1
|
Tang X, Zhou F, Wang S, Wang G, Bai L, Su J. Bioinspired injectable hydrogels for bone regeneration. J Adv Res 2024:S2090-1232(24)00486-7. [PMID: 39505143 DOI: 10.1016/j.jare.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
The effective regeneration of bone/cartilage defects remains a significant clinical challenge, causing irreversible damage to millions annually.Conventional therapies such as autologous or artificial bone grafting often yield unsatisfactory outcomes, emphasizing the urgent need for innovative treatment methods. Biomaterial-based strategies, including hydrogels and active scaffolds, have shown potential in promoting bone/cartilage regeneration. Among them, injectable hydrogels have garnered substantial attention in recent years on account of their minimal invasiveness, shape adaptation, and controlled spatiotemporal release. This review systematically discusses the synthesis of injectable hydrogels, bioinspired approaches-covering microenvironment, structural, compositional, and bioactive component-inspired strategies-and their applications in various bone/cartilage disease models, highlighting bone/cartilage regeneration from an innovative perspective of bioinspired design. Taken together, bioinspired injectable hydrogels offer promising and feasible solutions for promoting bone/cartilage regeneration, ultimately laying the foundations for clinical applications. Furthermore, insights into further prospective directions for AI in injectable hydrogels screening and organoid construction are provided.
Collapse
Affiliation(s)
- Xuan Tang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, China
| | - Sicheng Wang
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai 201900, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Wenzhou Institute of Shanghai University, Wenzhou 325000, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
2
|
Li H, Zhao T, Yuan Z, Gao T, Yang Y, Li R, Tian Q, Tang P, Guo Q, Zhang L. Cartilage lacuna-biomimetic hydrogel microspheres endowed with integrated biological signal boost endogenous articular cartilage regeneration. Bioact Mater 2024; 41:61-82. [PMID: 39104774 PMCID: PMC11299526 DOI: 10.1016/j.bioactmat.2024.06.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 08/07/2024] Open
Abstract
Despite numerous studies on chondrogenesis, the repair of cartilage-particularly the reconstruction of cartilage lacunae through an all-in-one advanced drug delivery system remains limited. In this study, we developed a cartilage lacuna-like hydrogel microsphere system endowed with integrated biological signals, enabling sequential immunomodulation and endogenous articular cartilage regeneration. We first integrated the chondrogenic growth factor transforming growth factor-β3 (TGF-β3) into mesoporous silica nanoparticles (MSNs). Then, TGF-β3@MSNs and insulin-like growth factor 1 (IGF-1) were encapsulated within microspheres made of polydopamine (pDA). In the final step, growth factor-loaded MSN@pDA and a chitosan (CS) hydrogel containing platelet-derived growth factor-BB (PDGF-BB) were blended to produce growth factors loaded composite microspheres (GFs@μS) using microfluidic technology. The presence of pDA reduced the initial acute inflammatory response, and the early, robust release of PDGF-BB aided in attracting endogenous stem cells. Over the subsequent weeks, the continuous release of IGF-1 and TGF-β3 amplified chondrogenesis and matrix formation. μS were incorporated into an acellular cartilage extracellular matrix (ACECM) and combined with a polydopamine-modified polycaprolactone (PCL) structure to produce a tissue-engineered scaffold that mimicked the structure of the cartilage lacunae evenly distributed in the cartilage matrix, resulting in enhanced cartilage repair and patellar cartilage protection. This research provides a strategic pathway for optimizing growth factor delivery and ensuring prolonged microenvironmental remodeling, leading to efficient articular cartilage regeneration.
Collapse
Affiliation(s)
- Hao Li
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
- Department of Orthopaedics, Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
| | - Zhiguo Yuan
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tianze Gao
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Yongkang Yang
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Runmeng Li
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Qinyu Tian
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Peifu Tang
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| |
Collapse
|
3
|
Torabi Rahvar P, Abdekhodaie MJ, Jooybar E, Gantenbein B. An enzymatically crosslinked collagen type II/hyaluronic acid hybrid hydrogel: A biomimetic cell delivery system for cartilage tissue engineering. Int J Biol Macromol 2024; 279:134614. [PMID: 39127277 DOI: 10.1016/j.ijbiomac.2024.134614] [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: 12/27/2023] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
This study presents new injectable hydrogels based on hyaluronic acid and collagen type II that mimic the polysaccharide-protein structure of natural cartilage. After collagen isolation from chicken sternal cartilage, tyramine-grafted hyaluronic acid and collagen type II (HA-Tyr and COL-II-Tyr) were synthesized. Hybrid hydrogels were prepared with different ratios of HA-Tyr/COL-II-Tyr using horseradish peroxidase and noncytotoxic concentrations of hydrogen peroxide to encapsulate human bone marrow-derived mesenchymal stromal cells (hBM-MSCs). The findings showed that a higher HA-Tyr content resulted in a higher storage modulus and a lower hydrogel shrinkage, resulting in hydrogel swelling. Incorporating COL-II-Tyr into HA-Tyr hydrogels induced a more favorable microenvironment for hBM-MSCs chondrogenic differentiation. Compared to HA-Tyr alone, the hybrid HA-Tyr/COL-II-Tyr hydrogel promoted enhanced chondrocyte adhesion, spreading, proliferation, and upregulation of cartilage-related gene expression. These results highlight the promising potential of injectable HA-Tyr/COL-II-Tyr hybrid hydrogels to deliver cells for cartilage regeneration.
Collapse
Affiliation(s)
- Parisa Torabi Rahvar
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Mohammad J Abdekhodaie
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran; Environmental and Applied Science Management, Yeates School of Graduate Studies, Toronto Metropolitan University, Toronto, Canada.
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland; Inselspital, Bern University Hospital, Department of Orthopedic Surgery & Traumatology, Bern, Switzerland
| |
Collapse
|
4
|
Fu L, Wu J, Li P, Zheng Y, Zhang Z, Yuan X, Ding Z, Ning C, Sui X, Liu S, Shi S, Guo Q, Lin Y. A novel mesenchymal stem cell-targeting dual-miRNA delivery system based on aptamer-functionalized tetrahedral framework nucleic acids: Application to endogenous regeneration of articular cartilage. Bioact Mater 2024; 40:634-648. [PMID: 39253616 PMCID: PMC11381621 DOI: 10.1016/j.bioactmat.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/25/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
Articular cartilage injury (ACI) remains one of the key challenges in regenerative medicine, as current treatment strategies do not result in ideal regeneration of hyaline-like cartilage. Enhancing endogenous repair via microRNAs (miRNAs) shows promise as a regenerative therapy. miRNA-140 and miRNA-455 are two key and promising candidates for regulating the chondrogenic differentiation of mesenchymal stem cells (MSCs). In this study, we innovatively synthesized a multifunctional tetrahedral framework in which a nucleic acid (tFNA)-based targeting miRNA codelivery system, named A-T-M, was used. With tFNAs as vehicles, miR-140 and miR-455 were connected to and modified on tFNAs, while Apt19S (a DNA aptamer targeting MSCs) was directly integrated into the nanocomplex. The relevant results showed that A-T-M efficiently delivered miR-140 and miR-455 into MSCs and subsequently regulated MSC chondrogenic differentiation through corresponding mechanisms. Interestingly, a synergistic effect between miR-140 and miR-455 was revealed. Furthermore, A-T-M successfully enhanced the endogenous repair capacity of articular cartilage in vivo and effectively inhibited hypertrophic chondrocyte formation. A-T-M provides a new perspective and strategy for the regeneration of articular cartilage, showing strong clinical application value in the future treatment of ACI.
Collapse
Affiliation(s)
- Liwei Fu
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
| | - Jiang Wu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Pinxue Li
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
| | - Yazhe Zheng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Zhichao Zhang
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
| | - Xun Yuan
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Zhengang Ding
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
- Guizhou Medical University, Guiyang, 550004, Guizhou Province, People's Republic of China
| | - Chao Ning
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
| | - Shuyun Liu
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, People's Republic of China
- Department of Orthopedics, The Fourth Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, People's Republic of China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| |
Collapse
|
5
|
Huang CY, Chen CY, Wei CH, Yang JW, Lin YC, Kao CF, Chung JHY, Chen GY. Patterned graphene oxide via one-step thermal annealing for controlling collective cell migration. J Mater Chem B 2024; 12:8733-8745. [PMID: 39138950 DOI: 10.1039/d4tb01091d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Graphene oxide (GO) is a two-dimensional metastable nanomaterial. Interestingly, GO formed oxygen clusterings in addition to oxidized and graphitic phases during the low-temperature thermal annealing process, which could be further used for biomolecule bonding. By harnessing this property of GO, we created a bio-interface with patterned structures with a common laboratory hot plate that could tune cellular behavior by physical contact. Due to the regional distribution of oxygen clustering at the interface, we refer to it as patterned annealed graphene oxide (paGO). In addition, since the paGO was a heterogeneous interface and bonded biomolecules to varying degrees, arginine-glycine-aspartic acid (RGD) was modified on it and successfully regulated cellular-directed growth and migration. Finally, we investigated the FRET phenomenon of this heterogeneous interface and found that it has potential as a biosensor. The paGO interface has the advantages of easy regulation and fabrication, and the one-step thermal reduction method is suitable for biological applications. We believe that this low-temperature thermal annealing method would make GO interfaces more accessible, especially for the development of nano-interfacial modifications for biological applications, revealing its potential for biomedical applications.
Collapse
Affiliation(s)
- Chien-Yu Huang
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Chong-You Chen
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Chia-Hung Wei
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Jia-Wei Yang
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Yu-Chien Lin
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Chih-Fei Kao
- Department of Biological Science and Technology, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Johnson H Y Chung
- Intelligent Polymer Research Institute, Institute for Innovative Materials, University of Wollongong, 2500, NSW, Australia
| | - Guan-Yu Chen
- Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan.
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| |
Collapse
|
6
|
Guarnera D, Restaino F, Vannozzi L, Trucco D, Mazzocchi T, Worwąg M, Gapinski T, Lisignoli G, Zaffagnini S, Russo A, Ricotti L. Arthroscopic device with bendable tip for the controlled extrusion of hydrogels on cartilage defects. Sci Rep 2024; 14:19904. [PMID: 39191817 DOI: 10.1038/s41598-024-70426-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 08/16/2024] [Indexed: 08/29/2024] Open
Abstract
Advanced tools for the in situ treatment of articular cartilage lesions are attracting a growing interest in both surgery and bioengineering communities. The interest is particularly high concerning the delivery of cell-laden hydrogels. The tools currently available in the state-of-the-art hardly find an effective compromise between treatment accuracy and invasiveness. This paper presents a novel arthroscopic device provided with a bendable tip for the controlled extrusion of cell-laden hydrogels. The device consists of a handheld extruder and a supply unit that allows the extrusion of hydrogels. The extruder is equipped with a disposable, bendable nitinol tip (diameter: 4 mm, length: 92 mm, maximum bending angle: 90°) that guarantees access to hard-to-reach areas of the joint, which are difficult to get to, with conventional arthroscopic instruments. The tip accommodates a biocompatible polymer tube that is directly connected to the cartridge containing the hydrogel, whose plunger is actuated by a volumetric or pneumatic supply unit (both tested, in this study). Three different chondrocyte-laden hydrogels (RGD-modified Vitrogel®, methacrylated gellan gum, and an alginate-gelatine blend) were considered. First, the performance of the device in terms of resolution in hydrogel delivery was assessed, finding values in the range between 4 and 102 µL, with better performance found for the pneumatic supply unit and no significant differences between straight tip and bent tip conditions. Finite element simulations suggested that the shear stresses and pressure levels generated during the extrusion process were compatible with a safe deposition of the hydrogels. Biological analyses confirmed a high chondrocyte viability over a 7-day period after the extrusion of the three cell-laden hydrogel types, with no differences between the two supply units. The arthroscopic device was finally tested ex vivo by nine orthopedic surgeons on human cadaver knees. The device allowed surgeons to easily deliver hydrogels even in hard-to-reach cartilage areas. The outcomes of a questionnaire completed by the surgeons demonstrated a high usability of the device, with an overall preference for the pneumatic supply unit. Our findings provide evidence supporting the future arthroscopic device translation in pre-clinical and clinical scenarios, dealing with osteoarticular treatments.
