1
|
Han D, Wang W, Gong J, Ma Y, Li Y. Controlled delivery of mesenchymal stem cells via biodegradable scaffolds for fracture healing. Nanomedicine (Lond) 2025; 20:207-224. [PMID: 39686770 PMCID: PMC11731254 DOI: 10.1080/17435889.2024.2439242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Biodegradable controlled delivery systems for mesenchymal stem cells (MSCs) have emerged as novel advancements in the field of regenerative medicine, particularly for accelerating bone fracture healing. This detailed study emphasizes the importance of quick and adequate fracture treatment and the limitations of existing methods. New approaches employing biodegradable scaffolds can be placed within a fracture to serve as a mechanical support and allow controlled release of in situ MSCs and bioactive agents. They are made up of polymers and composites which degrade over time, aiding in natural tissue regrowth. The fabrication methods, including 3D printing, electrospinning, and solvent casting, with particulate leaching that enable precise control over scaffold architecture and properties, are discussed. Progress in controlled drug delivery systems including encapsulation techniques and release kinetics is described, highlighting the potential of such strategies to maintain therapeutic benefits over a prolonged time as well as improving outcomes for fracture repair. MSCs play a role in bone regeneration through differentiation using biodegradable scaffolds, paracrine effects, and regulation of inflammation focusing on fracture healing. Current trends and future directions in scaffold technology and MSC delivery, including smart scaffolds with growth factor incorporation and innovative delivery approaches for fracture healing are also discussed.
Collapse
Affiliation(s)
- Dong Han
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Weijiao Wang
- Otolaryngology Department, Yantaishan Hospital, Yantai, China
| | - Jinpeng Gong
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yupeng Ma
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| | - Yu Li
- Trauma Orthopedics Department, Yantaishan Hospital, Yantai, China
| |
Collapse
|
2
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2025; 14:e2402737. [PMID: 39506433 PMCID: PMC11730424 DOI: 10.1002/adhm.202402737] [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: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Wei Liu
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Jagadeesh K. Venkatesan
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Henning Madry
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| | - Magali Cucchiarini
- Center of Experimental OrthopaedicsSaarland University and Saarland University Medical CenterKirrbergerstr. Bldg 37D‐66421Homburg/SaarGermany
| |
Collapse
|
3
|
Liang W, Zhou C, Deng Y, Fu L, Zhao J, Long H, Ming W, Shang J, Zeng B. The current status of various preclinical therapeutic approaches for tendon repair. Ann Med 2024; 56:2337871. [PMID: 38738394 PMCID: PMC11095292 DOI: 10.1080/07853890.2024.2337871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 03/27/2024] [Indexed: 05/14/2024] Open
Abstract
Tendons are fibroblastic structures that link muscle and bone. There are two kinds of tendon injuries, including acute and chronic. Each form of injury or deterioration can result in significant pain and loss of tendon function. The recovery of tendon damage is a complex and time-consuming recovery process. Depending on the anatomical location of the tendon tissue, the clinical outcomes are not the same. The healing of the wound process is divided into three stages that overlap: inflammation, proliferation, and tissue remodeling. Furthermore, the curing tendon has a high re-tear rate. Faced with the challenges, tendon injury management is still a clinical issue that must be resolved as soon as possible. Several newer directions and breakthroughs in tendon recovery have emerged in recent years. This article describes tendon injury and summarizes recent advances in tendon recovery, along with stem cell therapy, gene therapy, Platelet-rich plasma remedy, growth factors, drug treatment, and tissue engineering. Despite the recent fast-growing research in tendon recovery treatment, still, none of them translated to the clinical setting. This review provides a detailed overview of tendon injuries and potential preclinical approaches for treating tendon injuries.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, China
| | - Yongjun Deng
- Department of Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Lifeng Fu
- Department of Orthopedics, Shaoxing City Keqiao District Hospital of Traditional Chinese Medicine, Shaoxing, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Hengguo Long
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Wenyi Ming
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| | - Jinxiang Shang
- Department of Orthopedics, Affiliated Hospital of Shaoxing University, Shaoxing, China
| | - Bin Zeng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, China
| |
Collapse
|
4
|
Li H, Li Y, Xiang L, Luo S, Zhang Y, Li S. Therapeutic potential of GDF-5 for enhancing tendon regenerative healing. Regen Ther 2024; 26:290-298. [PMID: 39022600 PMCID: PMC11252783 DOI: 10.1016/j.reth.2024.03.029] [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: 01/02/2024] [Revised: 03/24/2024] [Accepted: 03/28/2024] [Indexed: 07/20/2024] Open
Abstract
Tendon injury is a common disorder of the musculoskeletal system, with a higher possibility of occurrence in elderly individuals and athletes. After a tendon injury, the tendon suffers from inadequate and slow healing, resulting in the formation of fibrotic scar tissue, ending up with inferior functional properties. Therapeutic strategies involving the application of growth factors have been advocated to promote tendon healing. Growth and differentiation-5 (GDF-5) represents one such factor that has shown promising effect on tendon healing in animal models and in vitro cultures. Although promising, these studies are limited as the molecular mechanisms by which GDF-5 exerts its effect remain incompletely understood. Starting from broadly introducing essential elements of current understanding about GDF-5, the present review aims to define the effect of GDF-5 and its possible mechanisms of action in tendon healing. Nevertheless, we still need more in vivo studies to explore dosage, application time and delivery strategy of GDF-5, so as to pave the way for future clinical translation.
Collapse
Affiliation(s)
- Hanyue Li
- School of Physical Education, Southwest Medical University, PR China
| | - Yini Li
- Department of Ultrasound, The Affiliated Hospital of Southwest Medical University, Sichuan, PR China
| | - Linmei Xiang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Shengyu Luo
- School of Physical Education, Southwest Medical University, PR China
| | - Yan Zhang
- Luzhou Vocational and Technical College, PR China
| | - Sen Li
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, PR China
| |
Collapse
|
5
|
Wang X, Ma C, Zhang X, Yuan P, Wang Y, Fu M, Zhang Z, Shi R, Wei N, Wang J, Wu W. Mussel inspired 3D elastomer enabled rapid calvarial bone regeneration through recruiting more osteoprogenitors from the dura mater. Regen Biomater 2024; 11:rbae059. [PMID: 38911700 PMCID: PMC11193312 DOI: 10.1093/rb/rbae059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/17/2024] [Accepted: 05/10/2024] [Indexed: 06/25/2024] Open
Abstract
Currently, the successful healing of critical-sized calvarial bone defects remains a considerable challenge. The immune response plays a key role in regulating bone regeneration after material grafting. Previous studies mainly focused on the relationship between macrophages and bone marrow mesenchymal stem cells (BMSCs), while dural cells were recently found to play a vital role in the calvarial bone healing. In this study, a series of 3D elastomers with different proportions of polycaprolactone (PCL) and poly(glycerol sebacate) (PGS) were fabricated, which were further supplemented with polydopamine (PDA) coating. The physicochemical properties of the PCL/PGS and PCL/PGS/PDA grafts were measured, and then they were implanted as filling materials for 8 mm calvarial bone defects. The results showed that a matched and effective PDA interface formed on a well-proportioned elastomer, which effectively modulated the polarization of M2 macrophages and promoted the recruitment of dural cells to achieve full-thickness bone repair through both intramembranous and endochondral ossification. Single-cell RNA sequencing analysis revealed the predominance of dural cells during bone healing and their close relationship with macrophages. The findings illustrated that the crosstalk between dural cells and macrophages determined the vertical full-thickness bone repair for the first time, which may be the new target for designing bone grafts for calvarial bone healing.
Collapse
Affiliation(s)
- Xuqiao Wang
- The College of Life Sciences, Northwest University, Xi'an, 710127, PR China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Chaoqun Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Xinchi Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Pingping Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Yujiao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Mingdi Fu
- The College of Life Sciences, Northwest University, Xi'an, 710127, PR China
| | - Zheqian Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Ruiying Shi
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Na Wei
- The College of Life Sciences, Northwest University, Xi'an, 710127, PR China
| | - Juncheng Wang
- Institute of Stomatology, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Wei Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral & Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, PR China
| |
Collapse
|
6
|
Kawabata S, Nakasa T, Nekomoto A, Yimiti D, Miyaki S, Adachi N. Osteophyte Cartilage as a Potential Source for Minced Cartilage Implantation: A Novel Approach for Articular Cartilage Repair in Osteoarthritis. Int J Mol Sci 2024; 25:5563. [PMID: 38791601 PMCID: PMC11122408 DOI: 10.3390/ijms25105563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
Osteoarthritis (OA) is a common joint disorder characterized by cartilage degeneration, often leading to pain and functional impairment. Minced cartilage implantation (MCI) has emerged as a promising one-step alternative for large cartilage defects. However, the source of chondrocytes for MCI remains a challenge, particularly in advanced OA, as normal cartilage is scarce. We performed in vitro studies to evaluate the feasibility of MCI using osteophyte cartilage, which is present in patients with advanced OA. Osteophyte and articular cartilage samples were obtained from 22 patients who underwent total knee arthroplasty. Chondrocyte migration and proliferation were assessed using cartilage fragment/atelocollagen composites to compare the characteristics and regenerative potential of osteophytes and articular cartilage. Histological analysis revealed differences in cartilage composition between osteophytes and articular cartilage, with higher expression of type X collagen and increased chondrocyte proliferation in the osteophyte cartilage. Gene expression analysis identified distinct gene expression profiles between osteophytes and articular cartilage; the expression levels of COL2A1, ACAN, and SOX9 were not significantly different. Chondrocytes derived from osteophyte cartilage exhibit enhanced proliferation, and glycosaminoglycan production is increased in both osteophytes and articular cartilage. Osteophyte cartilage may serve as a viable alternative source of MCI for treating large cartilage defects in OA.
Collapse
Affiliation(s)
- Shingo Kawabata
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| | - Tomoyuki Nakasa
- Department of Artificial Joints and Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan
| | - Akinori Nekomoto
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| | - Dilimulati Yimiti
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| | - Shigeru Miyaki
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima City 734-8551, Japan;
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima City 734-8551, Japan; (S.K.); (A.N.); (D.Y.); (N.A.)
| |
Collapse
|
7
|
Nguyen NTK, Lee SS, Chen PH, Chang YH, Pham NN, Chang CW, Pham DH, Ngo DKT, Dang QT, Truong VA, Truong VA, Chang YH, Hu YC. Enhanced Calvarial Bone Repair Using ASCs Engineered with RNA-Guided Split dCas12a System that Co-Activates Sox 5, Sox6, and Long Non-Coding RNA H19. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306612. [PMID: 38126683 DOI: 10.1002/smll.202306612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/16/2023] [Indexed: 12/23/2023]
Abstract
Healing of large calvarial bone defects remains challenging. An RNA-guided Split dCas12a system is previously harnessed to activate long non-coding RNA H19 (lncRNA H19, referred to as H19 thereafter) in bone marrow-derived mesenchymal stem cells (BMSCs). H19 activation in BMSCs induces chondrogenic differentiation, switches bone healing pathways, and improves calvarial bone repair. Since adipose-derived stem cells (ASCs) can be harvested more easily in large quantity, here it is aimed to use ASCs as an alternative cell source. However, H19 activation alone using the Split dCas12a system in ASCs failed to elicit evident chondrogenesis. Therefore, split dCas12a activators are designed more to co-activate other chondroinductive transcription factors (Sox5, Sox6, and Sox9) to synergistically potentiate differentiation. It is found that co-activation of H19/Sox5/Sox6 in ASCs elicited more potent chondrogenic differentiation than activation of Sox5/Sox6/Sox9 or H19 alone. Co-activating H19/Sox5/Sox6 in ASCs significantly augmented in vitro cartilage formation and in vivo calvarial bone healing. These data altogether implicated the potentials of the Split dCas12a system to trigger multiplexed gene activation in ASCs for differentiation pathway reprogramming and tissue regeneration.
