a biocompatible viscose hydrogel. Conducting a combination of experimental techniques and molecular dynamics simulations, it is found that the composition ratio of Alg/C3S governs intermolecular interactions among the polymer and ceramic, affecting the product performance. Investigating the effects of various C3S amounts in the bioinks, the 90/10 composition ratio of Alg/C3S is known as the optimum content in developed bioinks. Accordingly, the printability of high-viscosity inks is boosted by improved hierarchical interactions among assemblies, which in turn leads to better nanoscale alignment in extruded macroscopic filaments. Conducting multiple tests on specimens, the GelMA-coated Alg/C3S scaffolds (with a composition ratio of 90/10) were shown to have improved mechanical qualities and cell adhesion, spreading, proliferation, and osteogenic differentiation, compared to the bare scaffolds, making them better candidates for further future research. Overall, the in-silico and in vitro studies of GelMA-coated 3D-printed Alg/C3S scaffolds open new aspects for biomaterials aimed at the regeneration of large- and complicated-bone defects through modifying the extrusion-based 3D-printed constructs.
Collapse
Affiliation(s)
- Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Ali Farzin
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sima Rezvantalab
- Renewable Energies Department, Faculty of Chemical Engineering, Urmia University of Technology, 57166-419 Urmia, Iran
| | - Zahra Pazhouhnia
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Noori
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Regenerative Medicine Group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Joint Reconstruction Research Center (JRRC), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Ju Y, Hu Y, Yang P, Xie X, Fang B. Extracellular vesicle-loaded hydrogels for tissue repair and regeneration. Mater Today Bio 2023; 18:100522. [PMID: 36593913 PMCID: PMC9803958 DOI: 10.1016/j.mtbio.2022.100522] [Citation(s) in RCA: 158] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/04/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Extracellular vesicles (EVs) are a collective term for nanoscale or microscale vesicles secreted by cells that play important biological roles. Mesenchymal stem cells are a class of cells with the potential for self-healing and multidirectional differentiation. In recent years, numerous studies have shown that EVs, especially those secreted by mesenchymal stem cells, can promote the repair and regeneration of various tissues and, thus, have significant potential in regenerative medicine. However, due to the rapid clearance capacity of the circulatory system, EVs are barely able to act persistently at specific sites for repair of target tissues. Hydrogels have good biocompatibility and loose and porous structural properties that allow them to serve as EV carriers, thereby prolonging the retention in certain specific areas and slowing the release of EVs. When EVs are needed to function at specific sites, the EV-loaded hydrogels can stand as an excellent approach. In this review, we first introduce the sources, roles, and extraction and characterization methods of EVs and describe their current application status. We then review the different types of hydrogels and discuss factors influencing their abilities to carry and release EVs. We summarize several strategies for loading EVs into hydrogels and characterizing EV-loaded hydrogels. Furthermore, we discuss application strategies for EV-loaded hydrogels and review their specific applications in tissue regeneration and repair. This article concludes with a summary of the current state of research on EV-loaded hydrogels and an outlook on future research directions, which we hope will provide promising ideas for researchers.