Collapse
Affiliation(s)
- Daniele Guarnera
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy.
- Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy.
| | - Francesco Restaino
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
- Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
| | - Lorenzo Vannozzi
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
- Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
| | - Diego Trucco
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
- Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | | | - Michał Worwąg
- Vimex Endoscopy, Ul. Toruńska 27, 44-122, Gliwice, Poland
| | - Tomasz Gapinski
- Lega Medical Sp. Z o. O, ul. Majowa 11, 44-217, Rybnik, Poland
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | - Stefano Zaffagnini
- IRCCS Istituto Ortopedico Rizzoli, Orthopaedic and Traumatologic Clinic, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | - Alessandro Russo
- IRCCS Istituto Ortopedico Rizzoli, Orthopaedic and Traumatologic Clinic, Via di Barbiano, 1/10, 40136, Bologna, Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
- Department of Excellence in Robotics and AI, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy
| |
Collapse
|
7
|
Jakutowicz T, Wasyłeczko M, Płończak M, Wojciechowski C, Chwojnowski A, Czubak J. Comparative Study of Autogenic and Allogenic Chondrocyte Transplants on Polyethersulfone Scaffolds for Cartilage Regeneration. Int J Mol Sci 2024; 25:9075. [PMID: 39201763 PMCID: PMC11354243 DOI: 10.3390/ijms25169075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/10/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
The aim of this study was to evaluate the chondrogenic potential of chondrocyte transplants cultured in vitro on polyethersulfone (PES) membranes. Forty-eight rabbits (96 knee joints) were used in the project. The synthetic, macro-porous PES membranes were used as scaffolds. Fragments of articular cartilage were harvested from non-weight-bearing areas of the joints of the animals. Chondrocytes were isolated and then cultivated on PES scaffolds for 3 weeks. The animals were divided into four groups. All the lesions in the articular cartilage were full thickness defects. In Group I, autogenic chondrocytes on PES membranes were transplanted into the defect area; in Group II, allogenic chondrocytes on PES membranes were transplanted into the defect area; in Group III, pure PES membranes were transplanted into the defect area; and in Group IV, lesions were left untreated. Half of the animals from each group were terminated after 8 weeks, and the remaining half were terminated 12 weeks postoperatively. The samples underwent macroscopic evaluation using the Brittberg scale and microscopic evaluation using the O'Driscoll scale. The best regeneration was observed in Groups II and I. In Group I, the results were achieved with two surgeries, while in Group II, only one operation was needed. This indicates that allogenic chondrocytes do not require two surgeries, highlighting the importance of further in vivo studies to better understand this advantage. The success of the study and the desired properties of PES scaffolds are attributed mainly to the presence of sulfonic groups in the structure of the material. These groups, similar to chondroitin sulfate, which naturally occurs in hyaline cartilage, likely enable mutual affinity between the scaffold and cells and promote scaffold colonization by the cells.
Collapse
Affiliation(s)
- Tomasz Jakutowicz
- Paediatric Orthopaedics and Traumatology Department, Children’s Hospital, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Monika Wasyłeczko
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Maciej Płończak
- Department of Orthopedics, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
- Gruca Teaching Hospital, 05-400 Otwock, Poland
- Mazovia Regional Hospital in Siedlce, 08-110 Siedlce, Poland
| | - Cezary Wojciechowski
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Jarosław Czubak
- Department of Orthopedics, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
- Gruca Teaching Hospital, 05-400 Otwock, Poland
| |
Collapse
|
8
|
Cao M, Sheng R, Sun Y, Cao Y, Wang H, Zhang M, Pu Y, Gao Y, Zhang Y, Lu P, Teng G, Wang Q, Rui Y. Delivering Microrobots in the Musculoskeletal System. NANO-MICRO LETTERS 2024; 16:251. [PMID: 39037551 PMCID: PMC11263536 DOI: 10.1007/s40820-024-01464-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/16/2024] [Indexed: 07/23/2024]
Abstract
Disorders of the musculoskeletal system are the major contributors to the global burden of disease and current treatments show limited efficacy. Patients often suffer chronic pain and might eventually have to undergo end-stage surgery. Therefore, future treatments should focus on early detection and intervention of regional lesions. Microrobots have been gradually used in organisms due to their advantages of intelligent, precise and minimally invasive targeted delivery. Through the combination of control and imaging systems, microrobots with good biosafety can be delivered to the desired area for treatment. In the musculoskeletal system, microrobots are mainly utilized to transport stem cells/drugs or to remove hazardous substances from the body. Compared to traditional biomaterial and tissue engineering strategies, active motion improves the efficiency and penetration of local targeting of cells/drugs. This review discusses the frontier applications of microrobotic systems in different tissues of the musculoskeletal system. We summarize the challenges and barriers that hinder clinical translation by evaluating the characteristics of different microrobots and finally point out the future direction of microrobots in the musculoskeletal system.
Collapse
Affiliation(s)
- Mumin Cao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Renwang Sheng
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yimin Sun
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Ying Cao
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China
| | - Hao Wang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Ming Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yunmeng Pu
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
| | - Yucheng Gao
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Yuanwei Zhang
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Panpan Lu
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China
| | - Gaojun Teng
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Qianqian Wang
- Jiangsu Key Laboratory for Design and Manufacture of Micro-Nano Biomedical Instruments, School of Mechanical Engineering, Southeast University, Nanjing, 210009, People's Republic of China.
| | - Yunfeng Rui
- Department of Orthopaedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, People's Republic of China.
- School of Medicine, Southeast University, Nanjing, 210009, People's Republic of China.
- Orthopaedic Trauma Institute (OTI), Southeast University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
9
|
Epanomeritakis IE, Eleftheriou A, Economou A, Lu V, Khan W. Mesenchymal Stromal Cells for the Enhancement of Surgical Flexor Tendon Repair in Animal Models: A Systematic Review and Meta-Analysis. Bioengineering (Basel) 2024; 11:656. [PMID: 39061739 PMCID: PMC11274147 DOI: 10.3390/bioengineering11070656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/09/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Flexor tendon lacerations are primarily treated by surgical repair. Limited intrinsic healing ability means the repair site can remain weak. Furthermore, adhesion formation may reduce range of motion post-operatively. Mesenchymal stromal cells (MSCs) have been trialled for repair and regeneration of multiple musculoskeletal structures. Our goal was to determine the efficacy of MSCs in enhancing the biomechanical properties of surgically repaired flexor tendons. A PRISMA systematic review was conducted using four databases (PubMed, Ovid, Web of Science, and CINAHL) to identify studies using MSCs to augment surgical repair of flexor tendon injuries in animals compared to surgical repair alone. Nine studies were included, which investigated either bone marrow- or adipose-derived MSCs. Results of biomechanical testing were extracted and meta-analyses were performed regarding the maximum load, friction and properties relating to viscoelastic behaviour. There was no significant difference in maximum load at final follow-up. However, friction, a surrogate measure of adhesions, was significantly reduced following the application of MSCs (p = 0.04). Other properties showed variable results and dissipation of the therapeutic benefits of MSCs over time. In conclusion, MSCs reduce adhesion formation following tendon injury. This may result from their immunomodulatory function, dampening the inflammatory response. However, this may come at the cost of favourable healing which will restore the tendon's viscoelastic properties. The short duration of some improvements may reflect MSCs' limited survival or poor retention. Further investigation is needed to clarify the effect of MSC therapy and optimise its duration of action.
Collapse
Affiliation(s)
| | - Andreas Eleftheriou
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (A.E.); (A.E.); (V.L.)
| | - Anna Economou
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (A.E.); (A.E.); (V.L.)
| | - Victor Lu
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK; (A.E.); (A.E.); (V.L.)
| | - Wasim Khan
- Department of Trauma and Orthopaedic Surgery, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
10
|
Chen TY, Dai NT, Wen TK, Hsu SH. An Acellular, Self-Healed Trilayer Cryogel for Osteochondral Regeneration in Rabbits. Adv Healthc Mater 2024:e2400462. [PMID: 38948966 DOI: 10.1002/adhm.202400462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/13/2024] [Indexed: 07/02/2024]
Abstract
Osteochondral regeneration remains formidable challenges despite significant advances in microsurgery. Herein, an acellular trilayer cryogel (TC) with injectability, tunable pore sizes (80-200 µm), and appropriate compressive modulus (10.8 kPa) is manufactured from self-healable hydrogel under different gelling times through Schiff reaction between chitosan and difunctionalized polyurethane (DFPU). Bioactive molecules (Y27632 and dexamethasone) are respectively loaded in the top and bottom layers to form the Y27632/dexamethasone-loaded trilayer cryogel (Y/DEX-TC). Mesenchymal stem cells (MSCs) seeded in Y/DEX-TC proliferated ≈350% in vitro and underwent chondrogenesis or osteogenesis in response to the respective release of Y or DEX in 14 days. Acupuncture is administered to animals in an attempt to modulate the innate regulatory system and mobilize endogenous MSCs for osteochondral defect regeneration. In vivo rabbit experiments using Y/DEX-TC combined with acupuncture successfully regulate SDF-1 and TGF-β1 levels, which possibly cause MSC migration toward Y/DEX-TC. The synergistic effect of cryogel and acupuncture on immunomodulation is verified with a ≈7.3-fold enhancement of the M2-/M1-macrophage population ratio by treatment of Y/DEX-TC combining acupuncture, significantly greater than ≈1.5-fold increase by acupuncture or ≈2.2-fold increase by Y/DEX-TC alone. This novel strategy using acellular drug-loaded cryogel and accessible acupuncture shows promise in treating osteochondral defects of joint damage.