Collapse
Affiliation(s)
- Nuong Thi Kieu Nguyen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Shang-Shung Lee
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Pin-Hsin Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yi-Hao Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Nam Ngoc Pham
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Chin-Wei Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Dang Huu Pham
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Dung Kim Thi Ngo
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Quyen Thuc Dang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Vy Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Vu Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yu-Han Chang
- Department of Orthopaedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, 33305, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300044, Taiwan
| |
Collapse
|
8
|
Chen M, Jiang Z, Zou X, You X, Cai Z, Huang J. Advancements in tissue engineering for articular cartilage regeneration. Heliyon 2024; 10:e25400. [PMID: 38352769 PMCID: PMC10862692 DOI: 10.1016/j.heliyon.2024.e25400] [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: 11/03/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
Articular cartilage injury is a prevalent clinical condition resulting from trauma, tumors, infection, osteoarthritis, and other factors. The intrinsic lack of blood vessels, nerves, and lymphatic vessels within cartilage tissue severely limits its self-regenerative capacity after injury. Current treatment options, such as conservative drug therapy and joint replacement, have inherent limitations. Achieving perfect regeneration and repair of articular cartilage remains an ongoing challenge in the field of regenerative medicine. Tissue engineering has emerged as a key focus in articular cartilage injury research, aiming to utilize cultured and expanded tissue cells combined with suitable scaffold materials to create viable, functional tissues. This review article encompasses the latest advancements in seed cells, scaffolds, and cytokines. Additionally, the role of stimulatory factors including cytokines and growth factors, genetic engineering techniques, biophysical stimulation, and bioreactor systems, as well as the role of scaffolding materials including natural scaffolds, synthetic scaffolds, and nanostructured scaffolds in the regeneration of cartilage tissues are discussed. Finally, we also outline the signaling pathways involved in cartilage regeneration. Our review provides valuable insights for scholars to address the complex problem of cartilage regeneration and repair.
Collapse
Affiliation(s)
- Maohua Chen
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhiyuan Jiang
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiuyuan Zou
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Xiaobo You
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Zhen Cai
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jinming Huang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Poerio A, Mano JF, Cleymand F. Advanced 3D Printing Strategies for the Controlled Delivery of Growth Factors. ACS Biomater Sci Eng 2023; 9:6531-6547. [PMID: 37968925 DOI: 10.1021/acsbiomaterials.3c00873] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
The controlled delivery of growth factors (GFs) from tissue engineered constructs represents a promising strategy to improve tissue repair and regeneration. However, despite their established key role in tissue regeneration, the use of GFs is limited by their short half-life in the in vivo environment, their dose-dependent effectiveness, and their space- and time-dependent activity. Promising results have been obtained both in vitro and in vivo in animal models. Nevertheless, the clinical application of tissue engineered constructs releasing GFs is still challenging due to the several limitations and risks associated with their use. 3D printing and bioprinting, by allowing the microprecise spatial deposition of multiple materials and the fabrication of complex geometries with high resolution, offer advanced strategies for an optimal release of GFs from tissue engineered constructs. This review summarizes the strategies that have been employed to include GFs and their delivery system into biomaterials used for 3D printing applications to optimize their controlled release and to improve both the in vitro and in vivo regeneration processes. The approaches adopted to overcome the above-mentioned limitations are presented, showing the potential of the technology of 3D printing to get one step closer to clinical applications.
Collapse
Affiliation(s)
- Aurelia Poerio
- Institut Jean Lamour, University of Lorraine, Nancy 54011, France
| | - João F Mano
- Department of Chemistry, CICECO─Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Franck Cleymand
- Institut Jean Lamour, University of Lorraine, Nancy 54011, France
| |
Collapse
|
10
|
Yao H, Wang C, Zhang Y, Wan Y, Min Q. Manufacture of Bilayered Composite Hydrogels with Strong, Elastic, and Tough Properties for Osteochondral Repair Applications. Biomimetics (Basel) 2023; 8:biomimetics8020203. [PMID: 37218789 DOI: 10.3390/biomimetics8020203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Layered composite hydrogels have been considered attractive materials for use in osteochondral repair and regeneration. These hydrogel materials should be mechanically strong, elastic, and tough besides fulfilling some basic requirements such as biocompatibility and biodegradability. A novel type of bilayered composite hydrogel with multi-network structures and well-defined injectability was thus developed for osteochondral tissue engineering using chitosan (CH), hyaluronic acid (HA), silk fibroin (SF), CH nanoparticles (NPs), and amino-functionalized mesoporous bioglass (ABG) NPs. CH was combined with HA and CH NPs to build the chondral phase of the bilayered hydrogel, and CH, SF, and ABG NPs were used together to construct the subchondral phase of the bilayer hydrogel. Rheological measurements showed that the optimally achieved gels assigned to the chondral and subchondral layers had their elastic moduli of around 6.5 and 9.9 kPa, respectively, with elastic modulus/viscous modulus ratios higher than 36, indicating that they behaved like strong gels. Compressive measurements further demonstrated that the bilayered hydrogel with an optimally formulated composition had strong, elastic, and tough characteristics. Cell culture revealed that the bilayered hydrogel had the capacity to support the in-growth of chondrocytes in the chondral phase and osteoblasts in the subchondral phase. Results suggest that the bilayered composite hydrogel can act as an injective biomaterial for osteochondral repair applications.
Collapse
Affiliation(s)
- Hui Yao
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, China
| | - Congcong Wang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yuchen Zhang
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, China
| | - Ying Wan
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning 437100, China
- Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning 437100, China
| |
Collapse
|
11
|
Su D, Swearson S, Krongbaramee T, Sun H, Hong L, Amendt BA. Exploring microRNAs in craniofacial regenerative medicine. Biochem Soc Trans 2023; 51:841-854. [PMID: 37073783 PMCID: PMC11244734 DOI: 10.1042/bst20221448] [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: 11/14/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023]
Abstract
microRNAs (miRs) have been reported over the decades as important regulators in bone development and bone regeneration. They play important roles in maintaining the stem cell signature as well as regulating stem cell fate decisions. Thus, delivering miRs and miR inhibitors to the defect site is a potential treatment towards craniofacial bone defects. However, there are challenges in translation of basic research to clinics, including the efficiency, specificity, and efficacy of miR manipulation methods and the safety of miR delivery systems. In this review, we will compare miR oligonucleotides, mimics and antagomirs as therapeutic reagents to treat disease and regenerate tissues. Newer technology will be discussed as well as the efficiency and efficacy of using these technologies to express or inhibit miRs in treating and repairing oral tissues. Delivery of these molecules using extracellular vesicles and nanoparticles can achieve different results and depending on their composition will elicit specific effects. We will highlight the specificity, toxicity, stability, and effectiveness of several miR systems in regenerative medicine.
Collapse
Affiliation(s)
- Dan Su
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
| | - Samuel Swearson
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
| | - Tadkamol Krongbaramee
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
- Division of Endodontics, Department of Restorative Dentistry & Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Hongli Sun
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Liu Hong
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| |
Collapse
|
12
|
Xue Y, Li Y, Zhang D, Xu W, Ning C, Han D. Calcium Phosphate Silicate Microspheres with Soybean Lecithin as a Sustained-Release Bone Morphogenetic Protein-Delivery System for Bone Tissue Regeneration. ACS Biomater Sci Eng 2023; 9:2596-2607. [PMID: 36947498 DOI: 10.1021/acsbiomaterials.2c01065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Bone morphogenetic protein (BMP) is a growth factor that effectively promotes osteogenesis. Microsphere-based drug-delivery systems can facilitate an increase in the local concentration of BMP, thus promoting bone formation. In this study, calcium phosphate silicate (CPS) microspheres were used as drug-loading systems for BMP. Three groups─CPS, CPS + BMP, and CPS + BMP + soy lecithin (SL)─were set up, where SL was used to prolong the osteogenic effect of the microsphere system. Bone marrow mesenchymal stem cells and femoral defects in rats were used to compare the osteogenic ability of the three groups. The results indicated that CPS microspheres were good carriers of BMP, facilitating a smoother release into the cells and tissues. SL loading improved the loading rate of BMP, which promoted the osteogenic effect of the microspheres with BMP. We propose CPS microspheres as potential drug-delivery systems that can be effectively used in the treatment of bone defects.
Collapse
Affiliation(s)
- Yaxin Xue
- Department of Plastic and Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai Ninth People's Hospital, 639 Zhizaoju Road, 200011 Shanghai, People's Republic of China
| | - Yun Li
- Department of Plastic and Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai Ninth People's Hospital, 639 Zhizaoju Road, 200011 Shanghai, People's Republic of China
| | - Dong Zhang
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Wei Xu
- Department of Plastic and Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai Ninth People's Hospital, 639 Zhizaoju Road, 200011 Shanghai, People's Republic of China
| | - Congqin Ning
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China
| | - Dong Han
- Department of Plastic and Reconstructive Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai Ninth People's Hospital, 639 Zhizaoju Road, 200011 Shanghai, People's Republic of China
| |
Collapse
|
13
|
Kieu Nguyen NT, Tu Y, Lee HS, Truong VA, Chang YH, Pham NN, Chang CW, Lin YH, Lai PL, Chen PH, Parfyonova YV, Menshikov M, Chang YH, Hu YC. Split dCas12a activator for lncRNA H19 activation to enhance BMSC differentiation and promote calvarial bone healing. Biomaterials 2023; 297:122106. [PMID: 37030110 DOI: 10.1016/j.biomaterials.2023.122106] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/05/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Healing of large calvarial bone defects in adults is challenging. We previously showed that inducing chondrogenic differentiation of mesenchymal stem cells from bone marrow (BMSC) or adipose tissue (ASC) before implantation can switch the repair pathway and improve calvarial bone healing. Split dCas12a activator is a new CRISPR activation system comprising the amino (N) and carboxyl (C) fragments of dCas12a protein, each being fused with synthetic transcription activators at both termini. The split dCas12a activator was shown to induce programmable gene expression in cell lines. Here we exploited the split dCas12a activator to activate the expression of chondroinductive long non-coding RNA H19. We showed that co-expression of the split N- and C-fragments resulted in spontaneous dimerization, which elicited stronger activation of H19 than full-length dCas12a activator in rat BMSC and ASC. We further packaged the entire split dCas12a activator system (13.2 kb) into a hybrid baculovirus vector, which enhanced and prolonged H19 activation for at least 14 days in BMSC and ASC. The extended H19 activation elicited potent chondrogenic differentiation and inhibited adipogenesis. Consequently, the engineered BMSC promoted in vitro cartilage formation and augmented calvarial bone healing in rats. These data implicated the potentials of the split dCas12a activator for stem cell engineering and regenerative medicine.