Collapse
Key Words
- 4-arm-PEG-MAL, four-armed polyethylene glycol (PEG) functionalized with maleimide group
- AD/CS/RSF, alginate-dopamine chondroitin sulfate and regenerated silk fibroin
- ADSC, Adipose derived mesenchymal stem cells
- ADSC-EVs, adipose mesenchymal stem cells derived EVs
- ADSC-Exos, adipose mesenchymal stem cells derived exosomes
- ATRP, Atom transfer radical polymerization
- BCA, bicinchoninic acid
- BMSC, Bone marrow mesenchymal stem cells
- BMSC-EVs, bone marrow mesenchymal stem cells derived EVs
- BMSC-Exos, bone marrow mesenchymal stem cells derived exosomes
- CGC, chitosan-gelatin-chondroitin sulfate
- CL, chitosan lactate
- CNS, central nervous system
- CPCs, cardiac progenitor cells
- CS-g-PEG, chitosan-g-PEG
- DPSC-Exos, dental pulp stem cells derived exosomes
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EVMs, extracellular vesicles mimetics
- EVs, Extracellular vesicles
- Exos, Exosomes
- Exosome
- Extracellular vesicle
- FEEs, functionally engineered EVs
- FGF, fibroblast growth factor
- GelMA, Gelatin methacryloyl
- HA, Hyaluronic acid
- HAMA, Hyaluronic acid methacryloyl
- HG, nano-hydroxyapatite-gelatin
- HIF-1 α, hypoxia-inducible factor-1 α
- HS-HA, hypoxia-sensitive hyaluronic acid
- HUVEC, human umbilical vein endothelial cell
- Hydrogel
- LAP, Lithium Phenyl (2,4,6-trimethylbenzoyl) phosphinate
- LSCM, laser scanning confocal microscopy
- MC-CHO, Aldehyde methylcellulose
- MMP, matrix metalloproteinase
- MNs, microneedles
- MSC-EVs, mesenchymal stem cells derived EVs
- MSC-Exos, mesenchymal stem cells derived exosomes
- MSCs, mesenchymal stem cells
- NPCs, neural progenitor cells
- NTA, nanoparticle tracking analysis
- OHA, oxidized hyaluronic acid
- OSA, oxidized sodium alginate
- PDA, Polydopamine
- PDLLA, poly(D l-lactic acid)
- PDNPs-PELA, Polydopamine nanoparticles incorporated poly (ethylene glycol)-poly(ε-cap-rolactone-co-lactide)
- PEG, Polyethylene glycol
- PF-127, Pluronic F-127
- PHEMA, phenoxyethyl methacrylate
- PIC, photo-induced imine crosslinking
- PKA, protein kinase A system
- PLA, Poly lactic acid
- PLGA, polylactic acid-hydroxy acetic acid copolymer
- PLLA, poly(l-lactic acid)
- PPy, polypyrrole
- PVA, polyvinyl alcohol
- RDRP, Reversible deactivation radical polymerization
- Regeneration
- SCI, spinal cord injury
- SEM, Scanning electron microscopy
- SF, Silk fibroin
- SPT, single-particle tracking
- TEM, transmission electron microscopy
- Tissue repair
- UMSC, umbilical cord mesenchymal stem cells
- UMSC-EVs, umbilical cord mesenchymal stem cells derived EVs
- UMSC-Exos, umbilical cord mesenchymal stem cells derived exosomes
- UV, ultraviolet
- VEGF, vascular endothelial growth factor
- VEGF-R, vascular endothelial growth factor receptor
- WB, western blotting
- dECM, decellularized ECM
- hiPS-MSC-Exos, human induced pluripotent stem cell-MSC-derived exosomes
- iPS-CPCs, pluripotent stem cell-derived cardiac progenitors
- nHP, nanohydroxyapatite/poly-ε-caprolactone
- sEVs, small extracellular vesicles
- β-TCP, β-Tricalcium Phosphate
Collapse
Affiliation(s)
- Yikun Ju
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Yue Hu
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, 637000, People's Republic of China
| | - Pu Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Xiaoyan Xie
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Bairong Fang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| |
Collapse
|
34
|
Shen N, Maggio M, Woods I, C. Lowry M, Almasri R, Gorgun C, Eichholz K, Stavenschi E, Hokamp K, Roche F, O’Driscoll L, Hoey D. Mechanically activated mesenchymal-derived bone cells drive vessel formation via an extracellular vesicle mediated mechanism. J Tissue Eng 2023; 14:20417314231186918. [PMID: 37654438 PMCID: PMC10467237 DOI: 10.1177/20417314231186918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/23/2023] [Indexed: 09/02/2023] Open
Abstract
Blood vessel formation is an important initial step for bone formation during development as well as during remodelling and repair in the adult skeleton. This results in a heavily vascularized tissue where endothelial cells and skeletal cells are constantly in crosstalk to facilitate homeostasis, a process that is mediated by numerous environmental signals, including mechanical loading. Breakdown in this communication can lead to disease and/or poor fracture repair. Therefore, this study aimed to determine the role of mature bone cells in regulating angiogenesis, how this is influenced by a dynamic mechanical environment, and understand the mechanism by which this could occur. Herein, we demonstrate that both osteoblasts and osteocytes coordinate endothelial cell proliferation, migration, and blood vessel formation via a mechanically dependent paracrine mechanism. Moreover, we identified that this process is mediated via the secretion of extracellular vesicles (EVs), as isolated EVs from mechanically stimulated bone cells elicited the same response as seen with the full secretome, while the EV-depleted secretome did not elicit any effect. Despite mechanically activated bone cell-derived EVs (MA-EVs) driving a similar response to VEGF treatment, MA-EVs contain minimal quantities of this angiogenic factor. Lastly, a miRNA screen identified mechanoresponsive miRNAs packaged within MA-EVs which are linked with angiogenesis. Taken together, this study has highlighted an important mechanism in osteogenic-angiogenic coupling in bone and has identified the mechanically activated bone cell-derived EVs as a therapeutic to promote angiogenesis and potentially bone repair.