Collapse
Affiliation(s)
- Tsai-Yu Chen
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, 106319, R.O.C
| | - Niann-Tzyy Dai
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, 114202, R.O.C
| | - Tsung-Kai Wen
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan, 970374, R.O.C
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, 106319, R.O.C
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan, 350401, R.O.C
| |
Collapse
|
11
|
Amirhekmat A, Brown WE, Salinas EY, Hu JC, Athanasiou KA, Wang D. Mechanical Evaluation of Commercially Available Fibrin Sealants for Cartilage Repair. Cartilage 2024; 15:147-155. [PMID: 36974340 PMCID: PMC11368899 DOI: 10.1177/19476035231163273] [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: 06/17/2022] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/29/2023] Open
Abstract
OBJECTIVE Fibrin sealants are routinely used for intra-articular surgical fixation of cartilage fragments and implants. However, the mechanical properties of fibrin sealants in the context of cartilage repair are unknown. The purpose of this study was to characterize the adhesive and frictional properties of fibrin sealants using an ex vivo model. DESIGN Native bovine cartilage-bone composites were assembled with a single application of Tisseel or Vistaseal. Composites were tested in tension and lap shear. In addition, the coefficient of friction (COF) was measured in a native cartilage annulus model alone and with minced cartilage. Finally, the effect of a double application of fibrin sealant was evaluated. RESULTS There were no significant differences in tensile modulus, ultimate tensile strength (UTS), shear modulus, or ultimate shear strength (USS) between the 2 fibrin sealants. Both fibrin sealants demonstrated a UTS and USS of <8 and <30 kPa, respectively. There were no differences in COF between the sealants when tested alone or with minced cartilage. A double application of fibrin sealant did not alter the mechanical properties compared with a single application of fibrin sealant. CONCLUSIONS Fibrin sealant adhesive properties are not affected by the sealant type studied or the number of applications in a bovine cartilage-bone model. Fibrin sealant tribological properties are not affected by sealant type or the addition of minced cartilage. The adhesive properties of Tisseel and Vistaseal were less than those desired for the in vivo fixation of cartilage repair implants. These findings motivate the development of an improved cartilage-specific adhesive for cartilage repair applications.
Collapse
Affiliation(s)
- Arya Amirhekmat
- School of Medicine, University of California, Irvine, Irvine, CA, USA
- Department of Orthopaedic Surgery, University of California, Irvine, Orange, CA, USA
| | - Wendy E. Brown
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Evelia Y. Salinas
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Jerry C. Hu
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | | | - Dean Wang
- School of Medicine, University of California, Irvine, Irvine, CA, USA
- Department of Orthopaedic Surgery, University of California, Irvine, Orange, CA, USA
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
12
|
Nguyen M, Battistoni CM, Babiak PM, Liu JC, Panitch A. Chondroitin Sulfate/Hyaluronic Acid-Blended Hydrogels Suppress Chondrocyte Inflammation under Pro-Inflammatory Conditions. ACS Biomater Sci Eng 2024; 10:3242-3254. [PMID: 38632852 PMCID: PMC11094685 DOI: 10.1021/acsbiomaterials.4c00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
Osteoarthritis is characterized by enzymatic breakdown of the articular cartilage via the disruption of chondrocyte homeostasis, ultimately resulting in the destruction of the articular surface. Decades of research have highlighted the importance of inflammation in osteoarthritis progression, with inflammatory cytokines shifting resident chondrocytes into a pro-catabolic state. Inflammation can result in poor outcomes for cells implanted for cartilage regeneration. Therefore, a method to promote the growth of new cartilage and protect the implanted cells from the pro-inflammatory cytokines found in the joint space is required. In this study, we fabricate two gel types: polymer network hydrogels composed of chondroitin sulfate and hyaluronic acid, glycosaminoglycans (GAGs) known for their anti-inflammatory and prochondrogenic activity, and interpenetrating networks of GAGs and collagen I. Compared to a collagen-only hydrogel, which does not provide an anti-inflammatory stimulus, chondrocytes in GAG hydrogels result in reduced production of pro-inflammatory cytokines and enzymes as well as preservation of collagen II and aggrecan expression. Overall, GAG-based hydrogels have the potential to promote cartilage regeneration under pro-inflammatory conditions. Further, the data have implications for the use of GAGs to generally support tissue engineering in pro-inflammatory environments.
Collapse
Affiliation(s)
- Michael Nguyen
- Department
of Biomedical Engineering, University of
California, Davis, California 95616, United States
| | - Carly M. Battistoni
- Davidson
School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Paulina M. Babiak
- Davidson
School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Julie C. Liu
- Davidson
School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
- Weldon
School of Biomedical Engineering, Purdue
University, West Lafayette, Indiana 47907, United States
| | - Alyssa Panitch
- Department
of Biomedical Engineering, University of
California, Davis, California 95616, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, United States
| |
Collapse
|
13
|
Epanomeritakis IE, Khan WS. Adipose-derived regenerative therapies for the treatment of knee osteoarthritis. World J Stem Cells 2024; 16:324-333. [PMID: 38690511 PMCID: PMC11056639 DOI: 10.4252/wjsc.v16.i4.324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 04/25/2024] Open
Abstract
Knee osteoarthritis is a degenerative condition with a significant disease burden and no disease-modifying therapy. Definitive treatment ultimately requires joint replacement. Therapies capable of regenerating cartilage could significantly reduce financial and clinical costs. The regenerative potential of mesenchymal stromal cells (MSCs) has been extensively studied in the context of knee osteoarthritis. This has yielded promising results in human studies, and is likely a product of immunomodulatory and chondroprotective biomolecules produced by MSCs in response to inflammation. Adipose-derived MSCs (ASCs) are becoming increasingly popular owing to their relative ease of isolation and high proliferative capacity. Stromal vascular fraction (SVF) and micro-fragmented adipose tissue (MFAT) are produced by the enzymatic and mechanical disruption of adipose tissue, respectively. This avoids expansion of isolated ASCs ex vivo and their composition of heterogeneous cell populations, including immune cells, may potentiate the reparative function of ASCs. In this editorial, we comment on a multicenter randomized trial regarding the efficacy of MFAT in treating knee osteoarthritis. We discuss the study's findings in the context of emerging evidence regarding adipose-derived regenerative therapies. An underlying mechanism of action of ASCs is proposed while drawing important distinctions between the properties of isolated ASCs, SVF, and MFAT.
Collapse
Affiliation(s)
- Ilias E Epanomeritakis
- Division of Trauma and Orthopaedic Surgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, United Kingdom
| | - Wasim S Khan
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, United Kingdom.
| |
Collapse
|
14
|
Tu P, Pan Y, Wang L, Li B, Sun X, Liang Z, Liu M, Zhao Z, Wu C, Wang J, Wang Z, Song Y, Zhang Y, Ma Y, Guo Y. CD62E- and ROS-Responsive ETS Improves Cartilage Repair by Inhibiting Endothelial Cell Activation through OPA1-Mediated Mitochondrial Homeostasis. Biomater Res 2024; 28:0006. [PMID: 38439927 PMCID: PMC10911934 DOI: 10.34133/bmr.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/03/2024] [Indexed: 03/06/2024] Open
Abstract
Background: In the environment of cartilage injury, the activation of vascular endothelial cell (VEC), marked with excessive CD62E and reactive oxygen species (ROS), can affect the formation of hyaluronic cartilage. Therefore, we developed a CD62E- and ROS-responsive drug delivery system using E-selectin binding peptide, Thioketal, and silk fibroin (ETS) to achieve targeted delivery and controlled release of Clematis triterpenoid saponins (CS) against activated VEC, and thus promote cartilage regeneration. Methods: We prepared and characterized ETS/CS and verified their CD62E- and ROS-responsive properties in vitro. We investigated the effect and underlying mechanism of ETS/CS on inhibiting VEC activation and promoting chondrogenic differentiation of bone marrow stromal cells (BMSCs). We also analyzed the effect of ETS/CS on suppressing the activated VEC-macrophage inflammatory cascade in vitro. Additionally, we constructed a rat knee cartilage defect model and administered ETS/CS combined with BMSC-containing hydrogels. We detected the cartilage differentiation, the level of VEC activation and macrophage in the new tissue, and synovial tissue. Results: ETS/CS was able to interact with VEC and inhibit VEC activation through the carried CS. Coculture experiments verified ETS/CS promoted chondrogenic differentiation of BMSCs by inhibiting the activated VEC-induced inflammatory cascade of macrophages via OPA1-mediated mitochondrial homeostasis. In the rat knee cartilage defect model, ETS/CS reduced VEC activation, migration, angiogenesis in new tissues, inhibited macrophage infiltration and inflammation, promoted chondrogenic differentiation of BMSCs in the defective areas. Conclusions: CD62E- and ROS-responsive ETS/CS promoted cartilage repair by inhibiting VEC activation and macrophage inflammation and promoting BMSC chondrogenesis. Therefore, it is a promising therapeutic strategy to promote articular cartilage repair.
Collapse
Affiliation(s)
- Pengcheng Tu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Yalan Pan
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Lining Wang
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Bin Li
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Xiaoxian Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Zhongqing Liang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education,
Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Acupuncture and Tuina, School of Health and Rehabilitation,
Nanjing University of Chinese Medicine, Nanjing 210046, Jiangsu, China
| | - Mengmin Liu
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Zitong Zhao
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Chengjie Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Jianwei Wang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| | - Zhifang Wang
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, P.R. China
| | - Yu Song
- Zhangjiagang First People’s Hospital Affiliated to Soochow University, Zhangjiagang 215638, P.R. China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| | - Yong Ma
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| | - Yang Guo
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, P.R. China
- Laboratory of New Techniques of Restoration and Reconstruction of Orthopedics and Traumatology,
Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease,
Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214072, P.R. China
| |
Collapse
|
15
|
Miller Naranjo B, Zollo M, Sieber SA, Lieleg O. Lubricity, wear prevention, and anti-biofouling properties of macromolecular coatings for endotracheal tubes. Biomater Sci 2024; 12:1228-1238. [PMID: 38230671 DOI: 10.1039/d3bm01985c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Macromolecular coatings can improve the surface properties of many medical devices by enhancing their wetting behavior, tribological performance, and anti-biofouling properties - and covalent coatings produced from mucin glycoproteins have been shown to be very powerful in all those aspects. However, obtaining highly functional mucin glycoproteins is, at the moment, still a time-consuming process, which renders mucins rather expensive compared to other biomacromolecules. Here, we study a set of commercially available macromolecules that have the potential of substituting mucins in coatings for endotracheal tubes (ETTs). We present an overview of the different properties these macromolecular coatings establish on the ETT surface and whether they withstand storage or sterilization processes. Our study pinpoints several strategies of how to enhance the lubricity of ETTs by applying macromolecular coatings but also demonstrates the limited anti-biofouling abilities of well-established macromolecules such as hyaluronic acid, polyethylene glycol, and dextran. Based on the obtained results, we discuss to what extent those coatings can be considered equivalent alternatives to mucin coatings for applications on medical devices - their applicability does not have to be limited to ETTs, but could be broadened to catheters and endoscopes as well.
Collapse
Affiliation(s)
- Bernardo Miller Naranjo
- TUM School of Engineering and Design Department of Materials Engineering, Technical University of Munich (TUM), Boltzmannstraße 15, 85748 Garching, Germany.
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), Ernst-Otto-Fischer Straße 8, 85748 Garching, Germany
| | - Michael Zollo
- TUM School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II Center for Functional Protein Assemblies (CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer-Str. 8, Garching 85748, Germany
| | - Stephan A Sieber
- TUM School of Natural Sciences, Department of Bioscience, Chair of Organic Chemistry II Center for Functional Protein Assemblies (CPA), Technical University of Munich (TUM), Ernst-Otto-Fischer-Str. 8, Garching 85748, Germany
| | - Oliver Lieleg
- TUM School of Engineering and Design Department of Materials Engineering, Technical University of Munich (TUM), Boltzmannstraße 15, 85748 Garching, Germany.