Collapse
Affiliation(s)
- Nuong Thi Kieu Nguyen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yi Tu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hsiang-Sheng Lee
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Vu Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yi-Hao Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Nam Ngoc Pham
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chin-Wei Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ya-Hui Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, 333, Taiwan; Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Pin-Hsin Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yelena V Parfyonova
- National Medical Research Center of Cardiology, Russian Ministry of Health, Russia; Faculty of Medicine, Lomonosov Moscow State University, Russia
| | - Mikhail Menshikov
- Institute of Experimental Cardiology, National Cardiology Research Center, Russia
| | - Yu-Han Chang
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, 333, Taiwan; Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
14
|
Lee J, Kim D, Park S, Baek S, Jung J, Kim T, Han DK. Nitric Oxide-Releasing Bioinspired Scaffold for Exquisite Regeneration of Osteoporotic Bone via Regulation of Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205336. [PMID: 36581472 PMCID: PMC9951336 DOI: 10.1002/advs.202205336] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Osteoporotic bone regeneration is a challenging process which involves the occurrence of sophisticated interactions. Although various polymeric scaffolds have been proposed for bone repair, research on osteoporotic bone regeneration remains practically limited. In particular, achieving satisfactory bone regeneration when using osteoporotic drugs is challenging including bisphosphonates. Here, a novel nitric oxide-releasing bioinspired scaffold with bioactive agents for the exquisite regeneration of osteoporotic bone is proposed. The bone-like biomimetic poly(lactic-co-glycolic acid) scaffold is first prepared in combination with organic/inorganic ECM and magnesium hydroxide as the base implant material. Nanoparticles containing bioactive agents of zinc oxide (ZO), alendronate, and BMP2 are incorporated to the biomimetic scaffold to impart multifunctionality such as anti-inflammation, angiogenesis, anti-osteoclastogenesis, and bone regeneration. Especially, nitric oxide (NO) generated from ZO stimulates the activity of cGMP and protein kinase G; in addition, ZO downregulates the RANKL/osteoprotegerin ratio by suppressing the Wnt/β-catenin signaling pathway. The new bone is formed much better in the osteoporotic rat model than in the normal model through the regulation of bone homeostasis via the scaffold. These synergistic effects suggest that such a bioinspired scaffold could be a comprehensive way to regenerate exceptionally osteoporotic bones.
Collapse
Affiliation(s)
- Jun‐Kyu Lee
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| | - Da‐Seul Kim
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- School of Integrative EngineeringChung‐Ang University84 Heukseok‐ro, Dongjak‐guSeoul06974Republic of Korea
| | - So‐Yeon Park
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- Division of BiotechnologyCollege of Life Sciences and BiotechnologyKorea UniversitySeongbuk‐guSeoul02841Republic of Korea
| | - Seung‐Woon Baek
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- Department of Biomedical EngineeringSKKU Institute for ConvergenceSungkyunkwan University (SKKU)2066 Seobu‐ro, Jangan‐gu, Suwon‐siGyeonggi‐do16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSKKU Institute for ConvergenceSungkyunkwan University (SKKU)2066 Seobu‐ro, Jangan‐gu, Suwon‐siGyeonggi‐do16419Republic of Korea
| | - Ji‐Won Jung
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| | - Tae‐Hyung Kim
- School of Integrative EngineeringChung‐Ang University84 Heukseok‐ro, Dongjak‐guSeoul06974Republic of Korea
| | - Dong Keun Han
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| |
Collapse
|
15
|
Priddy LB, Krishnan L, Hettiaratchi MH, Karthikeyakannan S, Gupte N, Guldberg RE. Amniotic membrane attenuates heterotopic ossification following high-dose bone morphogenetic protein-2 treatment of segmental bone defects. J Orthop Res 2023; 41:130-140. [PMID: 35340049 PMCID: PMC9512937 DOI: 10.1002/jor.25324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/31/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023]
Abstract
Treatment of large bone defects with supraphysiological doses of bone morphogenetic protein-2 (BMP-2) has been associated with complications including heterotopic ossification (HO), inflammation, and pain, presumably due to poor spatiotemporal control of BMP-2. We have previously recapitulated extensive HO in our rat femoral segmental defect model by treatment with high-dose BMP-2 (30 μg). Using this model and BMP-2 dose, our objective was to evaluate the utility of a clinically available human amniotic membrane (AM) around the defect space for guided bone regeneration and reduction of HO. We hypothesized that AM surrounding collagen sponge would attenuate heterotopic ossification compared with collagen sponge alone. In vitro, AM retained more BMP-2 than a synthetic poly(ε-caprolactone) membrane through 21 days. In vivo, as hypothesized, the collagen + AM resulted in significantly less heterotopic ossification and correspondingly, lower total bone volume (BV), compared with collagen sponge alone. Although bone formation within the defect was delayed with AM around the defect, by 12 weeks, defect BVs were equivalent. Torsional stiffness was significantly reduced with AM but was equivalent to that of intact bone. Collagen + AM resulted in the formation of dense fibrous tissue and mineralized tissue, while the collagen group contained primarily mineralized tissue surrounded by marrow-like structures. Especially in conjunction with high doses of growth factor delivered via collagen sponge, these findings suggest AM may be effective as an overlay adjacent to bone healing sites to spatially direct bone regeneration and minimize heterotopic ossification.
Collapse
Affiliation(s)
- Lauren B. Priddy
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Department of Agricultural and Biological Engineering, Mississippi State University, 130 Creelman Street, Mississippi State, MS 39762, USA
| | - Laxminarayanan Krishnan
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Marian H. Hettiaratchi
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA
| | - Sukhita Karthikeyakannan
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Nikhil Gupte
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive NW, Atlanta, GA 30332, USA
- Current affiliation: Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
16
|
Wang L, Jiang J, Lin H, Zhu T, Cai J, Su W, Chen J, Xu J, Li Y, Wang J, Zhang K, Zhao J. Advances in Regenerative Sports Medicine Research. Front Bioeng Biotechnol 2022; 10:908751. [PMID: 35646865 PMCID: PMC9136559 DOI: 10.3389/fbioe.2022.908751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/21/2022] [Indexed: 01/08/2023] Open
Abstract
Regenerative sports medicine aims to address sports and aging-related conditions in the locomotor system using techniques that induce tissue regeneration. It also involves the treatment of meniscus and ligament injuries in the knee, Achilles’ tendon ruptures, rotator cuff tears, and cartilage and bone defects in various joints, as well as the regeneration of tendon–bone and cartilage–bone interfaces. There has been considerable progress in this field in recent years, resulting in promising steps toward the development of improved treatments as well as the identification of conundrums that require further targeted research. In this review the regeneration techniques currently considered optimal for each area of regenerative sports medicine have been reviewed and the time required for feasible clinical translation has been assessed. This review also provides insights into the direction of future efforts to minimize the gap between basic research and clinical applications.
Collapse
Affiliation(s)
- Liren Wang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Jiang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People’ Hospital, Shanghai, China
| | - Hai Lin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Tonghe Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center of Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Non-Coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
| | - Jiangyu Cai
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Wei Su
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jiebo Chen
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Junjie Xu
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yamin Li
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jing Wang
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
- *Correspondence: Kai Zhang, ; Jinzhong Zhao,
| | - Jinzhong Zhao
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Regenerative Sports Medicine and Translational Youth Science and Technology Innovation Workroom, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Regenerative Sports Medicine Lab of the Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People’ Hospital, Shanghai, China
- *Correspondence: Kai Zhang, ; Jinzhong Zhao,
| |
Collapse
|
17
|
Alksne M, Kalvaityte M, Simoliunas E, Gendviliene I, Barasa P, Rinkunaite I, Kaupinis A, Seinin D, Rutkunas V, Bukelskiene V. Dental pulp stem cell-derived extracellular matrix: autologous tool boosting bone regeneration. Cytotherapy 2022; 24:597-607. [PMID: 35304075 DOI: 10.1016/j.jcyt.2022.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/22/2021] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND AIMS To facilitate artificial bone construct integration into a patient's body, scaffolds are enriched with different biologically active molecules. Among various scaffold decoration techniques, coating surfaces with cell-derived extracellular matrix (ECM) is a rapidly growing field of research. In this study, for the first time, this technology was applied using primary dental pulp stem cells (DPSCs) and tested for use in artificial bone tissue construction. METHODS Rat DPSCs were grown on three-dimensional-printed porous polylactic acid scaffolds for 7 days. After the predetermined time, samples were decellularized, and the remaining ECM detailed proteomic analysis was performed. Further, DPSC-secreated ECM impact to mesenchymal stromal cells (MSC) behaviour as well as its role in osteoregeneration induction were analysed. RESULTS It was identified that DPSC-specific ECM protein network ornamenting surface-enhanced MSC attachment, migration and proliferation and even promoted spontaneous stem cell osteogenesis. This protein network also demonstrated angiogenic properties and did not stimulate MSCs to secrete molecules associated with scaffold rejection. With regard to bone defects, DPSC-derived ECM recruited endogenous stem cells, initiating the bone self-healing process. Thus, the DPSC-secreted ECM network was able to significantly enhance artificial bone construct integration and induce successful tissue regeneration. CONCLUSIONS DPSC-derived ECM can be a perfect tool for decoration of various biomaterials in the context of bone tissue engineering.
Collapse
Affiliation(s)
- Milda Alksne
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - Migle Kalvaityte
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Egidijus Simoliunas
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ieva Gendviliene
- Institute of Odontology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Povilas Barasa
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ieva Rinkunaite
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Algirdas Kaupinis
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Dmitrij Seinin
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Vygandas Rutkunas
- Institute of Odontology, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Virginija Bukelskiene
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
18
|
Liu S, Deng Z, Chen K, Jian S, Zhou F, Yang Y, Fu Z, Xie H, Xiong J, Zhu W. Cartilage tissue engineering: From proinflammatory and anti‑inflammatory cytokines to osteoarthritis treatments (Review). Mol Med Rep 2022; 25:99. [PMID: 35088882 PMCID: PMC8809050 DOI: 10.3892/mmr.2022.12615] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA), one of the most common joint diseases, is characterized by fibrosis, rhagadia, ulcers and attrition of articular cartilage due to a number of factors. The etiology of OA remains unclear, but its occurrence has been associated with age, obesity, inflammation, trauma and genetic factors. Inflammatory cytokines are crucial for the occurrence and progression of OA. The intra-articular proinflammatory and anti-inflammatory cytokines jointly maintain a dynamic balance, in accordance with the physiological metabolism of articular cartilage. However, dynamic imbalance between proinflammatory and anti-inflammatory cytokines can cause abnormal metabolism in knee articular cartilage, which leads to deformation, loss and abnormal regeneration, and ultimately destroys the normal structure of the knee joint. The ability of articular cartilage to self-repair once damaged is limited, due to its inability to obtain nutrients from blood vessels, nerves and lymphatic vessels, as well as limitations in the extracellular matrix. There are several disadvantages inherent to conventional repair methods, while cartilage tissue engineering (CTE), which combines proinflammatory and anti-inflammatory cytokines, offers a new therapeutic approach for OA. The aim of the present review was to examine the proinflammatory factors implicated in OA, including IL-1β, TNF-α, IL-6, IL-15, IL-17 and IL-18, as well as the key anti-inflammatory factors reducing OA-related articular damage, including IL-4, insulin-like growth factor and TGF-β. The predominance of proinflammatory over anti-inflammatory cytokine effects ultimately leads to the development of OA. CTE, which employs mesenchymal stem cells and scaffolding technology, may prevent OA by maintaining the homeostasis of pro- and anti-inflammatory factors.
Collapse
Affiliation(s)
- Shuyu Liu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Kang Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Shengsheng Jian
- Department of Orthopedics, Luo Hu Hospital, Shenzhen, Guangdong 518001, P.R. China
| | - Feifei Zhou
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Yuan Yang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zicai Fu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Huanyu Xie
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Jianyi Xiong
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Weimin Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
19
|
Truong VA, Lin YH, Nguyen NTK, Hsu MN, Pham NN, Chang YH, Chang CW, Shen CC, Lee HS, Lai PL, Parfyonova YV, Menshikov M, Wu JC, Chang YH, Hu YC. Bi-directional gene activation and repression promote ASC differentiation and enhance bone healing in osteoporotic rats. Mol Ther 2022; 30:92-104. [PMID: 34450254 PMCID: PMC8753367 DOI: 10.1016/j.ymthe.2021.08.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/16/2021] [Accepted: 08/08/2021] [Indexed: 01/07/2023] Open
Abstract
Calvarial bone healing is challenging, especially for individuals with osteoporosis because stem cells from osteoporotic patients are highly prone to adipogenic differentiation. Based on previous findings that chondrogenic induction of adipose-derived stem cells (ASCs) can augment calvarial bone healing, we hypothesized that activating chondroinductive Sox Trio genes (Sox5, Sox6, Sox9) and repressing adipoinductive genes (C/ebp-α, Ppar-γ) in osteoporotic ASCs can reprogram cell differentiation and improve calvarial bone healing after implantation. However, simultaneous gene activation and repression in ASCs is difficult. To tackle this problem, we built a CRISPR-BiD system for bi-directional gene regulation. Specifically, we built a CRISPR-AceTran system that exploited both histone acetylation and transcription activation for synergistic Sox Trio activation. We also developed a CRISPR interference (CRISPRi) system that exploited DNA methylation for repression of adipoinductive genes. We combined CRISPR-AceTran and CRISPRi to form the CRISPR-BiD system, which harnessed three mechanisms (transcription activation, histone acetylation, and DNA methylation). After delivery into osteoporotic rat ASCs, CRISPR-BiD significantly enhanced chondrogenesis and in vitro cartilage formation. Implantation of the engineered osteoporotic ASCs into critical-sized calvarial bone defects significantly improved bone healing in osteoporotic rats. These results implicated the potential of the CRISPR-BiD system for bi-directional regulation of cell fate and regenerative medicine.