Collapse
Affiliation(s)
- N. Shen
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - M. Maggio
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - I. Woods
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - M. C. Lowry
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, and Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - R. Almasri
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, and Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - C. Gorgun
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - K.F. Eichholz
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - E. Stavenschi
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - K. Hokamp
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, College Green, Dublin, Ireland
| | - F.M. Roche
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, College Green, Dublin, Ireland
| | - L. O’Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, and Trinity St. James’s Cancer Institute, Trinity College Dublin, Dublin, Ireland
| | - D.A. Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Department of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
35
|
Three-Dimensional Bio-Printed Cardiac Patch for Sustained Delivery of Extracellular Vesicles from the Interface. Gels 2022; 8:gels8120769. [PMID: 36547293 PMCID: PMC9777613 DOI: 10.3390/gels8120769] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiac tissue engineering has emerged as a promising strategy to treat infarcted cardiac tissues by replacing the injured region with an ex vivo fabricated functional cardiac patch. Nevertheless, integration of the transplanted patch with the host tissue is still a burden, limiting its clinical application. Here, a bi-functional, 3D bio-printed cardiac patch (CP) design is proposed, composed of a cell-laden compartment at its core and an extracellular vesicle (EV)-laden compartment at its shell for better integration of the CP with the host tissue. Alginate-based bioink solutions were developed for each compartment and characterized rheologically, examined for printability and their effect on residing cells or EVs. The resulting 3D bio-printed CP was examined for its mechanical stiffness, showing an elastic modulus between 4-5 kPa at day 1 post-printing, suitable for transplantation. Affinity binding of EVs to alginate sulfate (AlgS) was validated, exhibiting dissociation constant values similar to those of EVs with heparin. The incorporation of AlgS-EVs complexes within the shell bioink sustained EV release from the CP, with 88% cumulative release compared with 92% without AlgS by day 4. AlgS also prolonged the release profile by an additional 2 days, lasting 11 days overall. This CP design comprises great potential at promoting more efficient patch assimilation with the host.
Collapse
|
36
|
Pan Z, Sun W, Chen Y, Tang H, Lin W, Chen J, Chen C. Extracellular Vesicles in Tissue Engineering: Biology and Engineered Strategy. Adv Healthc Mater 2022; 11:e2201384. [PMID: 36053562 DOI: 10.1002/adhm.202201384] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/07/2022] [Indexed: 01/28/2023]
Abstract
Extracellular vesicles (EVs), acting as an important ingredient of intercellular communication through paracrine actions, have gained tremendous attention in the field of tissue engineering (TE). Moreover, these nanosized extracellular particles (30-140 nm) can be incorporated into biomaterials according to different principles to facilitate signal delivery in various regenerative processes directly or indirectly. Bioactive biomaterials as the carrier will extend the retention time and realize the controlled release of EVs, which further enhance their therapeutic efficiency in tissue regeneration. Herein, the basic biological characteristics of EVs are first introduced, and then their outstanding performance in exerting direct impacts on target cells in tissue regeneration as well as indirect effects on promoting angiogenesis and regulating the immune environment, due to specific functional components of EVs (nucleic acid, protein, lipid, etc.), is emphasized. Furthermore, different design ideas for suitable EV-loaded biomaterials are also demonstrated. In the end, this review also highlights the engineered strategies, which aim at solving the problems related to natural EVs such as highly heterogeneous functions, inadequate tissue targeting capabilities, insufficient yield and scalability, etc., thus promoting the therapeutic pertinence and clinical potential of EV-based approaches in TE.