- Center for Protein Assemblies (CPA) and Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), Ernst-Otto-Fischer Straße 8, 85748 Garching, Germany
| |
Collapse
|
16
|
Wang M, Wu Y, Li G, Lin Q, Zhang W, Liu H, Su J. Articular cartilage repair biomaterials: strategies and applications. Mater Today Bio 2024; 24:100948. [PMID: 38269053 PMCID: PMC10806349 DOI: 10.1016/j.mtbio.2024.100948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/09/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
Articular cartilage injury is a frequent worldwide disease, while effective treatment is urgently needed. Due to lack of blood vessels and nerves, the ability of cartilage to self-repair is limited. Despite the availability of various clinical treatments, unfavorable prognoses and complications remain prevalent. However, the advent of tissue engineering and regenerative medicine has generated considerable interests in using biomaterials for articular cartilage repair. Nevertheless, there remains a notable scarcity of comprehensive reviews that provide an in-depth exploration of the various strategies and applications. Herein, we present an overview of the primary biomaterials and bioactive substances from the tissue engineering perspective to repair articular cartilage. The strategies include regeneration, substitution, and immunization. We comprehensively delineate the influence of mechanically supportive scaffolds on cellular behavior, shedding light on emerging scaffold technologies, including stimuli-responsive smart scaffolds, 3D-printed scaffolds, and cartilage bionic scaffolds. Biologically active substances, including bioactive factors, stem cells, extracellular vesicles (EVs), and cartilage organoids, are elucidated for their roles in regulating the activity of chondrocytes. Furthermore, the composite bioactive scaffolds produced industrially to put into clinical use, are also explicitly presented. This review offers innovative solutions for treating articular cartilage ailments and emphasizes the potential of biomaterials for articular cartilage repair in clinical translation.
Collapse
Affiliation(s)
- Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- College of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai, 200941, China
| | - Qiushui Lin
- Department of Spine Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Wencai Zhang
- Department of Orthopedics, The First Affiliated Hospital Jinan University, Guangzhou, 510632, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Organoid Research Center, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| |
Collapse
|
17
|
Zhu W, Yang X, Liu S, Wang Y, Li W, Zhong Q, Zhang L, Xu J. Lentivirus-based shRNA of Caspase-3 gene silencing inhibits chondrocyte apoptosis and delays the progression of surgically induced osteoarthritis. Biotechnol J 2024; 19:e2300031. [PMID: 37750185 DOI: 10.1002/biot.202300031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Chondrocyte apoptosis is an important pathological feature of osteoarthritis (OA). Excessive apoptosis of chondrocytes disrupts the dynamic balance of cell proliferation and apoptosis, with a marked reduction in chondrocytes and cartilage matrix disintegration, which represents the main pathology of OA. Caspases, especially Caspase-3, play a central role in cell apoptosis. In this study, a lentiviral vector was used to transduce caspase-3 short hairpin RNA (shRNA) into rat chondrocytes (RCs), and the apoptotic and phenotypic genes of RCs were analyzed using real-time PCR and western blotting in vitro. In addition, in vivo intra-articular injection of Caspase-3 shRNA lentivirus was performed in a surgically induced OA rat model. Our results showed that Caspase-3 gene silencing could down-regulate the TNF-α-mediated inflammatory gene expression of TNFR1, FADD, and IL-1β, apoptotic gene expression of APAF1, Caspase-3, and Caspase-9, thereby attenuating the apoptotic pathway in vitro. Caspase-3 gene silencing also attenuated TNF-α-mediated decreased gene expression of ACAN, Col1-a1, and Col2-a1. Furthermore, Caspase-3 gene silencing could effectively reduce the OARSI score, and gene expression of Caspase-3, Caspase-9, MMP13, and TNF-α in a surgically induced OA rat model. Caspase-3 gene silencing may serve as a novel therapeutic strategy for cartilage injury and OA.
Collapse
Affiliation(s)
- Weicong Zhu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Xiaohong Yang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Shaojie Liu
- Surgical Department, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yiwen Wang
- Department of Pharmacy, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Wenxu Li
- Department of Orthopedics, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Qiguang Zhong
- Surgical Department, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Lihua Zhang
- Surgical Department, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
18
|
Fredrikson JP, Brahmachary PP, June RK, Cox LM, Chang CB. Pericellular Matrix Formation and Atomic Force Microscopy of Single Primary Human Chondrocytes Cultured in Alginate Microgels. Adv Biol (Weinh) 2024; 8:e2300268. [PMID: 37688354 PMCID: PMC10843004 DOI: 10.1002/adbi.202300268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/21/2023] [Indexed: 09/10/2023]
Abstract
One of the main components of articular cartilage is the chondrocyte's pericellular matrix (PCM), which is critical for regulating mechanotransduction, biochemical cues, and healthy cartilage development. Here, individual primary human chondrocytes (PHC) are encapsulated and cultured in 50 µm diameter alginate microgels using drop-based microfluidics. This unique culturing method enables PCM formation and manipulation of individual cells. Over ten days, matrix formation is observed using autofluorescence imaging, and the elastic moduli of isolated cells are measured using AFM. Matrix production and elastic modulus increase are observed for the chondrons cultured in microgels. Furthermore, the elastic modulus of cells grown in microgels increases ≈ten-fold over ten days, nearly reaching the elastic modulus of in vivo PCM. The AFM data is further analyzed using a Gaussian mixture model and shows that the population of PHCs grown in microgels exhibit two distinct populations with elastic moduli averaging 9.0 and 38.0 kPa. Overall, this work shows that microgels provide an excellent culture platform for the growth and isolation of PHCs, enabling PCM formation that is mechanically similar to native PCM. The microgel culture platform presented here has the potential to revolutionize cartilage regeneration procedures through the inclusion of in vitro developed PCM.
Collapse
Affiliation(s)
- Jacob P Fredrikson
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT, 59717, USA
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT, 59717, USA
| | - Priyanka P Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
- Department of Microbiology & Cell Biology, Montana State University, P.O. Box 173520, Bozeman, MT, 59717, USA
| | - Lewis M Cox
- Department of Mechanical & Industrial Engineering, Montana State University, P.O. Box 173800, Bozeman, MT, 59717, USA
| | - Connie B Chang
- Department of Chemical & Biological Engineering, Montana State University, P.O. Box 173920, Bozeman, MT, 59717, USA
- Center for Biofilm Engineering, Montana State University, P.O. Box 173980, Bozeman, MT, 59717, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| |
Collapse
|
19
|
Sun L, Xu Y, Han Y, Cui J, Jing Z, Li D, Liu J, Xiao C, Li D, Cai B. Collagen-Based Hydrogels for Cartilage Regeneration. Orthop Surg 2023; 15:3026-3045. [PMID: 37942509 PMCID: PMC10694028 DOI: 10.1111/os.13884] [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: 03/09/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 11/10/2023] Open
Abstract
Cartilage regeneration remains difficult due to a lack of blood vessels. Degradation of the extracellular matrix (ECM) causes cartilage defects, and the ECM provides the natural environment and nutrition for cartilage regeneration. Until now, collagen hydrogels are considered to be excellent material for cartilage regeneration due to the similar structure to ECM and good biocompatibility. However, collagen hydrogels also have several drawbacks, such as low mechanical strength, limited ability to induce stem cell differentiation, and rapid degradation. Thus, there is a demanding need to optimize collagen hydrogels for cartilage regeneration. In this review, we will first briefly introduce the structure of articular cartilage and cartilage defect classification and collagen, then provide an overview of the progress made in research on collagen hydrogels with chondrocytes or stem cells, comprehensively expound the research progress and clinical applications of collagen-based hydrogels that integrate inorganic or organic materials, and finally present challenges for further clinical translation.
Collapse
Affiliation(s)
- Lihui Sun
- Division of Bone and Joint Surgery, Center of OrthopaedicsFirst Hospital of Jilin UniversityChangchunPeople's Republic of China
| | - Yan Xu
- Division of Bone and Joint Surgery, Center of OrthopaedicsFirst Hospital of Jilin UniversityChangchunPeople's Republic of China
| | - Yu Han
- Division of Bone and Joint Surgery, Center of OrthopaedicsFirst Hospital of Jilin UniversityChangchunPeople's Republic of China
| | - Jing Cui
- Jilin Provincial Key Laboratory of Oral Biomedical Engineering, School and Hospital of StomatologyJilin UniversityChangchunChina
| | - Zheng Jing
- Division of Bone and Joint Surgery, Center of OrthopaedicsFirst Hospital of Jilin UniversityChangchunPeople's Republic of China
| | - Dongbo Li
- Division of Bone and Joint Surgery, Center of OrthopaedicsFirst Hospital of Jilin UniversityChangchunPeople's Republic of China
| | - Jianguo Liu
- Division of Bone and Joint Surgery, Center of OrthopaedicsFirst Hospital of Jilin UniversityChangchunPeople's Republic of China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunPeople's Republic of China
| | - Dongsong Li
- Division of Bone and Joint Surgery, Center of OrthopaedicsFirst Hospital of Jilin UniversityChangchunPeople's Republic of China
| | - Bo Cai
- Department of Ultrasound DiagnosisThe 964 Hospital of Chinese People's Liberation ArmyChangchunPeople's Republic of China
| |
Collapse
|
20
|
Yu X, Deng Z, Li H, Ma Y, Zheng Q. In situ fabrication of an anisotropic double-layer hydrogel as a bio-scaffold for repairing articular cartilage and subchondral bone injuries. RSC Adv 2023; 13:34958-34971. [PMID: 38046634 PMCID: PMC10688539 DOI: 10.1039/d3ra06222h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/15/2023] [Indexed: 12/05/2023] Open
Abstract
Articular cartilage is a smooth and elastic connective tissue playing load-bearing and lubricating roles in the human body. Normal articular cartilage comprises no blood vessels, lymphatic vessels, nerves, or undifferentiated cells, so damage self-repair is very unlikely. The injuries of articular cartilage are often accompanied by damage to the subchondral bone. The subchondral bone mainly provides mechanical support for the joint, and the successful repair of articular cartilage depends on the ability of the subchondral bone to provide a suitable environment. Currently, conventional repair treatments for articular cartilage and subchondral bone defects can hardly achieve good results due to the poor self-repairing ability of the cartilage Here, we propose a bioactive injectable double-layer hydrogel to repair articular cartilage and subchondral bone. The hydrogel scaffold mimics the multilayer structure of articular cartilage and subchondral bone. Agarose was used as a common base material for the double-layer hydrogel scaffold, in which a sodium alginate (SA)/agarose layer was used for the repair of artificially produced subchondral bone defects, while a decellularized extracellular matrix (dECM)/agarose layer was used for the repair of articular cartilage defects. The double-layer hydrogel scaffold is injectable, easy to use, and can fill in the damaged area. The hydrogel scaffold is also anisotropic both chemically and structurally. Animal experiments showed that the surface of the new cartilage tissue in the double-layer hydrogel scaffold group was closest to normal articular cartilage, with a structure similar to that of hyaline cartilage and a preliminary calcified layer. Moreover, the new subchondral bone in this group exhibited many regular bone trabeculae, and the new cartilage and subchondral bone were mechanically bound without mutual intrusion and tightly integrated with the surrounding tissue. The continuous double-layer hydrogel scaffold prepared in this study mimics the multilayer structure of articular cartilage and subchondral bone and promotes the functional repair of articular cartilage and subchondral bone, favoring close integration between the newborn tissue and the original tissue.