Collapse
Affiliation(s)
- Vu Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Hui Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | | | - Mu-Nung Hsu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Nam Ngoc Pham
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Hao Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chin-Wei Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Che Shen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiang-Sheng Lee
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan,Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yelena V. Parfyonova
- National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow, Russia,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail Menshikov
- Institute of Experimental Cardiology, National Cardiology Research Center, Moscow, Russia
| | - Jaw-Ching Wu
- Medical Research Department, Taipei Veterans General Hospital, Taipei, Taiwan,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yu-Han Chang
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan,College of Medicine, Chang Gung University, Taoyuan, Taiwan,Corresponding author: Yu-Chen Hu, Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan,Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan,Corresponding author: Yu-Han Chang, Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan.
| |
Collapse
|
20
|
Oliveira AS, Silva JC, Figueiredo L, Ferreira FC, Kotov NA, Colaço R, Serro AP. High-performance bilayer composites for the replacement of osteochondral defects. Biomater Sci 2022; 10:5856-5875. [DOI: 10.1039/d2bm00716a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Two novel bilayer constructs for the repair of osteochondral defects were developed from nanofibers and ceramic particles embedded into PVA matrices, exhibiting multiple promising properties similar to those of corresponding natural tissues.
Collapse
Affiliation(s)
- A. S. Oliveira
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
- Instituto de Engenharia Mecânica and Department of Mechanical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
- Centro de Investigação Interdisciplinar Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal
| | - J. C. Silva
- Centre for Rapid and Sustainable Product Development, Politécnico de Leiria, Rua de Portugal – Zona Industrial, 2430-028 Marinha Grande, Portugal
- Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
| | - L. Figueiredo
- Bioceramed S.A., Rua José Gomes Ferreira 1 Arm. D, 2660-360 São Julião do Tojal, Portugal
| | - F. C. Ferreira
- Institute for Bioengineering and Biosciences and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB – Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
| | - N. A. Kotov
- Biointerfaces Institute and Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - R. Colaço
- Instituto de Engenharia Mecânica and Department of Mechanical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
| | - A. P. Serro
- Centro de Química Estrutural, Institute of Molecular Sciences and Department of Chemical Engineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
- Centro de Investigação Interdisciplinar Egas Moniz, Instituto Universitário Egas Moniz, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal
| |
Collapse
|
21
|
pH-Sensitive mesoporous bisphosphonate-based TiO2 nanoparticles utilized for controlled drug delivery of dexamethasone. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-021-01870-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Van Eps JL, Boada C, Scherba JC, Zavlin D, Arrighetti N, Shi A, Wang X, Tasciotti E, Buell JF, Ellsworth WA, Bonville DJ, Fernandez-Moure JS. Amniotic fluid allograft enhances the host response to ventral hernia repair using acellular dermal matrix. J Tissue Eng Regen Med 2021; 15:1092-1104. [PMID: 34599552 DOI: 10.1002/term.3255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/19/2021] [Accepted: 08/31/2021] [Indexed: 11/08/2022]
Abstract
Ventral hernia repair (VHR) with acellular dermal matrix (ADM) has high rates of recurrence that may be improved with allogeneic growth factor augmentation such as amniotic fluid allograft (AFA). We hypothesized that AFA would modulate the host response to improve ADM incorporation in VHR. Lewis rats underwent chronic VHR with porcine ADM alone or with AFA augmentation. Tissue harvested at 3, 14, or 28 days was assessed for region-specific cellularity, and a validated histomorphometric score was generated for tissue incorporation. Expression of pro-inflammatory (Nos1, Tnfα), anti-inflammatory (Arg1, Il-10, Mrc1) and tissue regeneration (Col1a1, Col3a1, Vegf, and alpha actinin-2) genes were quantified using quantitative reverse-transcription polymerase chain reaction. Amniotic fluid allograft treatment caused enhanced vascularization and cellularization translating to increased histomorphometric scores at 14 days, likely mediated by upregulation of pro-regeneration genes throughout the study period and molecular evidence of anti-inflammatory, M2-polarized macrophage phenotype. Collectively, this suggests AFA may have a therapeutic role as a VHR adjunct.
Collapse
Affiliation(s)
- Jeffrey L Van Eps
- Department of Surgery, Section of Colon & Rectal Surgery, University of Texas Health Science Center, McGovern Medical School, Houston, Texas, USA
| | - Christian Boada
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Houston Methodist Orthopedic and Sports Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Jacob C Scherba
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Dmitry Zavlin
- Department of Surgery, Plastic & Reconstructive Surgery, Houston Methodist Hospital, Houston, Texas, USA
| | - Noemi Arrighetti
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, Texas, USA
| | - Aaron Shi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, Texas, USA
| | - Xin Wang
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, Texas, USA
| | - Ennio Tasciotti
- Department of Human Sciences and Quality of Life Promotion, University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Joseph F Buell
- Department of Surgery, Mission Health, Asheville, North Carolina, USA
| | - Warren A Ellsworth
- Department of Surgery, Plastic & Reconstructive Surgery, Houston Methodist Hospital, Houston, Texas, USA
| | - Daniel J Bonville
- Department of Surgery, Division of Acute Care Surgery, Houston Methodist Hospital, Houston, Texas, USA
| | - Joseph S Fernandez-Moure
- Department of Surgery, Division of Trauma, Acute, and Critical Care Surgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
23
|
Chen YW, Shie MY, Chang WC, Shen YF. Approximate Optimization Study of Light Curing Waterborne Polyurethane Materials for the Construction of 3D Printed Cytocompatible Cartilage Scaffolds. MATERIALS 2021; 14:ma14226804. [PMID: 34832205 PMCID: PMC8626041 DOI: 10.3390/ma14226804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 11/18/2022]
Abstract
Articular cartilage, which is a white transparent tissue with 1–2 mm thickness, is located in the interface between the two hard bones. The main functions of articular cartilage are stress transmission, absorption, and friction reduction. The cartilage cannot be repaired and regenerated once it has been damaged, and it needs to be replaced by artificial joints. Many approaches, such as artificial joint replacement, hyaluronic acid injection, microfracture surgery and cartilage tissue engineering have been applied in clinical treatment. Basically, some of these approaches are foreign material implantation for joint replacement to reach the goal of pain reduction and mechanism support. This study demonstrated another frontier in the research of cartilage reconstruction by applying regeneration medicine additive manufacturing (3D Printing) and stem cell technology. Light curing materials have been modified and tested to be printable and cytocompatible for stem cells in this research. Design of experiments (DOE) is adapted in this investigation to search for the optimal manufacturing parameter for biocompatible scaffold fabrication and stem cell attachment and growth. Based on the results, an optimal working process of biocompatible and printable scaffolds for cartilage regeneration is reported. We expect this study will facilitate the development of cartilage tissue engineering.
Collapse
Affiliation(s)
- Yi-Wen Chen
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 40447, Taiwan; (Y.-W.C.); (M.-Y.S.); (W.-C.C.)
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 40447, Taiwan
| | - Ming-You Shie
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 40447, Taiwan; (Y.-W.C.); (M.-Y.S.); (W.-C.C.)
- School of Dentistry, China Medical University, Taichung City 40447, Taiwan
| | - Wen-Ching Chang
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 40447, Taiwan; (Y.-W.C.); (M.-Y.S.); (W.-C.C.)
| | - Yu-Fang Shen
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan
- High Performance Materials Institute for xD Printing, Asia University, Taichung City 41354, Taiwan
- Correspondence:
| |
Collapse
|
24
|
Remy MT, Akkouch A, He L, Eliason S, Sweat ME, Krongbaramee T, Fei F, Qian F, Amendt BA, Song X, Hong L. Rat Calvarial Bone Regeneration by 3D-Printed β-Tricalcium Phosphate Incorporating MicroRNA-200c. ACS Biomater Sci Eng 2021; 7:4521-4534. [PMID: 34437807 PMCID: PMC8441974 DOI: 10.1021/acsbiomaterials.0c01756] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
![]()
Advanced fabrication
methods for bone grafts designed to match
defect sites that combine biodegradable, osteoconductive materials
with potent, osteoinductive biologics would significantly impact the
clinical treatment of large bone defects. In this study, we engineered
synthetic bone grafts using a hybrid approach that combined three-dimensional
(3D-)printed biodegradable, osteoconductive β-tricalcium phosphate
(β-TCP) with osteoinductive microRNA(miR)-200c. 3D-printed β-TCP
scaffolds were fabricated utilizing a suspension-enclosing projection-stereolithography
(SEPS) process to produce constructs with reproducible microarchitectures
that enhanced the osteoconductive properties of β-TCP. Collagen
coating on 3D-printed β-TCP scaffolds slowed the release of
plasmid DNA encoding miR-200c compared to noncoated
constructs. 3D-printed β-TCP scaffolds coated with miR-200c-incorporated collagen increased the transfection efficiency of miR-200c of both rat and human BMSCs and additionally increased
osteogenic differentiation of hBMSCs in vitro. Furthermore, miR-200c-incorporated scaffolds significantly enhanced bone
regeneration in critical-sized rat calvarial defects. These results
strongly indicate that bone grafts combining SEPS 3D-printed osteoconductive
biomaterial-based scaffolds with osteoinductive miR-200c can be used
as superior bone substitutes for the clinical treatment of large bone
defects.
Collapse
Affiliation(s)
- Matthew T Remy
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Adil Akkouch
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Li He
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Steven Eliason
- Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Mason E Sweat
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Tadkamol Krongbaramee
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Fan Fei
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Fang Qian
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Brad A Amendt
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States.,Department of Anatomy and Cell Biology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States.,Center for Craniofacial Anomalies Research, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Xuan Song
- Department of Industrial and Systems Engineering, College of Engineering, The University of Iowa, Iowa City, Iowa 52242, United States
| | - Liu Hong
- Iowa Institute for Oral Health Research, College of Dentistry, The University of Iowa, Iowa City, Iowa 52242, United States.,Center for Craniofacial Anomalies Research, Carver College of Medicine, The University of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
25
|
Nguyen NTK, Chang YH, Truong VA, Hsu MN, Pham NN, Chang CW, Wu YH, Chang YH, Li H, Hu YC. CRISPR activation of long non-coding RNA DANCR promotes bone regeneration. Biomaterials 2021; 275:120965. [PMID: 34147719 DOI: 10.1016/j.biomaterials.2021.120965] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 06/06/2021] [Indexed: 01/20/2023]
Abstract
Healing of large calvarial bone defects in adults adopts intramembranous pathway and is difficult. Implantation of adipose-derived stem cells (ASC) that differentiate towards chondrogenic lineage can switch the bone repair pathway and improve calvarial bone healing. Long non-coding RNA DANCR was recently uncovered to promote chondrogenesis, but its roles in rat ASC (rASC) chondrogenesis and bone healing stimulation have yet to be explored. Here we first verified that DANCR expression promoted rASC chondrogenesis, thus we harnessed CRISPR activation (CRISPRa) technology to upregulate endogenous DANCR, stimulate rASC chondrogenesis and improve calvarial bone healing in rats. We generated 4 different dCas9-VPR orthologues by fusing a tripartite transcription activator domain VPR to catalytically dead Cas9 (dCas9) derived from 4 different bacteria, and compared the degree of activation using the 4 different dCas9-VPR. We unveiled surprisingly that the most commonly used dCas9-VPR derived from Streptococcus pyogenes barely activated DANCR. Nonetheless dCas9-VPR from Staphylococcus aureus (SadCas9-VPR) triggered efficient activation of DANCR in rASC. Delivery of SadCas9-VPR and the associated guide RNA into rASC substantially enhanced chondrogenic differentiation of rASC and augmented cartilage formation in vitro. Implantation of the engineered rASC remarkably potentiated the calvarial bone healing in rats. Furthermore, we identified that DANCR improved the rASC chondrogenesis through inhibition of miR-203a and miR-214. These results collectively proved that DANCR activation by SadCas9-VPR-based CRISPRa provides a novel therapeutic approach to improving calvarial bone healing.