Collapse
Affiliation(s)
- Ziyin Pan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weiyan Sun
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Yi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Hai Tang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Weikang Lin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| | - Jiafei Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School Of Medicine, Shanghai, 200092, China.,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, 200433, China
| |
Collapse
|
37
|
He W, Wang H, Zhang X, Mao T, Lu Y, Gu Y, Ju D, Qi L, Wang Q, Dong C. Construction of a decellularized spinal cord matrix/GelMA composite scaffold and its effects on neuronal differentiation of neural stem cells. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:2124-2144. [PMID: 35835455 DOI: 10.1080/09205063.2022.2102275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 06/15/2023]
Abstract
Spinal cord injury (SCI) leads to severe loss of motor and sensory functions, and the rehabilitation of SCI is a worldwide problem. Tissue-engineered scaffolds offer new hope for SCI patients, while the newly developed materials encountered a challenge in modeling the microenvironment around the lesion site. We constructed a new composite scaffold by mixing decellularized spinal cord extracellular matrix (dECM) with gelatin methacryloyl (GelMA). The dECM, as a natural biological material, retained a large number of proteins and growth factors related to neurogenesis. GelMA was a photopolymerizable material, harbored a polymer network structure, soft texture, certain shape and plenty of water. The viability, proliferation, and differentiation of neural stem cells (NSCs) on the composite scaffold were evaluated by cell count kit-8 (CCK8), Live/Dead assay, phalloidin staining, 5-Ethynyl-2'-deoxyurdine (EdU), immunofluorescence staining and western blot. The Live/Dead assay, phalloidin staining, EdU, and CCK8 assay showed that the composite scaffold had good biocompatibility and provided better support for proliferation of NSCs. Results of immunocytochemistry and western blot showed that the composite scaffolds promoted the specific differentiation of NSCs into neuron cells. Together, this dECM/GelMA composite scaffold can be used as a cell culture coating, the isolated NSCs seeded on the surface of composite scaffold expressed neuronal markers and assumed neuronal morphology. Our work provided a new method that would be widely used in tissue engineering of SCI.
Collapse
Affiliation(s)
- Wenhua He
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| | - Hui Wang
- Department of Emergency, Affiliated Hospital of Nantong University, Nantong, China
| | - Xuanxuan Zhang
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| | - Tiantian Mao
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| | - Yan Lu
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| | - Yu Gu
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| | - Dingyue Ju
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| | - Longju Qi
- Department of Hepatic Intervention, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Qinghua Wang
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| | - Chuanming Dong
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong, China
| |
Collapse
|
38
|
Tipa C, Cidade MT, Borges JP, Costa LC, Silva JC, Soares PIP. Clay-Based Nanocomposite Hydrogels for Biomedical Applications: A Review. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3308. [PMID: 36234440 PMCID: PMC9565291 DOI: 10.3390/nano12193308] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 06/16/2023]
Abstract
In recent decades, new and improved materials have been developed with a significant interest in three-dimensional (3D) scaffolds that can cope with the diverse needs of the expanding biomedical field and promote the required biological response in multiple applications. Due to their biocompatibility, ability to encapsulate and deliver drugs, and capacity to mimic the extracellular matrix (ECM), typical hydrogels have been extensively investigated in the biomedical and biotechnological fields. The major limitations of hydrogels include poor mechanical integrity and limited cell interaction, restricting their broad applicability. To overcome these limitations, an emerging approach, aimed at the generation of hybrid materials with synergistic effects, is focused on incorporating nanoparticles (NPs) within polymeric gels to achieve nanocomposites with tailored functionality and improved properties. This review focuses on the unique contributions of clay nanoparticles, regarding the recent developments of clay-based nanocomposite hydrogels, with an emphasis on biomedical applications.