Collapse
Affiliation(s)
- Xiaotian Yu
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou, 510000 P.R. China
- Guangdong Cardiovascular Institute Guangzhou Guangdong China
| | - Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou, 510000 P.R. China
| | - Han Li
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou, 510000 P.R. China
- Guangdong Cardiovascular Institute Guangzhou Guangdong China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou, 510000 P.R. China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou, 510000 P.R. China
| |
Collapse
|
21
|
Chitchongyingcharoen N, Tawonsawatruk T, Phetfong J, Aroontanee W, Supokawej A. Application of human platelet lysate in chondrocyte expansion promotes chondrogenic phenotype and slows senescence progression via BMP-TAK1-p38 pathway. Sci Rep 2023; 13:21106. [PMID: 38036641 PMCID: PMC10689743 DOI: 10.1038/s41598-023-48544-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/28/2023] [Indexed: 12/02/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common musculoskeletal degenerative. OA treatments are aiming to slow down disease progression; however, lack of cartilage regeneration efficacy. Autologous chondrocyte implantation (ACI) is a promising cartilage-regeneration strategy that uses human articular chondrocytes (HACs) as cellular materials. However, the unreadiness of HACs from prolonged expansion, cellular senescence, and chondrogenic dedifferentiation occurred during conventional expansion, thus, minimizing the clinical efficacy of ACI. We aimed to examine the effects of a human platelet lysate (HPL) as an alternative human-derived HAC medium supplement to overcome the limitations of conventional expansion, and to explain the mechanism underlying the effects of HPL. During passages 2-4 (P2-P4), HPL significantly increased HAC proliferation capacities and upregulated chondrogenic markers. Simultaneously, HPL significantly reduced HAC senescence compared with conventional condition. HACs treated with LDN193189 exhibited a reduction in proliferation capacity and chondrogenic marker expression, whereas the HAC senescence increased slightly. These findings indicated involvement of BMP-2 signaling transduction in the growth-assistive, anti-senescent, and chondrogenic-inductive properties of HPL, which demonstrated its beneficial effects for application as HAC medium supplement to overcome current expansion limitations. Finally, our findings support the roles of platelets in platelet-rich plasma as a promising treatment for patients with OA.
Collapse
Affiliation(s)
- Narong Chitchongyingcharoen
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Tulyapruek Tawonsawatruk
- Department of Orthopedics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jitrada Phetfong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Wrattya Aroontanee
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
22
|
Zhang Z, Mu Y, Zhou H, Yao H, Wang DA. Cartilage Tissue Engineering in Practice: Preclinical Trials, Clinical Applications, and Prospects. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:473-490. [PMID: 36964757 DOI: 10.1089/ten.teb.2022.0190] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Articular cartilage defects significantly compromise the quality of life in the global population. Although many strategies are needed to repair articular cartilage, including microfracture, autologous osteochondral transplantation, and osteochondral allograft, the therapeutic effects remain suboptimal. In recent years, with the development of cartilage tissue engineering, scientists have continuously improved the formulations of therapeutic cells, biomaterial-based scaffolds, and biological factors, which have opened new avenues for better therapeutics of cartilage lesions. This review focuses on advances in cartilage tissue engineering, particularly in preclinical trials and clinical applications, prospects, and challenges.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, P.R. China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong SAR
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, P.R. China
| |
Collapse
|
23
|
Wang T, Dogru S, Dai Z, Kim SY, Vickers NA, Albro MB. Physiologic Doses of TGF-β Improve the Composition of Engineered Articular Cartilage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559554. [PMID: 37808691 PMCID: PMC10557735 DOI: 10.1101/2023.09.27.559554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
For cartilage regeneration applications, transforming growth factor beta (TGF-β) is conventionally administered at highly supraphysiologic doses (10-10,000 ng/mL) in an attempt to cue cells to fabricate neocartilage that matches the composition, structure, and functional properties of native hyaline cartilage. While supraphysiologic doses enhance ECM biosynthesis, they are also associated with inducing detrimental tissue features, such as fibrocartilage matrix deposition, pathologic-like chondrocyte clustering, and tissue swelling. Here we investigate the hypothesis that moderated TGF-β doses (0.1-1 ng/mL), akin to those present during physiological cartilage development, can improve neocartilage composition. Variable doses of media-supplemented TGF-β were administered to a model system of reduced-size cylindrical constructs (Ø2-Ø3 mm), which mitigate the TGF-β spatial gradients observed in conventional-size constructs (Ø4-Ø6 mm), allowing for a novel assessment of the intrinsic effect of TGF-β doses on macroscale neocartilage properties and composition. The administration of physiologic TGF-β to reduced-size constructs yields neocartilage with native-matched sGAG content and mechanical properties while providing a more hyaline cartilage-like composition, marked by: 1) reduced fibrocartilage-associated type I collagen, 2) 77% reduction in the fraction of cells present in a clustered morphology, and 3) 45% reduction in the degree of tissue swelling. Physiologic TGF-β appears to achieve an important balance of promoting requisite ECM biosynthesis, while mitigating hyaline cartilage compositional deficits. These results can guide the development of novel physiologic TGF-β-delivering scaffolds to improve the regeneration clinical-sized neocartilage tissues.
Collapse
|
24
|
Philippe V, Jeannerat A, Peneveyre C, Jaccoud S, Scaletta C, Hirt-Burri N, Abdel-Sayed P, Raffoul W, Darwiche S, Applegate LA, Martin R, Laurent A. Autologous and Allogeneic Cytotherapies for Large Knee (Osteo)Chondral Defects: Manufacturing Process Benchmarking and Parallel Functional Qualification. Pharmaceutics 2023; 15:2333. [PMID: 37765301 PMCID: PMC10536774 DOI: 10.3390/pharmaceutics15092333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Cytotherapies are often necessary for the management of symptomatic large knee (osteo)-chondral defects. While autologous chondrocyte implantation (ACI) has been clinically used for 30 years, allogeneic cells (clinical-grade FE002 primary chondroprogenitors) have been investigated in translational settings (Swiss progenitor cell transplantation program). The aim of this study was to comparatively assess autologous and allogeneic approaches (quality, safety, functional attributes) to cell-based knee chondrotherapies developed for clinical use. Protocol benchmarking from a manufacturing process and control viewpoint enabled us to highlight the respective advantages and risks. Safety data (telomerase and soft agarose colony formation assays, high passage cell senescence) and risk analyses were reported for the allogeneic FE002 cellular active substance in preparation for an autologous to allogeneic clinical protocol transposition. Validation results on autologous bioengineered grafts (autologous chondrocyte-bearing Chondro-Gide scaffolds) confirmed significant chondrogenic induction (COL2 and ACAN upregulation, extracellular matrix synthesis) after 2 weeks of co-culture. Allogeneic grafts (bearing FE002 primary chondroprogenitors) displayed comparable endpoint quality and functionality attributes. Parameters of translational relevance (transport medium, finished product suturability) were validated for the allogeneic protocol. Notably, the process-based benchmarking of both approaches highlighted the key advantages of allogeneic FE002 cell-bearing grafts (reduced cellular variability, enhanced process standardization, rationalized logistical and clinical pathways). Overall, this study built on our robust knowledge and local experience with ACI (long-term safety and efficacy), setting an appropriate standard for further clinical investigations into allogeneic progenitor cell-based orthopedic protocols.
Collapse
Affiliation(s)
- Virginie Philippe
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Annick Jeannerat
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| | - Cédric Peneveyre
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| | - Sandra Jaccoud
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- Laboratory of Biomechanical Orthopedics, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Corinne Scaletta
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- STI School of Engineering, Federal Polytechnic School of Lausanne, CH-1015 Lausanne, Switzerland
| | - Wassim Raffoul
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
| | - Salim Darwiche
- Musculoskeletal Research Unit, Vetsuisse Faculty, University of Zurich, CH-8057 Zurich, Switzerland;
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, CH-8057 Zurich, Switzerland
- Oxford OSCAR Suzhou Center, Oxford University, Suzhou 215123, China
| | - Robin Martin
- Orthopedics and Traumatology Service, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland;
| | - Alexis Laurent
- Regenerative Therapy Unit, Plastic, Reconstructive and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, CH-1066 Epalinges, Switzerland; (S.J.); (C.S.); (N.H.-B.); (P.A.-S.); (W.R.); (L.A.A.)
- Preclinical Research Department, LAM Biotechnologies SA, CH-1066 Epalinges, Switzerland; (A.J.); (C.P.)
| |
Collapse
|
25
|
Wang S, Chen H, Huang J, Shen S, Tang Z, Tan X, Lei D, Zhou G. Gelatin-modified 3D printed PGS elastic hierarchical porous scaffold for cartilage regeneration. APL Bioeng 2023; 7:036105. [PMID: 37547670 PMCID: PMC10404141 DOI: 10.1063/5.0152151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Regenerative cartilage replacements are increasingly required in clinical settings for various defect repairs, including bronchial cartilage deficiency, articular cartilage injury, and microtia reconstruction. Poly (glycerol sebacate) (PGS) is a widely used bioelastomer that has been developed for various regenerative medicine applications because of its excellent elasticity, biodegradability, and biocompatibility. However, because of inadequate active groups, strong hydrophobicity, and limited ink extrusion accuracy, 3D printed PGS scaffolds may cause insufficient bioactivity, inefficient cell inoculation, and inconsistent cellular composition, which seriously hinders its further cartilage regenerative application. Here, we combined 3D printed PGS frameworks with an encapsulated gelatin hydrogel to fabricate a PGS@Gel composite scaffold. PGS@Gel scaffolds have a controllable porous microstructure, with suitable pore sizes and enhanced hydrophilia, which could significantly promote the cells' penetration and adhesion for efficient chondrocyte inoculation. Furthermore, the outstanding elasticity and fatigue durability of the PGS framework enabled the regenerated cartilage built by the PGS@Gel scaffolds to resist the dynamic in vivo environment and maintain its original morphology. Importantly, PGS@Gel scaffolds increased the rate of cartilage regeneration concurrent with scaffold degradation. The scaffold was gradually degraded and integrated to form uniform, dense, and mature regenerated cartilage tissue with little scaffold residue.