Collapse
Affiliation(s)
- Nuong Thi Kieu Nguyen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Han Chang
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Linkou, 333, Taiwan; College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Vu Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Mu-Nung Hsu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Nam Ngoc Pham
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chin-Wei Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yi-Hsiu Wu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yi-Hao Chang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Hung Li
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
26
|
Liu Z, Liu X, Bao L, Liu J, Zhu X, Mo X, Tang R. The evaluation of functional small intestinal submucosa for abdominal wall defect repair in a rat model: Potent effect of sequential release of VEGF and TGF-β1 on host integration. Biomaterials 2021; 276:120999. [PMID: 34273685 DOI: 10.1016/j.biomaterials.2021.120999] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/14/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Ineffective vessel penetration and extracellular matrix (ECM) remodeling are responsible for the failure of porcine small intestinal submucosa (SIS)-repaired abdominal wall defects. Combined growth factors could be used as directing signals in a nature-mimicking strategy to improve this repair through mesh functionalization. In this work, vascular endothelial growth factor (VEGF) and transforming growth factor β1 (TGF-β1) were incorporated into a silk fibroin membrane via coaxial aqueous electrospinning to exploit their benefits of biological interactions. The membrane was sandwiched into the SIS bilayer as a functional mesh to repair partial-thickness defects in a rat model. Membrane characterization demonstrated that the core-shell structure ensured the independent distribution and sequential release of two regulators and protection of their bioactivities, which were confirmed by cell viability and protein expression. The mesh was further assessed to facilitate vasculature formation and collagen secretion in vitro, and exhibited better host integration than VEGF- or TGF-β1-containing mesh and developed reinforced mechanical properties compared with the VEGF-containing mesh after 28 days in vivo. Determination of the underlying biological interactions revealed that rapid VEGF release promotes angiogenesis and collagen secretion but initially potentiates the inflammatory response. Sustained TGF-β1 release at relatively low concentrations promoted VEGF for vessel permeation and maturation and steadily induced ECM remodeling under milder foreign body reactions. The functionalization of SIS improves repair by sufficient integration with timely remodeling and helps elucidate the related regulatory interactions.
Collapse
Affiliation(s)
- Zhengni Liu
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China
| | - Xuezhe Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Luhan Bao
- Group of Microbiological Engineering and Industrial Biotechnology, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Jiajie Liu
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China
| | - Xiaoqiang Zhu
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China
| | - Rui Tang
- Department of Hernia and Abdominal Wall Surgery, Shanghai East Hospital, TongJi University, 150 Ji Mo Road, Shanghai, 200120, PR China.
| |
Collapse
|
27
|
Khodr V, Machillot P, Migliorini E, Reiser JB, Picart C. High-throughput measurements of bone morphogenetic protein/bone morphogenetic protein receptor interactions using biolayer interferometry. Biointerphases 2021; 16:031001. [PMID: 34241280 PMCID: PMC7614001 DOI: 10.1116/6.0000926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/11/2021] [Indexed: 01/03/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are an important family of growth factors playing a role in a large number of physiological and pathological processes, including bone homeostasis, tissue regeneration, and cancers. In vivo, BMPs bind successively to both BMP receptors (BMPRs) of type I and type II, and a promiscuity has been reported. In this study, we used biolayer interferometry to perform parallel real-time biosensing and to deduce the kinetic parameters (ka, kd) and the equilibrium constant (KD) for a large range of BMP/BMPR combinations in similar experimental conditions. We selected four members of the BMP family (BMP-2, 4, 7, 9) known for their physiological relevance and studied their interactions with five type-I BMP receptors (ALK1, 2, 3, 5, 6) and three type-II BMP receptors (BMPR-II, ACTR-IIA, ACTR-IIB). We reveal that BMP-2 and BMP-4 behave differently, especially regarding their kinetic interactions and affinities with the type-II BMPR. We found that BMP-7 has a higher affinity for the type-II BMPR receptor ACTR-IIA and a tenfold lower affinity with the type-I receptors. While BMP-9 has a high and similar affinity for all type-II receptors, it can interact with ALK5 and ALK2, in addition to ALK1. Interestingly, we also found that all BMPs can interact with ALK5. The interaction between BMPs and both type-I and type-II receptors in a ternary complex did not reveal further cooperativity. Our work provides a synthetic view of the interactions of these BMPs with their receptors and paves the way for future studies on their cell-type and receptor specific signaling pathways.
Collapse
Affiliation(s)
- Valia Khodr
- Interdisciplinary Research Institute of Grenoble (IRIG), ERL BRM 5000 (CNRS/UGA/CEA), CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble cedex, France
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble
| | - Paul Machillot
- Interdisciplinary Research Institute of Grenoble (IRIG), ERL BRM 5000 (CNRS/UGA/CEA), CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble cedex, France
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble
| | - Elisa Migliorini
- Interdisciplinary Research Institute of Grenoble (IRIG), ERL BRM 5000 (CNRS/UGA/CEA), CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble cedex, France
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble
| | - Jean-Baptiste Reiser
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Catherine Picart
- Interdisciplinary Research Institute of Grenoble (IRIG), ERL BRM 5000 (CNRS/UGA/CEA), CEA Grenoble, 17 rue des Martyrs, 38054 Grenoble cedex, France
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble
| |
Collapse
|
28
|
Lou Y, Wang H, Ye G, Li Y, Liu C, Yu M, Ying B. Periosteal Tissue Engineering: Current Developments and Perspectives. Adv Healthc Mater 2021; 10:e2100215. [PMID: 33938636 DOI: 10.1002/adhm.202100215] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/18/2021] [Indexed: 12/22/2022]
Abstract
Periosteum, a highly vascularized bilayer connective tissue membrane plays an indispensable role in the repair and regeneration of bone defects. It is involved in blood supply and delivery of progenitor cells and bioactive molecules in the defect area. However, sources of natural periosteum are limited, therefore, there is a need to develop tissue-engineered periosteum (TEP) mimicking the composition, structure, and function of natural periosteum. This review explores TEP construction strategies from the following perspectives: i) different materials for constructing TEP scaffolds; ii) mechanical properties and surface topography in TEP; iii) cell-based strategies for TEP construction; and iv) TEP combined with growth factors. In addition, current challenges and future perspectives for development of TEP are discussed.
Collapse
Affiliation(s)
- Yiting Lou
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| | - Huiming Wang
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Guanchen Ye
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Yongzheng Li
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Chao Liu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Mengfei Yu
- The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Key Laboratory of Oral Biomedical Research of Zhejiang Province, 395 Yan'an road, Hangzhou, Zhejiang, 310003, China
| | - Binbin Ying
- Department of Stomatology, The Ningbo Hospital of Zhejiang University, and Ningbo First Hospital, 59 Liuting street, Ningbo, Zhejiang, 315000, China
| |
Collapse
|
29
|
Zhang Y, Fang C, Zhang S, Campbell RE, Serpe MJ. Controlled Osteogenic Differentiation of Human Mesenchymal Stem Cells Using Dexamethasone-Loaded Light-Responsive Microgels. ACS APPLIED MATERIALS & INTERFACES 2021; 13:7051-7059. [PMID: 33528987 DOI: 10.1021/acsami.0c17664] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Human mesenchymal stem cells (hMSCs), which have the ability to differentiate into osteoblasts, show promise for bone tissue engineering and bone defect treatment. While there are a number of approaches currently available to accomplish this, e.g., utilizing biodegradable materials loaded with the synthetic glucocorticoid osteogenic inducer dexamethasone (DEX), there are still many disadvantages with the current technologies. Here, we generated light-responsive microgels that we showed are capable of loading and releasing DEX in a light-triggered fashion, with the released DEX being able to induce hMSC differentiation into osteoblasts. Specifically, light-responsive poly(N-isopropylacrylamide-co-nitrobenzyl methacrylate) (pNIPAm-co-NBMA) microgels were synthesized via free radical precipitation polymerization and their size, morphology, and chemical composition were characterized. We then went on to show that the microgels could be loaded with DEX (via what we think are hydrophobic interactions) and released upon exposure to UV light. We went on to show that the DEX released from the microgels was still capable of inducing osteogenic differentiation of hMSCs using an alamarBlue assay and normalized alkaline phosphatase (ALP) activity assay. We also investigated how hMSC differentiation was impacted by intermittent DEX released from UV-exposed microgels. Finally, we confirmed that the microgels themselves were not cytotoxic to hMSCs. Taken together, the DEX-loaded light-responsive microgels reported here may find a use for niche clinical applications, e.g., bone tissue repair.
Collapse
Affiliation(s)
- Yingnan Zhang
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Changhao Fang
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Shuce Zhang
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Robert E Campbell
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| | - Michael J Serpe
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
| |
Collapse
|
30
|
Garot C, Bettega G, Picart C. Additive Manufacturing of Material Scaffolds for Bone Regeneration: Toward Application in the Clinics. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2006967. [PMID: 33531885 PMCID: PMC7116655 DOI: 10.1002/adfm.202006967] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Indexed: 05/07/2023]
Abstract
Additive manufacturing (AM) allows the fabrication of customized bone scaffolds in terms of shape, pore size, material type and mechanical properties. Combined with the possibility to obtain a precise 3D image of the bone defects using computed tomography or magnetic resonance imaging, it is now possible to manufacture implants for patient-specific bone regeneration. This paper reviews the state-of-the-art of the different materials and AM techniques used for the fabrication of 3D-printed scaffolds in the field of bone tissue engineering. Their advantages and drawbacks are highlighted. For materials, specific criteria, were extracted from a literature study: biomimetism to native bone, mechanical properties, biodegradability, ability to be imaged (implantation and follow-up period), histological performances and sterilization process. AM techniques can be classified in three major categories: extrusion-based, powder-based and liquid-base. Their price, ease of use and space requirement are analyzed. Different combinations of materials/AM techniques appear to be the most relevant depending on the targeted clinical applications (implantation site, presence of mechanical constraints, temporary or permanent implant). Finally, some barriers impeding the translation to human clinics are identified, notably the sterilization process.