Collapse
Affiliation(s)
- Cezar Tipa
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Maria T. Cidade
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - João P. Borges
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Luis C. Costa
- I3N and Physics Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Jorge C. Silva
- CENIMAT|i3N, Department of Physics, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Paula I. P. Soares
- CENIMAT|i3N, Department of Materials Science, School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| |
Collapse
|
39
|
Leung KS, Shirazi S, Cooper LF, Ravindran S. Biomaterials and Extracellular Vesicle Delivery: Current Status, Applications and Challenges. Cells 2022; 11:2851. [PMID: 36139426 PMCID: PMC9497093 DOI: 10.3390/cells11182851] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 12/14/2022] Open
Abstract
In this review, we will discuss the current status of extracellular vesicle (EV) delivery via biopolymeric scaffolds for therapeutic applications and the challenges associated with the development of these functionalized scaffolds. EVs are cell-derived membranous structures and are involved in many physiological processes. Naïve and engineered EVs have much therapeutic potential, but proper delivery systems are required to prevent non-specific and off-target effects. Targeted and site-specific delivery using polymeric scaffolds can address these limitations. EV delivery with scaffolds has shown improvements in tissue remodeling, wound healing, bone healing, immunomodulation, and vascular performance. Thus, EV delivery via biopolymeric scaffolds is becoming an increasingly popular approach to tissue engineering. Although there are many types of natural and synthetic biopolymers, the overarching goal for many tissue engineers is to utilize biopolymers to restore defects and function as well as support host regeneration. Functionalizing biopolymers by incorporating EVs works toward this goal. Throughout this review, we will characterize extracellular vesicles, examine various biopolymers as a vehicle for EV delivery for therapeutic purposes, potential mechanisms by which EVs exert their effects, EV delivery for tissue repair and immunomodulation, and the challenges associated with the use of EVs in scaffolds.
Collapse
Affiliation(s)
- Kasey S. Leung
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Sajjad Shirazi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Lyndon F. Cooper
- School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Sriram Ravindran
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL 60612, USA
| |
Collapse
|
40
|
GelMA Hydrogel Reinforced with 3D Printed PEGT/PBT Scaffolds for Supporting Epigenetically-Activated Human Bone Marrow Stromal Cells for Bone Repair. J Funct Biomater 2022; 13:jfb13020041. [PMID: 35466223 PMCID: PMC9036254 DOI: 10.3390/jfb13020041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/28/2022] [Accepted: 04/06/2022] [Indexed: 12/15/2022] Open
Abstract
Epigenetic approaches using the histone deacetylase 2 and 3 inhibitor-MI192 have been reported to accelerate stem cells to form mineralised tissues. Gelatine methacryloyl (GelMA) hydrogels provide a favourable microenvironment to facilitate cell delivery and support tissue formation. However, their application for bone repair is limited due to their low mechanical strength. This study aimed to investigate a GelMA hydrogel reinforced with a 3D printed scaffold to support MI192-induced human bone marrow stromal cells (hBMSCs) for bone formation. Cell culture: The GelMA (5 wt%) hydrogel supported the proliferation of MI192-pre-treated hBMSCs. MI192-pre-treated hBMSCs within the GelMA in osteogenic culture significantly increased alkaline phosphatase activity (p ≤ 0.001) compared to control. Histology: The MI192-pre-treated group enhanced osteoblast-related extracellular matrix deposition and mineralisation (p ≤ 0.001) compared to control. Mechanical testing: GelMA hydrogels reinforced with 3D printed poly(ethylene glycol)-terephthalate/poly(butylene terephthalate) (PEGT/PBT) scaffolds exhibited a 1000-fold increase in the compressive modulus compared to the GelMA alone. MI192-pre-treated hBMSCs within the GelMA–PEGT/PBT constructs significantly enhanced extracellular matrix collagen production and mineralisation compared to control (p ≤ 0.001). These findings demonstrate that the GelMA–PEGT/PBT construct provides enhanced mechanical strength and facilitates the delivery of epigenetically-activated MSCs for bone augmentation strategies.