Collapse
Affiliation(s)
| | | | | | - Sisi Shen
- Department of Plastic and Reconstructive Surgery, Department of Cardiology, Shanghai Key Lab of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Zhengya Tang
- Department of Plastic and Reconstructive Surgery, Department of Cardiology, Shanghai Key Lab of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Xiaoyan Tan
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, China
| | - Dong Lei
- Authors to whom correspondence should be addressed:; ; and
| | - Guangdong Zhou
- Authors to whom correspondence should be addressed:; ; and
| |
Collapse
|
26
|
Wen Y, Chen Y, Wu W, Zhang H, Peng Z, Yao X, Zhang X, Jiang W, Liao Y, Xie Y, Shen X, Sun H, Hu J, Liu H, Chen X, Chen J, Ouyang H. Hyperplastic Human Macromass Cartilage for Joint Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301833. [PMID: 37395375 PMCID: PMC10502860 DOI: 10.1002/advs.202301833] [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: 03/21/2023] [Revised: 06/07/2023] [Indexed: 07/04/2023]
Abstract
Cartilage damage affects millions of people worldwide. Tissue engineering strategies hold the promise to provide off-the-shelf cartilage analogs for tissue transplantation in cartilage repair. However, current strategies hardly generate sufficient grafts, as tissues cannot maintain size growth and cartilaginous phenotypes simultaneously. Herein, a step-wise strategy is developed for fabricating expandable human macromass cartilage (macro-cartilage) in a 3D condition by employing human polydactyly chondrocytes and a screen-defined serum-free customized culture (CC). CC-induced chondrocytes demonstrate improved cell plasticity, expressing chondrogenic biomarkers after a 14.59-times expansion. Crucially, CC-chondrocytes form large-size cartilage tissues with average diameters of 3.25 ± 0.05 mm, exhibiting abundant homogenous matrix and intact structure without a necrotic core. Compared with typical culture, the cell yield in CC increases 2.57 times, and the expression of cartilage marker collagen type II increases 4.70 times. Transcriptomics reveal that this step-wise culture drives a proliferation-to-differentiation process through an intermediate plastic stage, and CC-chondrocytes undergo a chondral lineage-specific differentiation with an activated metabolism. Animal studies show that CC macro-cartilage maintains a hyaline-like cartilage phenotype in vivo and significantly promotes the healing of large cartilage defects. Overall, an efficient expansion of human macro-cartilage with superior regenerative plasticity is achieved, providing a promising strategy for joint regeneration.
Collapse
|
27
|
Zhu J, Lun W, Feng Q, Cao X, Li Q. Mesenchymal stromal cells modulate YAP by verteporfin to mimic cartilage development and construct cartilage organoids based on decellularized matrix scaffolds. J Mater Chem B 2023; 11:7442-7453. [PMID: 37439116 DOI: 10.1039/d3tb01114c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The mechanical elasticity or stiffness of the ECM modulates YAP activity to regulate the differentiation of stem cells during the development and defect regeneration of cartilage tissue. However, the understanding of the scaffold-associated mechanobiology during the initiation of chondrogenesis and hyaline cartilaginous phenotype maintenance remains unclear. In order to elucidate such mechanisms to promote articular cartilage repair by producing more hyaline cartilage, we identify the relationship between YAP subcellular localization and variation of the cartilage structure and organization during the early postnatal explosive growth in incipient articular cartilage. Next, we prepared a decellularized cartilage scaffold with different stiffness (2-33 kPa) to investigate the effect of scaffold stiffness on the formation of hyaline cartilage by mesenchymal stem cells and the change of YAP activity. Furthermore, we simulated the decrease of cellular YAP activity during postnatal cartilage development by inhibiting YAP activity with verteporfin, and realized that the timing of drug incorporation was critical to regulate the differentiation of MSCs to hyaline chondrocytes and inhibit their hypertrophy and fibrosis. On this basis, we constructed hyaline cartilage organoids by decellularized matrix scaffolds. Collectively, the results herein demonstrate that YAP plays a critical role during in vitro chondrogenic differentiation which is tightly regulated by biochemical and mechanical regulation.
Collapse
Affiliation(s)
- Jiayi Zhu
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China.
| | - Wanqing Lun
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China.
| | - Qi Feng
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China.
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Xiaodong Cao
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China.
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Qingtao Li
- School of Medicine, South China University of Technology, Guangzhou 510006, P. R. China.
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou 510006, P. R. China.
| |
Collapse
|
28
|
Shi W, Meng Q, Hu X, Cheng J, Shao Z, Yang Y, Ao Y. Using a Xenogeneic Acellular Dermal Matrix Membrane to Enhance the Reparability of Bone Marrow Mesenchymal Stem Cells for Cartilage Injury. Bioengineering (Basel) 2023; 10:916. [PMID: 37627801 PMCID: PMC10451227 DOI: 10.3390/bioengineering10080916] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Due to its avascular organization and low mitotic ability, articular cartilage possesses limited intrinsic regenerative capabilities. The aim of this study is to achieve one-step cartilage repair in situ via combining bone marrow stem cells (BMSCs) with a xenogeneic Acellular dermal matrix (ADM) membrane. The ADM membranes were harvested from Sprague-Dawley (SD) rats through standard decellularization procedures. The characterization of the scaffolds was measured, including the morphology and physical properties of the ADM membrane. The in vitro experiments included the cell distribution, chondrogenic matrix quantification, and viability evaluation of the scaffolds. Adult male New Zealand white rabbits were used for the in vivo evaluation. Isolated microfracture was performed in the control (MF group) in the left knee and the tested ADM group was included as an experimental group when an ADM scaffold was implanted through matching with the defect after microfracture in the right knee. At 6, 12, and 24 weeks post-surgery, the rabbits were sacrificed for further research. The ADM could adsorb water and had excellent porosity. The bone marrow stem cells (BMSCs) grew well when seeded on the ADM scaffold, demonstrating a characteristic spindle-shaped morphology. The ADM group exhibited an excellent proliferative capacity as well as the cartilaginous matrix and collagen production of the BMSCs. In the rabbit model, the ADM group showed earlier filling, more hyaline-like neo-tissue formation, and better interfacial integration between the defects and normal cartilage compared with the microfracture (MF) group at 6, 12, and 24 weeks post-surgery. In addition, neither intra-articular inflammation nor a rejection reaction was observed after the implantation of the ADM scaffold. This study provides a promising biomaterial-based strategy for cartilage repair and is worth further investigation in large animal models.
Collapse
Affiliation(s)
- Weili Shi
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (W.S.); (Q.M.); (X.H.); (J.C.); (Z.S.)
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (W.S.); (Q.M.); (X.H.); (J.C.); (Z.S.)
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (W.S.); (Q.M.); (X.H.); (J.C.); (Z.S.)
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (W.S.); (Q.M.); (X.H.); (J.C.); (Z.S.)
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Zhenxing Shao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (W.S.); (Q.M.); (X.H.); (J.C.); (Z.S.)
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Yuping Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (W.S.); (Q.M.); (X.H.); (J.C.); (Z.S.)
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (W.S.); (Q.M.); (X.H.); (J.C.); (Z.S.)
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
29
|
Vágó J, Takács R, Kovács P, Hajdú T, van der Veen DR, Matta C. Combining biomechanical stimulation and chronobiology: a novel approach for augmented chondrogenesis? Front Bioeng Biotechnol 2023; 11:1232465. [PMID: 37456723 PMCID: PMC10349586 DOI: 10.3389/fbioe.2023.1232465] [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: 05/31/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
The unique structure and composition of articular cartilage is critical for its physiological function. However, this architecture may get disrupted by degeneration or trauma. Due to the low intrinsic regeneration properties of the tissue, the healing response is generally poor. Low-grade inflammation in patients with osteoarthritis advances cartilage degradation, resulting in pain, immobility, and reduced quality of life. Generating neocartilage using advanced tissue engineering approaches may address these limitations. The biocompatible microenvironment that is suitable for cartilage regeneration may not only rely on cells and scaffolds, but also on the spatial and temporal features of biomechanics. Cell-autonomous biological clocks that generate circadian rhythms in chondrocytes are generally accepted to be indispensable for normal cartilage homeostasis. While the molecular details of the circadian clockwork are increasingly well understood at the cellular level, the mechanisms that enable clock entrainment by biomechanical signals, which are highly relevant in cartilage, are still largely unknown. This narrative review outlines the role of the biomechanical microenvironment to advance cartilage tissue engineering via entraining the molecular circadian clockwork, and highlights how application of this concept may enhance the development and successful translation of biomechanically relevant tissue engineering interventions.
Collapse
Affiliation(s)
- Judit Vágó
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Roland Takács
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Patrik Kovács
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Tibor Hajdú
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| | - Daan R. van der Veen
- Chronobiology Section, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Csaba Matta
- Department of Anatomy, Faculty of Medicine, Histology and Embryology, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
30
|
Semerci Sevimli T, Sevimli M, Qomi Ekenel E, Altuğ Tasa B, Nur Soykan M, Demir Güçlüer Z, İnan U, Uysal O, Güneş Bağış S, Çemrek F, Eker Sarıboyacı A. Comparison of exosomes secreted by synovial fluid-derived mesenchymal stem cells and adipose tissue-derived mesenchymal stem cells in culture for microRNA-127-5p expression during chondrogenesis. Gene 2023; 865:147337. [PMID: 36878417 DOI: 10.1016/j.gene.2023.147337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/14/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
This study aimed to investigate the differences between the exosomal microRNA-127-5p expression profiles of human adipose tissue-derived mesenchymal stem cells (hAT-MSCs) and human synovial fluid-derived mesenchymal stem cells (hSF-MSCs) during chondrogenesis in terms of regenerative treatment of cartilage. Synovial fluid-derived mesenchymal stem cells, adipose tissue-derived mesenchymal stem cells, and human fetal chondroblast cells (hfCCs) were directed to chondrogenic differentiation. Alcian Blue and Safranin O stainings were performed to detect chondrogenic differentiation histochemically. Exosomes derived from chondrogenic differentiated cells and their exosomes were isolated and characterized. microRNA-127-5p expressions were measured by Quantitative reverse transcription PCR (qRT-PCR). Significantly higher levels of microRNA-127-5p expression in differentiated hAT-MSCs exosomes, similar to human fetal chondroblast cells, which are the control group in the chondrogenic differentiation process, were observed. hAT-MSCs are better sources of microRNA-127-5p than hSF-MSCs for stimulating chondrogenesis or in the regenerative therapy of cartilage-related pathologies. hAT-MSCs exosomes are rich sources of microRNA-127-5p and can be an essential candidate for cartilage regeneration treatments.
Collapse
Affiliation(s)
- Tuğba Semerci Sevimli
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Murat Sevimli
- Department of Histology and Embryology, Faculty of Medicine, Suleyman Demirel University, 32260 Isparta, Turkey.
| | - Emilia Qomi Ekenel
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Burcugül Altuğ Tasa
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Merve Nur Soykan
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Zilif Demir Güçlüer
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Ulukan İnan
- Department of Orthopedics and Traumatology, Faculty of Medicine, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Onur Uysal
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Sibel Güneş Bağış
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Fatih Çemrek
- Department of Statistics, Faculty of Science and Letters, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| | - Ayla Eker Sarıboyacı
- Department of Stem Cell, Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, 26040 Eskisehir, Turkey; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, 26040 Eskisehir, Turkey.
| |
Collapse
|
31
|
Płończak M, Wasyłeczko M, Jakutowicz T, Chwojnowski A, Czubak J. Intraarticular Implantation of Autologous Chondrocytes Placed on Collagen or Polyethersulfone Scaffolds: An Experimental Study in Rabbits. Polymers (Basel) 2023; 15:polym15102360. [PMID: 37242936 DOI: 10.3390/polym15102360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Hyaline cartilage has very limited repair capability and cannot be rebuilt predictably using conventional treatments. This study presents Autologous Chondrocyte Implantation (ACI) on two different scaffolds for the treatment of lesions in hyaline cartilage in rabbits. The first one is a commercially available scaffold (Chondro-Gide) made of collagen type I/III and the second one is a polyethersulfone (PES) synthetic membrane, manufactured by phase inversion. The revolutionary idea in the present study is the fact that we used PES membranes, which have unique features and benefits that are desirable for the 3D cultivation of chondrocytes. Sixty-four White New Zealand rabbits were used in this research. Defects penetrating into the subchondral bone were filled with or without the placement of chondrocytes on collagen or PES membranes after two weeks of culture. The expression of the gene encoding type II procollagen, a molecular marker of chondrocytes, was evaluated. Elemental analysis was performed to estimate the weight of tissue grown on the PES membrane. The reparative tissue was analyzed macroscopically and histologically after surgery at 12, 25, and 52 weeks. RT-PCR analysis of the mRNA isolated from cells detached from the polysulphonic membrane revealed the expression of type II procollagen. The elementary analysis of polysulphonic membrane slices after 2 weeks of culture with chondrocytes revealed a concentration of 0.23 mg of tissue on one part of the membrane. Macroscopic and microscopic evaluation indicated that the quality of regenerated tissue was similar after the transplantation of cells placed on polysulphonic or collagen membranes. The established method for the culture and transplantation of chondrocytes placed on polysulphonic membranes resulted in the growth of the regenerated tissue, revealing the morphology of hyaline-like cartilage to be of similar quality to collagen membranes.