Collapse
Affiliation(s)
- Charlotte Garot
- CEA, Université de Grenoble Alpes, CNRS, ERL 5000, IRIG Institute, 17 rue des Martyrs, F-38054, Grenoble, France
- CNRS and Grenoble Institute of Engineering, UMR 5628, LMGP, 3 parvis Louis Néel F-38016 Grenoble, France
| | - Georges Bettega
- Service de chirurgie maxillo-faciale, Centre Hospitalier Annecy-Genevois, 1 avenue de l’hôpital, F-74370 Epagny Metz-Tessy, France
- INSERM U1209, Institut Albert Bonniot, F-38000 Grenoble, France
| | - Catherine Picart
- CEA, Université de Grenoble Alpes, CNRS, ERL 5000, IRIG Institute, 17 rue des Martyrs, F-38054, Grenoble, France
- CNRS and Grenoble Institute of Engineering, UMR 5628, LMGP, 3 parvis Louis Néel F-38016 Grenoble, France
| |
Collapse
|
31
|
Liang W, Chen X, Dong Y, Zhou P, Xu F. Recent advances in biomaterials as instructive scaffolds for stem cells in tissue repair and regeneration. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1848832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| | - Xuerong Chen
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People’s Hospital, Shaoxing, P. R. China
| | - Ping Zhou
- Department of Orthopaedics, Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, P. R. China
| | - Fangming Xu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, P. R. China
| |
Collapse
|
32
|
Zhang J, Liu Z, Li Y, You Q, Yang J, Jin Y, Zou G, Tang J, Ge Z, Liu Y. FGF2: a key regulator augmenting tendon-to-bone healing and cartilage repair. Regen Med 2020; 15:2129-2142. [PMID: 33201773 DOI: 10.2217/rme-2019-0080] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Ligament/tendon and cartilage injuries are clinically common diseases that perplex most clinicians. Because of the lack of blood vessels and nerves, their self-repairing abilities are rather poor. Therefore, surgeries are necessary and also widely used to treat ligament/tendon or cartilage injuries. However, after surgery, there are still many problems that affect healing. In recent years, it has been found that exogenous FGF2 plays an important role in the repair of ligament/tendon and cartilage injuries and exerts a synergistic effect with endogenous FGF2. Therefore, FGF2 can be used as a new type of biomolecule to accelerate tendon-to-bone healing and cartilage repair after injury.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| | - Ziming Liu
- Peking University Institute of Sports Medicine, Beijing 100083, China
| | - Yuwan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qi You
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| | - Jibin Yang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| | - Ying Jin
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| | - Gang Zou
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| | - Jingfeng Tang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| | - Zhen Ge
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| | - Yi Liu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Guizhou 563000, China
| |
Collapse
|
33
|
Abstract
Dental implants are widely used in the field of oral restoration, but there are still problems leading to implant failures in clinical application, such as failed osseointegration, marginal bone resorption, and peri-implantitis, which restrict the success rate of dental implants and patient satisfaction. Poor osseointegration and bacterial infection are the most essential reasons resulting in implant failure. To improve the clinical outcomes of implants, many scholars devoted to modifying the surface of implants, especially to preparing different physical and chemical modifications to improve the osseointegration between alveolar bone and implant surface. Besides, the bioactive-coatings to promote the adhesion and colonization of ossteointegration-related proteins and cells also aim to improve the osseointegration. Meanwhile, improving the anti-bacterial performance of the implant surface can obstruct the adhesion and activity of bacteria, avoiding the occurrence of inflammation related to implants. Therefore, this review comprehensively investigates and summarizes the modifying or coating methods of implant surfaces, and analyzes the ossteointegration ability and anti-bacterial characteristics of emerging functional coatings in published references.
Collapse
|
34
|
Zhang LY, Bi Q, Zhao C, Chen JY, Cai MH, Chen XY. Recent Advances in Biomaterials for the Treatment of Bone Defects. Organogenesis 2020; 16:113-125. [PMID: 32799735 DOI: 10.1080/15476278.2020.1808428] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Bone defects or fractures generally heal in the absence of major interventions due to the high regenerative capacity of bone tissue. However, in situations of severe/large bone defects, these orchestrated regeneration mechanisms are impaired. With advances in modern medicine, natural and synthetic bio-scaffolds from bioceramics and polymers that support bone growth have emerged and gained intense research interest. In particular, scaffolds that recapitulate the molecular cues of extracellular signals, particularly growth factors, offer potential as therapeutic bone biomaterials. The current challenges for these therapies include the ability to engineer materials that mimic the biological and mechanical properties of the real bone tissue matrix, whilst simultaneously supporting bone vascularization. In this review, we discuss the very recent innovative strategies in bone biomaterial technology, including those of endogenous biomaterials and cell/drug delivery systems that promote bone regeneration. We present our understanding of their current value and efficacy, and the future perspectives for bone regenerative medicine.
Collapse
Affiliation(s)
- Le-Yi Zhang
- Department of General Surgery, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch) , Hangzhou, Zhejiang Province, China
| | - Qing Bi
- Department of Orthopedics, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College) , Hangzhou, China
| | - Chen Zhao
- Department of Orthopedics, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College) , Hangzhou, China
| | - Jin-Yang Chen
- Research and Development Department, Zhejiang Healthfuture Institute for Cell-Based Applied Technology , Hangzhou, Zhejiang Province, China
| | - Mao-Hua Cai
- Department of General Surgery, Chun'an First People's Hospital (Zhejiang Provincial People's Hospital Chun'an Branch) , Hangzhou, Zhejiang Province, China
| | - Xiao-Yi Chen
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College) , Hangzhou, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College) , Hangzhou, China
| |
Collapse
|
35
|
Tran HD, Park KD, Ching YC, Huynh C, Nguyen DH. A comprehensive review on polymeric hydrogel and its composite: Matrices of choice for bone and cartilage tissue engineering. J IND ENG CHEM 2020. [DOI: 10.1016/j.jiec.2020.06.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
36
|
Rodrigues EM, Viola KS, Gomes‐Cornélio AL, Soares‐Costa A, Henrique‐Silva F, Rossa‐Junior C, Guerreiro‐Tanomaru JM, Tanomaru‐Filho M. Sugarcane cystatin CaneCPI‐1 promotes osteogenic differentiation in human dental pulp cells: a new insight into cysteine proteases inhibitors. Int Endod J 2020; 53:1485-1493. [DOI: 10.1111/iej.13371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 01/09/2023]
Affiliation(s)
- E. M. Rodrigues
- Department of Restorative Dentistry Dental School of São Paulo State University Araraquara São PauloBrazil
| | - K. S. Viola
- Department of Restorative Dentistry Dental School of São Paulo State University Araraquara São PauloBrazil
| | - A. L. Gomes‐Cornélio
- Department of Restorative Dentistry Dental School of São Paulo State University Araraquara São PauloBrazil
| | - A. Soares‐Costa
- Department of Genetic and Evolution Federal University of Sao Carlos São CarlosBrazil
| | - F. Henrique‐Silva
- Department of Genetic and Evolution Federal University of Sao Carlos São CarlosBrazil
| | - C. Rossa‐Junior
- Department of Diagnosis and Surgery Dental School of São Paulo State University Araraquara São Paulo Brazil
| | - J. M. Guerreiro‐Tanomaru
- Department of Restorative Dentistry Dental School of São Paulo State University Araraquara São PauloBrazil
| | - M. Tanomaru‐Filho
- Department of Restorative Dentistry Dental School of São Paulo State University Araraquara São PauloBrazil
| |
Collapse
|
37
|
Chen L, Liu J, Guan M, Zhou T, Duan X, Xiang Z. Growth Factor and Its Polymer Scaffold-Based Delivery System for Cartilage Tissue Engineering. Int J Nanomedicine 2020; 15:6097-6111. [PMID: 32884266 PMCID: PMC7434569 DOI: 10.2147/ijn.s249829] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/02/2020] [Indexed: 02/05/2023] Open
Abstract
The development of biomaterials, stem cells and bioactive factors has led to cartilage tissue engineering becoming a promising tactic to repair cartilage defects. Various polymer three-dimensional scaffolds that provide an extracellular matrix (ECM) mimicking environment play an important role in promoting cartilage regeneration. In addition, numerous growth factors have been found in the regenerative process. However, it has been elucidated that the uncontrolled delivery of these factors cannot fully exert regenerative potential and can also elicit undesired side effects. Considering the complexity of the ECM, neither scaffolds nor growth factors can independently obtain successful outcomes in cartilage tissue engineering. Therefore, collectively, an appropriate combination of growth factors and scaffolds have great potential to promote cartilage repair effectively; this approach has become an area of considerable interest in recent investigations. Of late, an increasing trend was observed in cartilage tissue engineering towards this combination to develop a controlled delivery system that provides adequate physical support for neo-cartilage formation and also enables spatiotemporally delivery of growth factors to precisely and fully exert their chondrogenic potential. This review will discuss the role of polymer scaffolds and various growth factors involved in cartilage tissue engineering. Several growth factor delivery strategies based on the polymer scaffolds will also be discussed, with examples from recent studies highlighting the importance of spatiotemporal strategies for the controlled delivery of single or multiple growth factors in cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Li Chen
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China.,School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jiaxin Liu
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Ming Guan
- School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | - Tongqing Zhou
- School of Dentistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xin Duan
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Zhou Xiang
- Department of Orthopedics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| |
Collapse
|
38
|
Abstract
PURPOSE OF REVIEW The decreased contact area, edge loading, and increased stress in the adjacent area cartilage resulting from chondral defects are believed to predispose this tissue to degenerative changes that have significant economic implications, especially when considering its progression to osteoarthritis of the knee. Growth factors are considered therapeutic possibilities to enhance healing of chondral injuries and modify the progression to degenerative arthritis. Thus, the purposes of this review are to first to summarize important points for defect preparation and recent advances in techniques for marrow stimulation and second, and to identify specific growth factors and cytokines that have the capacity to advance cartilage regeneration and the treatment of osteoarthritis in light of recent laboratory and clinical studies. RECENT FINDINGS TGF-β, BMP-2, BMP-7, IGF-1, as IL-1 receptor antagonist, and recombinant human FGF-18 are some of the promising growth factor/cytokine treatments with pioneering and evolving clinical developments. The bulk of the review describes and discusses these developments in light of fundamental basic science. It is crucial to also understand the other underlying advances made in the surgical management of cartilage defects prior to onset of OA. These advances are in techniques for defect preparation and marrow stimulation, a common cartilage repair procedure used in combination with growth factor/cytokine augmentation. Multiple growth factor/cytokine modulation therapies are currently undergoing clinical trial investigation including Invossa (currently in phase III study), Kineret (currently in phase I study), and Sprifermin (currently in phase II study) for the treatment of symptomatic osteoarthritis.
Collapse
|
39
|
Montagna G, Cristofaro F, Fassina L, Bruni G, Cucca L, Kochen A, Divieti Pajevic P, Bragdon B, Visai L, Gerstenfeld L. An in vivo Comparison Study Between Strontium Nanoparticles and rhBMP2. Front Bioeng Biotechnol 2020; 8:499. [PMID: 32612980 PMCID: PMC7308719 DOI: 10.3389/fbioe.2020.00499] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/29/2020] [Indexed: 01/24/2023] Open
Abstract
The osteoinductive property of strontium was repeatedly proven in the last decades. Compelling in vitro data demonstrated that strontium hydroxyapatite nanoparticles exert a dual action, by promoting osteoblasts-driven matrix secretion and inhibiting osteoclasts-driven matrix resorption. Recombinant human bone morphogenetic protein 2 (rhBMP2) is a powerful osteoinductive biologic, used for the treatment of vertebral fractures and critically-sized bone defects. Although effective, the use of rhBMP2 has limitations due its recombinant morphogen nature. In this study, we examined the comparison between two osteoinductive agents: rhBMP2 and the innovative strontium-substituted hydroxyapatite nanoparticles. To test their effectiveness, we independently loaded Gelfoam sponges with the two osteoinductive agents and used the sponges as agent-carriers. Gelfoam are FDA-approved biodegradable medical devices used as delivery system for musculoskeletal defects. Their porous structure and spongy morphology make them attractive in orthopedic field. The abiotic characterization of the loaded sponges, involving ion release pattern and structure investigation, was followed by in vivo implantation onto the periosteum of healthy mice and comparison of the effects induced by each implant was performed. Abiotic analysis demonstrated that strontium was continuously released from the sponges over 28 days with a pattern similar to rhBMP2. Histological observations and gene expression analysis showed stronger endochondral ossification elicited by strontium compared to rhBMP2. Osteoclast activity was more inhibited by strontium than by rhBMP2. These results demonstrated the use of sponges loaded with strontium nanoparticles as potential bone grafts might provide better outcomes for complex fractures. Strontium nanoparticles are a novel and effective non-biologic treatment for bone injuries and can be used as novel powerful therapeutics for bone regeneration.