Collapse
|
41
|
Man K, Brunet MY, Federici AS, Hoey DA, Cox SC. An ECM-Mimetic Hydrogel to Promote the Therapeutic Efficacy of Osteoblast-Derived Extracellular Vesicles for Bone Regeneration. Front Bioeng Biotechnol 2022; 10:829969. [PMID: 35433655 PMCID: PMC9005798 DOI: 10.3389/fbioe.2022.829969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
The use of extracellular vesicles (EVs) is emerging as a promising acellular approach for bone regeneration, overcoming translational hurdles associated with cell-based therapies. Despite their potential, EVs short half-life following systemic administration hinders their therapeutic efficacy. EVs have been reported to bind to extracellular matrix (ECM) proteins and play an essential role in matrix mineralisation. Chitosan and collagen type I are naturally-derived pro-osteogenic biomaterials, which have been demonstrated to control EV release kinetics. Therefore, this study aimed to develop an injectable ECM-mimetic hydrogel capable of controlling the release of osteoblast-derived EVs to promote bone repair. Pure chitosan hydrogels significantly enhanced compressive modulus (2.48-fold) and osteogenic differentiation (3.07-fold), whilst reducing gelation times (2.09-fold) and proliferation (2.7-fold) compared to pure collagen gels (p ≤ 0.001). EV release was strongly associated with collagen concentration (R2 > 0.94), where a significantly increased EV release profile was observed from chitosan containing gels using the CD63 ELISA (p ≤ 0.001). Hydrogel-released EVs enhanced human bone marrow stromal cells (hBMSCs) proliferation (1.12-fold), migration (2.55-fold), and mineralisation (3.25-fold) compared to untreated cells (p ≤ 0.001). Importantly, EV-functionalised chitosan-collagen composites significantly promoted hBMSCs extracellular matrix mineralisation when compared to the EV-free gels in a dose-dependent manner (p ≤ 0.001). Taken together, these findings demonstrate the development of a pro-osteogenic thermosensitive chitosan-collagen hydrogel capable of enhancing the therapeutic efficacy of osteoblast-derived EVs as a novel acellular tool for bone augmentation strategy.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Mathieu Y. Brunet
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Angelica S. Federici
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland,Dept. of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland,Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and RCSI, Dublin, Ireland
| | - David A. Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland,Dept. of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland,Advanced Materials and Bioengineering Research Centre, Trinity College Dublin and RCSI, Dublin, Ireland
| | - Sophie C. Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom,*Correspondence: Sophie C. Cox,
| |
Collapse
|
42
|
Printable Hydrogels Based on Alginate and Halloysite Nanotubes. Int J Mol Sci 2022; 23:ijms23063294. [PMID: 35328714 PMCID: PMC8954042 DOI: 10.3390/ijms23063294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
The design of hydrogels for the controlled release of active species is an attractive challenge. In this work, we prepared hybrid hydrogels composed of halloysite nanotubes as the inorganic component, and alginate as the organic counterpart. The reported procedure allowed us to provide the resulting materials with a peculiar wire-like shape. Both optical and scanning electron microscopy were used to characterize the morphological properties of the hydrogel wires, whose diameters were ca. 0.19 and 0.47 mm, respectively. The possibility to be exploited as drug delivery systems was carried out by loading the nanoclay with salicylic acid and by studying the release profiles. Thermogravimetric experiments showed that the amount of encapsulated drug was 4.35 wt%, and the salicylic acid was thermally stabilized after the loading into the nanotubes, as observed by the shift of the degradation peak in the differential thermograms from 193 to 267 °C. The kinetics investigation was conducted using UV–Vis spectrophotometry, and it exhibited the profound effects of both the morphology and dimensions on the release of the drugs. In particular, the release of 50% of the payload occurred in 6 and 10 h for the filiform hydrogels, and it was slower compared to the bare drug-loaded halloysite, which occurred in 2 h. Finally, an induction period of 2 h was observed in the release profile from the thicker sample.
Collapse
|