Collapse
Affiliation(s)
- Maciej Płończak
- Mazovia Regional Hospital John Paul II, 08-110 Siedlce, Poland
| | - Monika Wasyłeczko
- Nałęcz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Tomasz Jakutowicz
- Department of Neurosurgery and Children Traumatology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nałęcz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Jarosław Czubak
- Department of Orthopedics, Pediatric Orthopedics and Traumatology, Centre of Postgraduate Medical Education, Gruca Orthopaedic and Trauma Teaching Hospital, 05-402 Otwock, Poland
| |
Collapse
|
32
|
Xiao F, Zheng P, Tang J, Huang X, Kang W, Zhou G, Sun K. Cartilage-bioinspired, tough and lubricated hydrogel based on nanocomposite enhancement effect. J Mater Chem B 2023; 11:4763-4775. [PMID: 37183499 DOI: 10.1039/d3tb00364g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The maintenance of high load-bearing tissues and joint lubrication is essential for suppressing osteoarthritis. The lubrication of natural joints is mainly attributed to the hydration lubrication mechanism of articular cartilage. Phospholipids on the cartilage surface attract water molecules to form a tough hydrated layer to reduce friction. In this work, inspired by the phosphatidylcholine lipids, we synthesized lubricated nanospheres by grafting hydrophilic polymer brushes and further synthesized a nanocomposite hydrogel. The addition of the lubricated nanospheres enhanced both the mechanical and lubricated properties of the hydrogel. The nanocomposite-lubricated hydrogel exhibited a friction coefficient 81.7% lower than the blank hydrogel because of grafting the polymer brushes. Also, the nanocomposite enhancement helped the hydrogel achieve high mechanical properties with a compressive strength of 6.63 MPa (50%). The nanocomposite hydrogel developed here could be a promising candidate material in bionic articular cartilage substitute materials.
Collapse
Affiliation(s)
- Fen Xiao
- Hunan Key Laboratory of Biomedical Nanometer and Device, Hunan University of Technology, Zhuzhou 412007, P. R. China.
| | - Pengshuo Zheng
- Hunan Key Laboratory of Biomedical Nanometer and Device, Hunan University of Technology, Zhuzhou 412007, P. R. China.
| | - Jianxin Tang
- Hunan Key Laboratory of Biomedical Nanometer and Device, Hunan University of Technology, Zhuzhou 412007, P. R. China.
| | - Xin Huang
- Hunan Key Laboratory of Biomedical Nanometer and Device, Hunan University of Technology, Zhuzhou 412007, P. R. China.
| | - Wenji Kang
- Hunan Key Laboratory of Biomedical Nanometer and Device, Hunan University of Technology, Zhuzhou 412007, P. R. China.
| | - Guiyin Zhou
- Hunan Key Laboratory of Biomedical Nanometer and Device, Hunan University of Technology, Zhuzhou 412007, P. R. China.
- School of Physics and Electronics, Central South University, Changsha 410083, China
| | - Kehui Sun
- School of Physics and Electronics, Central South University, Changsha 410083, China
| |
Collapse
|
33
|
Liu Z, Luo Z, Yu H, Zhao E, Guo J, Mou P, Chen A, Chen J, Zhou Z, Zeng W. Near-infrared light-controlled kartogenin delivery of multifunctional Prussian blue nanocomposites for cartilage defect repair. NANOSCALE 2023; 15:9076-9093. [PMID: 37129436 DOI: 10.1039/d3nr00205e] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Articular cartilage injury repair remains a challenge for clinicians and researchers. Mesenchymal stem cells (MSCs) have multiple differentiation potentials and can be induced to differentiate into the chondrogenic lineage for cartilage defect repair; however, the insufficient capacity of chondrogenic differentiation and excess reactive oxygen species (ROS)-mediated oxidative stress, which always lead to differentiation into hypertrophic chondrocytes, still need to be resolved. Accordingly, kartogenin (KGN), which can promote chondrogenic differentiation of MSCs, has shown promise in promoting infected cartilage repair. However, realizing controllable release to prolong its action time and avoid hypertrophic differentiation is critical. We herein developed a mesoporous Prussian blue nanoparticle (mPB)-based near-infrared (NIR) light-responsive controlled nanosystem. KGN was encapsulated in temperature-stimulated responsive phase change materials (PCMs), which were used as excellent gating materials (KGN-PCM@mPBs). In addition, the mPBs could efficiently scavenge ROS by their enzyme-like antioxidative activities. Our study demonstrates that the nanocomposites could efficiently promote chondrogenic differentiation and successfully inhibit the hypertrophic differentiation of MSCs. By intra-articular injection of KGN-PCM@mPBs and NIR-triggered precisely controlled release, satisfactory cartilage repair effects can be achieved in a rat chondral defect model. Thus, this constructed NIR-mediated KGN-PCM@mPB nanoplatform may represent an effective cartilage repair strategy with satisfactory biosafety in clinical applications.
Collapse
Affiliation(s)
- Zunhan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, the First Affiliated Hospital of the Army Military Medical University, Chongqing, 400038, China
| | - Zhenyu Luo
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Haoda Yu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Enze Zhao
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Junfeng Guo
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, the First Affiliated Hospital of the Army Military Medical University, Chongqing, 400038, China
| | - Ping Mou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Anjing Chen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Jiali Chen
- West China School of Nursing, Sichuan University/Department of Orthopedics, West China Hospital, Sichuan University Chengdu, 610041, P.R. China
| | - Zongke Zhou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Weinan Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
34
|
Huo Y, Bai B, Zheng R, Sun Y, Yu Y, Wang X, Chen H, Hua Y, Zhang Y, Zhou G, Wang X. In Vivo Stable Allogenic Cartilage Regeneration in a Goat Model Based on Immunoisolation Strategy Using Electrospun Semipermeable Membranes. Adv Healthc Mater 2023; 12:e2203084. [PMID: 36789972 PMCID: PMC11469122 DOI: 10.1002/adhm.202203084] [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: 11/28/2022] [Revised: 01/13/2023] [Indexed: 02/16/2023]
Abstract
Tissue engineering is a promising strategy for cartilage defect repair. However, autologous cartilage regeneration is limited by additional trauma to the donor site and a long in vitro culture period. Alternatively, allogenic cartilage regeneration has attracted attention because of the unique advantages of an abundant donor source and immediate supply, but it will cause immune rejection responses (IRRs), especially in immunocompetent large animals. Therefore, a universal technique needs to be established to overcome IRRs for allogenic cartilage regeneration in large animals. In the current study, a hybrid synthetic-natural electrospun thermoplastic polyurethane/gelatin (TPU/GT) semipermeable membrane to explore the feasibility of stable allogenic cartilage regeneration by an immunoisolation strategy is developed. In vitro results demonstrated that the rationally designed electrospun TPU/GT membranes has ideal biocompatibility, semipermeability, and an immunoisolation function. In vivo results further showed that the semipermeable membrane (SPM) efficiently blocked immune cell attack, decreased immune factor production, and cell apoptosis of the regenerated allogenic cartilage. Importantly, TPU/GT-encapsulated cartilage-sheet constructs achieved stable allogeneic cartilage regeneration in a goat model. The current study provides a novel strategy for allogenic cartilage regeneration and supplies a new cartilage donor source to repair various cartilage defects.
Collapse
Affiliation(s)
- Yingying Huo
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Key Laboratory of Tissue EngineeringShanghai200011PR China
- National Tissue Engineering Center of ChinaShanghai200241PR China
| | - Baoshuai Bai
- Research Institute of Plastic SurgeryWeifang Medical UniversityWeifangShandong261053PR China
| | - Rui Zheng
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Key Laboratory of Tissue EngineeringShanghai200011PR China
- National Tissue Engineering Center of ChinaShanghai200241PR China
| | - Yuyan Sun
- Research Institute of Plastic SurgeryWeifang Medical UniversityWeifangShandong261053PR China
| | - Yao Yu
- Research Institute of Plastic SurgeryWeifang Medical UniversityWeifangShandong261053PR China
| | - Xin Wang
- Department of Plastic SurgeryTongren HospitalShanghai Jiao Tong University School of MedicineShanghai Key Laboratory of Tissue EngineeringShanghai200050PR China
- Department of Hand SurgeryNingbo Sixth HospitalNingboZhejiang315042PR China
| | - Hong Chen
- Department of Hand SurgeryNingbo Sixth HospitalNingboZhejiang315042PR China
| | - Yujie Hua
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Key Laboratory of Tissue EngineeringShanghai200011PR China
- National Tissue Engineering Center of ChinaShanghai200241PR China
- Institute of Regenerative Medicine and OrthopedicsInstitutes of Health Central PlainXinxiang Medical UniversityXinxiangHenan453003PR China
| | - Yixin Zhang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Key Laboratory of Tissue EngineeringShanghai200011PR China
- National Tissue Engineering Center of ChinaShanghai200241PR China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai Key Laboratory of Tissue EngineeringShanghai200011PR China
- National Tissue Engineering Center of ChinaShanghai200241PR China
- Research Institute of Plastic SurgeryWeifang Medical UniversityWeifangShandong261053PR China
- Institute of Regenerative Medicine and OrthopedicsInstitutes of Health Central PlainXinxiang Medical UniversityXinxiangHenan453003PR China
| | - Xiaoyun Wang
- Department of Plastic SurgeryTongren HospitalShanghai Jiao Tong University School of MedicineShanghai Key Laboratory of Tissue EngineeringShanghai200050PR China
- Department of Hand SurgeryNingbo Sixth HospitalNingboZhejiang315042PR China
| |
Collapse
|
35
|
Arhebamen EP, Teodoro MT, Blonka AB, Matthew HWT. Long-Term Culture Performance of a Polyelectrolyte Complex Microcapsule Platform for Hyaline Cartilage Repair. Bioengineering (Basel) 2023; 10:bioengineering10040467. [PMID: 37106654 PMCID: PMC10135885 DOI: 10.3390/bioengineering10040467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Articular cartilage (AC) tissue repair and regeneration remains an ongoing challenge. One component of the challenge is the limited ability to scale an engineered cartilage graft to clinically relevant sizes while maintaining uniform properties. In this paper, we report on the evaluation of our polyelectrolyte complex microcapsule (PECM) platform technology as a technique for generating cartilage-like spherical modules. Bone marrow-derived mesenchymal stem cells (bMSCs) or primary articular chondrocytes were encapsulated within PECMs composed of methacrylated hyaluronan, collagen I, and chitosan. The formation of cartilage-like tissue in the PECMs over a 90-day culture was characterized. The results showed that chondrocytes exhibited superior growth and matrix deposition compared to either chondrogenically-induced bMSCs or a mixed PECM culture containing both chondrocytes and bMSCs. The chondrocyte-generated matrix filled the PECM and produced substantial increases in capsule compressive strength. The PECM system thus appears to support intracapsular cartilage tissue formation and the capsule approach promotes efficient culture and handling of these micro tissues. Since previous studies have proven the feasibility of fusing such capsules into large tissue constructs, the results suggest that encapsulating primary chondrocytes in PECM modules may be a viable route toward achieving a functional articular cartilage graft.