Collapse
Affiliation(s)
- Giulia Montagna
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Pavia, Italy.,Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Francesco Cristofaro
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Pavia, Italy
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Giovanna Bruni
- C.S.G.I. Department of Chemistry, Physical-Chemistry Section, University of Pavia, Pavia, Italy
| | - Lucia Cucca
- Department of Chemistry, University of Pavia, Pavia, Italy
| | - Alejandro Kochen
- Department of Translational Dental Medicine, Goldman School of Dental Medicine, Boston University, Boston, MA, United States
| | - Paola Divieti Pajevic
- Department of Translational Dental Medicine, Goldman School of Dental Medicine, Boston University, Boston, MA, United States
| | - Beth Bragdon
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, United States
| | - Livia Visai
- Department of Molecular Medicine (DMM), Center for Health Technologies (CHT), UdR INSTM, University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
| | - Louis Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
40
|
Childs RD, Nakao H, Isogai N, Murthy A, Landis WJ. An analytical study of neocartilage from microtia and otoplasty surgical remnants: A possible application for BMP7 in microtia development and regeneration. PLoS One 2020; 15:e0234650. [PMID: 32555733 PMCID: PMC7299323 DOI: 10.1371/journal.pone.0234650] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 05/30/2020] [Indexed: 02/07/2023] Open
Abstract
To investigate auricular reconstruction by tissue engineering means, this study compared cartilage regenerated from human chondrocytes obtained from either microtia or normal (conchal) tissues discarded from otoplasties. Isolated cells were expanded in vitro, seeded onto nanopolyglycolic acid (nanoPGA) sheets with or without addition of bone morphogenetic protein-7 (BMP7), and implanted in nude mice for 10 weeks. On specimen harvest, cartilage development was assessed by gross morphology, histology, and RT-qPCR and microarray analyses. Neocartilages from normal and microtia surgical tissues were found equivalent in their dimensions, qualitative degree of proteoglycan and elastic fiber staining, and quantitative gene expression levels of types II and III collagen, elastin, and SOX5. Microarray analysis, applied for the first time for normal and microtia neocartilage comparison, yielded no genes that were statistically significantly different in expression between these two sample groups. These results support use of microtia tissue as a cell source for normal auricular reconstruction. Comparison of normal and microtia cells, each seeded on nanoPGA and supplemented with BMP7 in a slow-release hydrogel, showed statistically significant differences in certain genes identified by microarray analysis. Such differences were also noted in several analyses comparing counterpart seeded cells without BMP7. Summary data suggest a possible application for BMP7 in microtia cartilage regeneration and encourage further studies to elucidate whether such genotypic differences translate to phenotypic characteristics of the human microtic ear. The present work advances understanding relevant to the potential clinical use of microtia surgical remnants as a suitable cell source for tissue engineering of the pinna.
Collapse
Affiliation(s)
- Robin DiFeo Childs
- Department of Polymer Science, University of Akron, Akron, Ohio, United States of America
- Department of Plastic and Reconstructive Surgery, Kindai University Medical School, Osaka sayama, Osaka, Japan
| | - Hitomi Nakao
- Division of Plastic and Reconstructive Surgery, Children’s Hospital Medical Center, Akron, Ohio, United States of America
| | - Noritaka Isogai
- Division of Plastic and Reconstructive Surgery, Children’s Hospital Medical Center, Akron, Ohio, United States of America
| | - Ananth Murthy
- Department of Plastic and Reconstructive Surgery, Kindai University Medical School, Osaka sayama, Osaka, Japan
| | - William J. Landis
- Department of Polymer Science, University of Akron, Akron, Ohio, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abalymov A, Parakhonskiy B, Skirtach AG. Polymer- and Hybrid-Based Biomaterials for Interstitial, Connective, Vascular, Nerve, Visceral and Musculoskeletal Tissue Engineering. Polymers (Basel) 2020; 12:E620. [PMID: 32182751 PMCID: PMC7182904 DOI: 10.3390/polym12030620] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/19/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022] Open
Abstract
In this review, materials based on polymers and hybrids possessing both organic and inorganic contents for repairing or facilitating cell growth in tissue engineering are discussed. Pure polymer based biomaterials are predominantly used to target soft tissues. Stipulated by possibilities of tuning the composition and concentration of their inorganic content, hybrid materials allow to mimic properties of various types of harder tissues. That leads to the concept of "one-matches-all" referring to materials possessing the same polymeric base, but different inorganic content to enable tissue growth and repair, proliferation of cells, and the formation of the ECM (extra cellular matrix). Furthermore, adding drug delivery carriers to coatings and scaffolds designed with such materials brings additional functionality by encapsulating active molecules, antibacterial agents, and growth factors. We discuss here materials and methods of their assembly from a general perspective together with their applications in various tissue engineering sub-areas: interstitial, connective, vascular, nervous, visceral and musculoskeletal tissues. The overall aims of this review are two-fold: (a) to describe the needs and opportunities in the field of bio-medicine, which should be useful for material scientists, and (b) to present capabilities and resources available in the area of materials, which should be of interest for biologists and medical doctors.
Collapse
Affiliation(s)
- Anatolii Abalymov
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | | | - Andre G. Skirtach
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
42
|
Porgham Daryasari M, Dusti Telgerd M, Hossein Karami M, Zandi-Karimi A, Akbarijavar H, Khoobi M, Seyedjafari E, Birhanu G, Khosravian P, SadatMahdavi F. Poly-l-lactic acid scaffold incorporated chitosan-coated mesoporous silica nanoparticles as pH-sensitive composite for enhanced osteogenic differentiation of human adipose tissue stem cells by dexamethasone delivery. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:4020-4029. [PMID: 31595797 DOI: 10.1080/21691401.2019.1658594] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nowadays, the development of drug-loaded electrospun organic-inorganic composite scaffolds for tissue engineering application is an attractive approach. In this study, a composite scaffold of Poly-l-lactic acid (PLLA) incorporated dexamethasone (Dexa) loaded Mesoporous Silica Nanoparticles (MSN) coated with Chitosan (CS) were fabricated by electrospinning for bone tissue engineering application. The MSN was prepared by precipitation method. After that, Dexamethasone (Dexa) was loaded into MSNs (MSN-Dexa). In the following, CS was coated over the prepared nanoparticles to form MSN-Dexa@CS and then, were mixed to PLLA solution to form MSN-Dexa@CS/PLLA composite for electrospinning. The surface morphology, hydrophilicity, tensile strength and the bioactivity of the scaffolds were characterized. The osteogenic proliferation and differentiation potential were evaluated by MTT assay and by measuring the basic osteogenic markers: the activity of the enzyme alkaline phosphatase and the level of calcium deposition. The composite scaffolds prepared here have conductive surface property and have a better osteogenic potential than pure PLLA scaffolds. Hence, the controlled release of nanoparticle containing Dexa from composite scaffold supported the osteogenesis and made the composite scaffolds ideal candidates for bone tissue engineering application and pH-sensitive delivery of drugs at the site of implantation in tissue regeneration.
Collapse
Affiliation(s)
- Mohammad Porgham Daryasari
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran.,Biomaterials Group, the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Dusti Telgerd
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | | | - Ali Zandi-Karimi
- Department of Biotechnology, College of Science, University of Tehran , Tehran , Iran
| | - Hamid Akbarijavar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Khoobi
- Biomaterials Group, the Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences , Tehran , Iran.,Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences , Tehran , Iran
| | - Ehsan Seyedjafari
- Department of Biotechnology, College of Science, University of Tehran , Tehran , Iran
| | - Gebremariam Birhanu
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, International campus (TUMS-IC) , Tehran , Iran.,School of Pharmacy, College of Health Sciences, Addis Ababa University , Addis Ababa , Ethiopia
| | - Pegah Khosravian
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences , Shahrekord , Iran
| | - Fatemeh SadatMahdavi
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran , Pakdasht, Tehran , Iran
| |
Collapse
|
43
|
PDGFB-expressing mesenchymal stem cells improve human hematopoietic stem cell engraftment in immunodeficient mice. Bone Marrow Transplant 2019; 55:1029-1040. [PMID: 31804621 PMCID: PMC7269905 DOI: 10.1038/s41409-019-0766-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
The bone marrow (BM) niche regulates multiple hematopoietic stem cell (HSC) processes. Clinical treatment for hematological malignancies by HSC transplantation often requires preconditioning via total body irradiation, which severely and irreversibly impairs the BM niche and HSC regeneration. Novel strategies are needed to enhance HSC regeneration in irradiated BM. We compared the effects of EGF, FGF2, and PDGFB on HSC regeneration using human mesenchymal stem cells (MSCs) that were transduced with these factors via lentiviral vectors. Among the above niche factors tested, MSCs transduced with PDGFB (PDGFB-MSCs) most significantly improved human HSC engraftment in immunodeficient mice. PDGFB-MSC-treated BM enhanced transplanted human HSC self-renewal in secondary transplantations more efficiently than GFP-transduced MSCs (GFP-MSCs). Gene set enrichment analysis showed increased antiapoptotic signaling in PDGFB-MSCs compared with GFP-MSCs. PDGFB-MSCs exhibited enhanced survival and expansion after transplantation, resulting in an enlarged humanized niche cell pool that provide a better humanized microenvironment to facilitate superior engraftment and proliferation of human hematopoietic cells. Our studies demonstrate the efficacy of PDGFB-MSCs in supporting human HSC engraftment.
Collapse
|
44
|
García-García A, Martin I. Extracellular Matrices to Modulate the Innate Immune Response and Enhance Bone Healing. Front Immunol 2019; 10:2256. [PMID: 31616429 PMCID: PMC6764079 DOI: 10.3389/fimmu.2019.02256] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/06/2019] [Indexed: 01/10/2023] Open
Abstract
Extracellular matrices (ECMs) have emerged as promising off-the-shelf products to induce bone regeneration, with the capacity not only to activate osteoprogenitors, but also to influence the immune response. ECMs generated starting from living cells such as mesenchymal stromal cells (MSCs) have the potential to combine advantages of native tissue-derived ECMs (e.g., physiological presentation of multiple regulatory factors) with those of synthetic ECMs (e.g., customization and reproducibility of composition). MSC-derived ECMs could be tailored by enrichment not only in osteogenic cytokines, but also in immunomodulatory factors, to skew the innate immune response toward regenerative processes. After reviewing the different immunoregulatory properties of ECM components, here we propose different approaches to engineer ECMs enriched in factors capable to regulate macrophage polarization, recruit host immune and mesenchymal cells, and stimulate the synthesis of other immunoinstructive cytokines. Finally, we offer a perspective on the possible evolution of the paradigm based on biological and chemico-physical design considerations, and the use of gene editing approaches.
Collapse
Affiliation(s)
- Andrés García-García
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
45
|
Lü J, Shi Y, Wang Y, Kang X, Bian X, Yuan B, Zhu M, Tang K. [Research progress of structured repair of tendon-bone interface]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:1064-1070. [PMID: 31512444 PMCID: PMC8355852 DOI: 10.7507/1002-1892.201811139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 07/15/2019] [Indexed: 01/08/2023]
Abstract
In sports system, the tendon-bone interface has the effect of tensile and bearing load, so the effect of healing plays a crucial role in restoring joint function. The process of repair is the formation of scar tissue, so it is difficult to achieve the ideal effect for morphology and biomechanical strength. The tissue engineering method can promote the tendon-bone interface healing from the seed cells, growth factors, and scaffolds, and is a new direction in the field of development of the tendon-bone interface healing.
Collapse
Affiliation(s)
- Jingtong Lü
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038, P.R.China
| | - Youxing Shi
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038, P.R.China
| | - Yunjiao Wang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038, P.R.China
| | - Xia Kang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038, P.R.China
| | - Xuting Bian
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038, P.R.China
| | - Bao Yuan
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038, P.R.China
| | - Min Zhu
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038, P.R.China
| | - Kanglai Tang
- Department of Orthopedics/Sports Medicine Center, State Key Laboratory of Trauma, Burn, and Combined Injury, the First Affiliated Hospital of Army Medical University (the Third Military Medical University), Chongqing, 400038,
| |
Collapse
|
46
|
García-Couce J, Almirall A, Fuentes G, Kaijzel E, Chan A, Cruz LJ. Targeting Polymeric Nanobiomaterials as a Platform for Cartilage Tissue Engineering. Curr Pharm Des 2019; 25:1915-1932. [DOI: 10.2174/1381612825666190708184745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023]
Abstract
Articular cartilage is a connective tissue structure that is found in anatomical areas that are important for the movement of the human body. Osteoarthritis is the ailment that most often affects the articular cartilage. Due to its poor intrinsic healing capacity, damage to the articular cartilage is highly detrimental and at present the reconstructive options for its repair are limited. Tissue engineering and the science of nanobiomaterials are two lines of research that together can contribute to the restoration of damaged tissue. The science of nanobiomaterials focuses on the development of different nanoscale structures that can be used as carriers of drugs / cells to treat and repair damaged tissues such as articular cartilage. This review article is an overview of the composition of articular cartilage, the causes and treatments of osteoarthritis, with a special emphasis on nanomaterials as carriers of drugs and cells, which reduce inflammation, promote the activation of biochemical factors and ultimately contribute to the total restoration of articular cartilage.
Collapse
Affiliation(s)
- Jomarien García-Couce
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Amisel Almirall
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Gastón Fuentes
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Eric Kaijzel
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| | - Alan Chan
- Percuros B.V., Zernikedreef 8, 2333 CL Leiden, Netherlands
| | - Luis J. Cruz
- Translational Nanobiomaterials and Imaging (TNI) group, Radiology department, Leiden University Medical Centrum, Leiden, Netherlands
| |
Collapse
|
47
|
Lepage SIM, Robson N, Gilmore H, Davis O, Hooper A, St John S, Kamesan V, Gelis P, Carvajal D, Hurtig M, Koch TG. Beyond Cartilage Repair: The Role of the Osteochondral Unit in Joint Health and Disease. TISSUE ENGINEERING PART B-REVIEWS 2019; 25:114-125. [PMID: 30638141 PMCID: PMC6486663 DOI: 10.1089/ten.teb.2018.0122] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Once believed to be limited to articular cartilage, osteoarthritis is now considered to be an organ disease of the “whole joint.” Damage to the articular surface can lead to, be caused by, or occur in parallel with, damage to other tissues in the joint. The relationship between cartilage and the underlying subchondral bone has particular importance when assessing joint health and determining treatment strategies. The articular cartilage is anchored to the subchondral bone through an interface of calcified cartilage, which as a whole makes up the osteochondral unit. This unit functions primarily by transferring load-bearing weight over the joint to allow for normal joint articulation and movement. Unfortunately, irreversible damage and degeneration of the osteochondral unit can severely limit joint function. Our understanding of joint pain, the primary complaint of patients, is poorly understood and past efforts toward structural cartilage restoration have often not been associated with a reduction in pain. Continued research focusing on the contribution of subchondral bone and restoration of the entire osteochondral unit are therefore needed, with the hope that this will lead to curative, and not merely palliative, treatment options. The purpose of this narrative review is to investigate the role of the osteochondral unit in joint health and disease. Topics of discussion include the crosstalk between cartilage and bone, the efficacy of diagnostic procedures, the origins of joint pain, current and emerging treatment paradigms, and suitable preclinical animal models for safety and efficacy assessment of novel osteochondral therapies. The goal of the review is to facilitate an appreciation of the important role played by the subchondral bone in joint pain and why the osteochondral unit as a whole should be considered in many cases of joint restoration strategies.
Collapse
Affiliation(s)
- Sarah I M Lepage
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Naomi Robson
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Hillary Gilmore
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Ola Davis
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Allyssa Hooper
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Stephanie St John
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Vashine Kamesan
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Paul Gelis
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Diana Carvajal
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| | - Mark Hurtig
- 2 Department of Clinical Studies, University of Guelph, Guelph, Canada
| | - Thomas G Koch
- 1 Department of Biomedical Sciences, University of Guelph, Guelph, Canada
| |
Collapse
|
48
|
Truong VA, Hsu MN, Kieu Nguyen NT, Lin MW, Shen CC, Lin CY, Hu YC. CRISPRai for simultaneous gene activation and inhibition to promote stem cell chondrogenesis and calvarial bone regeneration. Nucleic Acids Res 2019; 47:e74. [PMID: 30997496 PMCID: PMC6648329 DOI: 10.1093/nar/gkz267] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/28/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
Calvarial bone healing remains difficult but may be improved by stimulating chondrogenesis of implanted stem cells. To simultaneously promote chondrogenesis and repress adipogenesis of stem cells, we built a CRISPRai system that comprised inactive Cas9 (dCas9), two fusion proteins as activation/repression complexes and two single guide RNA (sgRNA) as scaffolds for recruiting activator (sgRNAa) or inhibitor (sgRNAi). By plasmid transfection and co-expression in CHO cells, we validated that dCas9 coordinated with sgRNAa to recruit the activator for mCherry activation and also orchestrated with sgRNAi to recruit the repressor for d2EGFP inhibition, without cross interference. After changing the sgRNA sequence to target endogenous Sox9/PPAR-γ, we packaged the entire CRISPRai system into an all-in-one baculovirus for efficient delivery into rat bone marrow-derived mesenchymal stem cells (rBMSC) and verified simultaneous Sox9 activation and PPAR-γ repression. The activation/inhibition effects were further enhanced/prolonged by using the Cre/loxP-based hybrid baculovirus. The CRISPRai system delivered by the hybrid baculovirus stimulated chondrogenesis and repressed adipogenesis of rBMSC in 2D culture and promoted the formation of engineered cartilage in 3D culture. Importantly, implantation of the rBMSC engineered by the CRISPRai improved calvarial bone healing. This study paves a new avenue to translate the CRISPRai technology to regenerative medicine.
Collapse
Affiliation(s)
- Vu Anh Truong
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Mu-Nung Hsu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Nuong Thi Kieu Nguyen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Mei-Wei Lin
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 300, Taiwan
| | - Chih-Che Shen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chin-Yu Lin
- Institute of New Drug Development, China Medical University, Taichung 404, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
49
|
Bai Y, Sha J, Kanno T, Miyamoto K, Hideshima K, Matsuzaki Y. Comparison of the Bone Regenerative Capacity of Three-Dimensional Uncalcined and Unsintered Hydroxyapatite/Poly-d/l-Lactide and Beta-Tricalcium Phosphate Used as Bone Graft Substitutes. J INVEST SURG 2019; 34:243-256. [PMID: 31122080 DOI: 10.1080/08941939.2019.1616859] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study compared the in vivo applicability of three-dimensional uncalcined and unsintered hydroxyapatite/poly-d/l-lactide (3D-HA/PDLLA) with beta-tricalcium phosphate (β-TCP). 3D-HA/PDLLA is a newly developed bioactive, osteoconductive, bioresorbable bone regenerative composite. We performed critical-defect surgery on the mandible body of rats; the defects were filled with one of two bone graft substitutes. After a 4-week follow-up period, the mandibular specimens were examined using hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) staining and micro-computed tomography (micro-CT). The H&E staining showed an increase in newly formed bone in both groups from week 1 to 4. The difference in the Runx2 IHC optical density (OD) scores of 3D-HA/PDLLA and β-TCP was not statistically significant (p > 0.05); however, the osteocalcin IHC OD scores of the groups differed significantly (p < 0.05). Micro-CT demonstrated a similar trabecular thickness, trabecular spacing, and bone volume per total volume in the two groups (p > 0.05), indicating that bone formation in the two groups was nearly the same from a macro-perspective of bone regeneration. These results demonstrated that a different bone regeneration pattern and earlier osteoblast differentiation occurred in 3D-HA/PDLLA compared with β-TCP. In conclusion, our study demonstrates that 3D-HA/PDLLA is feasible for clinical application as a new bioactive, osteoconductive/bioresorbable bone graft substitute for maxillofacial surgery.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Jingjing Sha
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Takahiro Kanno
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Kenichi Miyamoto
- Department of Cancer Biology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Katsumi Hideshima
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Yumi Matsuzaki
- Department of Cancer Biology, Shimane University Faculty of Medicine, Izumo, Japan
| |
Collapse
|
50
|
Lim ZXH, Rai B, Tan TC, Ramruttun AK, Hui JH, Nurcombe V, Teoh SH, Cool SM. Autologous bone marrow clot as an alternative to autograft for bone defect healing. Bone Joint Res 2019; 8:107-117. [PMID: 30997036 PMCID: PMC6444063 DOI: 10.1302/2046-3758.83.bjr-2018-0096.r1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Objectives Long bone defects often require surgical intervention for functional restoration. The ‘gold standard’ treatment is autologous bone graft (ABG), usually from the patient’s iliac crest. However, autograft is plagued by complications including limited supply, donor site morbidity, and the need for an additional surgery. Thus, alternative therapies are being actively investigated. Autologous bone marrow (BM) is considered as a candidate due to the presence of both endogenous reparative cells and growth factors. We aimed to compare the therapeutic potentials of autologous bone marrow aspirate (BMA) and ABG, which has not previously been done. Methods We compared the efficacy of coagulated autologous BMA and ABG for the repair of ulnar defects in New Zealand White rabbits. Segmental defects (14 mm) were filled with autologous clotted BM or morcellized autograft, and healing was assessed four and 12 weeks postoperatively. Harvested ulnas were subjected to radiological, micro-CT, histological, and mechanical analyses. Results Comparable results were obtained with autologous BMA clot and ABG, except for the quantification of new bone by micro-CT. Significantly more bone was found in the ABG-treated ulnar defects than in those treated with autologous BMA clot. This is possibly due to the remnants of necrotic autograft fragments that persisted within the healing defects at week 12 post-surgery. Conclusion As similar treatment outcomes were achieved by the two strategies, the preferred treatment would be one that is associated with a lower risk of complications. Hence, these results demonstrate that coagulated BMA can be considered as an alternative autogenous therapy for long bone healing. Cite this article: Z. X. H. Lim, B. Rai, T. C. Tan, A. K. Ramruttun, J. H. Hui, V. Nurcombe, S. H. Teoh, S. M. Cool. Autologous bone marrow clot as an alternative to autograft for bone defect healing. Bone Joint Res 2019;8:107–117. DOI: 10.1302/2046-3758.83.BJR-2018-0096.R1.
Collapse
Affiliation(s)
- Z X H Lim
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - B Rai
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Science and Maths Cluster, Singapore University of Technology & Design (SUTD), Singapore
| | - T C Tan
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore
| | - A K Ramruttun
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - J H Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - V Nurcombe
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College, Singapore
| | - S H Teoh
- Lee Kong Chian School of Medicine, Nanyang Technological University-Imperial College, Singapore; School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - S M Cool
- Glycotherapeutics Group, Institute of Medical Biology, Agency for Science, Technology and Research, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|