Collapse
Affiliation(s)
- Ehinor P Arhebamen
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| | - Maria T Teodoro
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| | - Amelia B Blonka
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| | - Howard W T Matthew
- Department of Biomedical Engineering, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
- Department of Chemical Engineering and Materials Science, Wayne State University, 5050 Anthony Wayne Dr., Detroit, MI 48202, USA
| |
Collapse
|
36
|
Wasyłeczko M, Remiszewska E, Sikorska W, Dulnik J, Chwojnowski A. Scaffolds for Cartilage Tissue Engineering from a Blend of Polyethersulfone and Polyurethane Polymers. Molecules 2023; 28:molecules28073195. [PMID: 37049957 PMCID: PMC10095814 DOI: 10.3390/molecules28073195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
In recent years, one of the main goals of cartilage tissue engineering has been to find appropriate scaffolds for hyaline cartilage regeneration, which could serve as a matrix for chondrocytes or stem cell cultures. The study presents three types of scaffolds obtained from a blend of polyethersulfone (PES) and polyurethane (PUR) by a combination of wet-phase inversion and salt-leaching methods. The nonwovens made of gelatin and sodium chloride (NaCl) were used as precursors of macropores. Thus, obtained membranes were characterized by a suitable structure. The top layers were perforated, with pores over 20 µm, which allows cells to enter the membrane. The use of a nonwoven made it possible to develop a three-dimensional network of interconnected macropores that is required for cell activity and mobility. Examination of wettability (contact angle, swelling ratio) showed a hydrophilic nature of scaffolds. The mechanical test showed that the scaffolds were suitable for knee joint applications (stress above 10 MPa). Next, the scaffolds underwent a degradation study in simulated body fluid (SBF). Weight loss after four weeks and changes in structure were assessed using scanning electron microscopy (SEM) and MeMoExplorer Software, a program that estimates the size of pores. The porosity measurements after degradation confirmed an increase in pore size, as expected. Hydrolysis was confirmed by Fourier-transform infrared spectroscopy (FT-IR) analysis, where the disappearance of ester bonds at about 1730 cm−1 wavelength is noticeable after degradation. The obtained results showed that the scaffolds meet the requirements for cartilage tissue engineering membranes and should undergo further testing on an animal model.
Collapse
Affiliation(s)
- Monika Wasyłeczko
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| | - Elżbieta Remiszewska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| | - Wioleta Sikorska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| | - Judyta Dulnik
- Institute of Fundamental Technological Research Polish Academy of Sciences, Laboratory of Polymers and Biomaterials, Pawińskiego 5b, 02-106 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4, 02-109 Warsaw, Poland
| |
Collapse
|
37
|
Márquez-Flórez K, Garzón-Alvarado DA, Carda C, Sancho-Tello M. Computational model of articular cartilage regeneration induced by scaffold implantation in vivo. J Theor Biol 2023; 561:111393. [PMID: 36572091 DOI: 10.1016/j.jtbi.2022.111393] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/22/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Computational models allow to explain phenomena that cannot be observed through an animal model, such as the strain and stress states which can highly influence regeneration of the tissue. For this purpose, we have developed a simulation tool to determine the mechanical conditions provided by the polymeric scaffold. The computational model considered the articular cartilage, the subchondral bone, and the scaffold. All materials were modeled as poroelastic, and the cartilage had linear-elastic oriented collagen fibers. This model was able to explain the remodeling process that subchondral bone goes through, and how the scaffold allowed the conditions for cartilage regeneration. These results suggest that the use of scaffolds might lead the cartilaginous tissue growth in vivo by providing a better mechanical environment. Moreover, the developed computational model demonstrated to be useful as a tool prior experimental in vivo studies, by predicting the possible outcome of newly proposed treatments allowing to discard approaches that might not bring good results.
Collapse
Affiliation(s)
- K Márquez-Flórez
- Department of Mechanical and Mechatronic Engineering, Universidad Nacional de Colombia, Bogotá, Colombia; Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia; Department of Pathology, Faculty of Medicine and Odontology, Universitat de València, Valencia, Spain
| | - D A Garzón-Alvarado
- Department of Mechanical and Mechatronic Engineering, Universidad Nacional de Colombia, Bogotá, Colombia; Numerical Methods and Modeling Research Group (GNUM), Universidad Nacional de Colombia, Bogotá, Colombia; Instituto de Biotecnología, Universidad Nacional de Colombia.
| | - C Carda
- Department of Pathology, Faculty of Medicine and Odontology, Universitat de València, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain; Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - M Sancho-Tello
- Department of Pathology, Faculty of Medicine and Odontology, Universitat de València, Valencia, Spain; INCLIVA Biomedical Research Institute, Valencia, Spain
| |
Collapse
|
38
|
Yang G, Li X, Zhang W, Wu N, Chen H, Liu X, Jiang H. Quantitative T2 mapping monitoring the maturation of engineered elastic cartilage in a rabbit model. BMC Med Imaging 2023; 23:36. [PMID: 36879206 PMCID: PMC9987110 DOI: 10.1186/s12880-023-00985-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 02/03/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Cartilage tissue engineering provides a promising approach to reconstruct craniofacial defects, and a noninvasive method is needed to assess its effectiveness. Although magnetic resonance imaging (MRI) has been used to evaluate articular cartilage in vivo, few studies focused on its feasibility in monitoring engineered elastic cartilage (EC). METHODS Auricular cartilage, silk fibroin (SF) scaffold, and EC consisting of rabbit auricular chondrocytes and SF scaffold were transplanted subcutaneously into the rabbit back. In eight weeks after transplantation, grafts were imaged by MRI using PROSET, PDW VISTA SPAIR, 3D T2 VISTA, 2D MIXED T2 Multislice, and SAG TE multiecho sequences, followed by histological examination and biochemical analysis. Statistical analyses were performed to identify the association between T2 values and biochemical indicator values of EC. RESULTS In vivo imaging shows that 2D MIXED T2 Multislice sequence (T2 mapping) clearly distinguished the native cartilage, engineered cartilage and fibrous tissue. T2 values showed high correlations with cartilage-specific biochemical parameters at different time points, especially the elastic cartilage specific protein elastin (ELN, r= -0.939, P < 0.001). CONCLUSION Quantitative T2 mapping can effectively detect the in vivo maturity of engineered elastic cartilage after subcutaneously transplantation. This study would promote the clinical application of MRI T2 mapping in monitoring engineered elastic cartilage in the repair of craniofacial defects.
Collapse
Affiliation(s)
- Guojun Yang
- Auricular Plastic and Reconstructive Surgery Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, 100144, Beijing, People's Republic of China
| | - Xue Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dongdan Santiao, Dongcheng District, 100005, Beijing, People's Republic of China
| | - Weiwei Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, 5 Dongdan Santiao, Dongcheng District, 100005, Beijing, People's Republic of China
| | - Nier Wu
- Department of Biomedical Engineering, College of Engineering, Peking University, 5 Yiheyuan Road, Haidian District, 100871, Beijing, People's Republic of China
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Engineering, Peking University, 5 Yiheyuan Road, Haidian District, 100871, Beijing, People's Republic of China
| | - Xia Liu
- Research Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, 100144, Beijing, People's Republic of China.
| | - Haiyue Jiang
- Auricular Plastic and Reconstructive Surgery Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 33 Badachu Road, Shijingshan District, 100144, Beijing, People's Republic of China.
| |
Collapse
|
39
|
Zhang G, Qi C, Rong L, Ju C. Intra-articular delivery of antioxidative polymer-based nanospheres reduces intracellular reactive oxygen species in macrophages and alleviates cartilage damage in a rat model. J Biomater Appl 2023; 37:1384-1392. [PMID: 36342066 DOI: 10.1177/08853282221137774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cartilage damage is frequent in various joint diseases, mainly manifested by the loss of type II collagen and the degradation of proteoglycans. Diclofenac sodium is a commonly used drug for the treatment of joint diseases, but simple administration is often affected by drug clearance and rapid metabolism. Intra-articular drug delivery is an effective method for local enrichment of high concentration of drugs. However, due to the short half-life of diclofenac sodium, prolonging the stability and duration of the drug can alleviate the disadvantages of direct intra-articular application. Nanospheres for delivering drugs to treat joint diseases could be a remedy for cartilage damage. In addition, excessive production of reactive oxygen species (ROS) by macrophages activated in damaged cartilage would aggravate cartilage damage. Therefore, this study intends to use poly lactic-co-glycolic acid nanospheres to load and deliver diclofenac sodium to inhibit chondrocyte death while regulating the generation of ROS, thereby promoting the treatment of cartilage damage.
Collapse
Affiliation(s)
- Guihua Zhang
- Department of Bone and Joint Surgery, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| | - Chaopeng Qi
- Department of Hand and Microsurgery, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| | - Lingyan Rong
- Department of Pharmacy, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| | - Chuanbao Ju
- Department of Emergency Trauma, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| |
Collapse
|
40
|
Kim Y, Zharkinbekov Z, Raziyeva K, Tabyldiyeva L, Berikova K, Zhumagul D, Temirkhanova K, Saparov A. Chitosan-Based Biomaterials for Tissue Regeneration. Pharmaceutics 2023; 15:pharmaceutics15030807. [PMID: 36986668 PMCID: PMC10055885 DOI: 10.3390/pharmaceutics15030807] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Chitosan is a chitin-derived biopolymer that has shown great potential for tissue regeneration and controlled drug delivery. It has numerous qualities that make it attractive for biomedical applications such as biocompatibility, low toxicity, broad-spectrum antimicrobial activity, and many others. Importantly, chitosan can be fabricated into a variety of structures including nanoparticles, scaffolds, hydrogels, and membranes, which can be tailored to deliver a desirable outcome. Composite chitosan-based biomaterials have been demonstrated to stimulate in vivo regeneration and the repair of various tissues and organs, including but not limited to, bone, cartilage, dental, skin, nerve, cardiac, and other tissues. Specifically, de novo tissue formation, resident stem cell differentiation, and extracellular matrix reconstruction were observed in multiple preclinical models of different tissue injuries upon treatment with chitosan-based formulations. Moreover, chitosan structures have been proven to be efficient carriers for medications, genes, and bioactive compounds since they can maintain the sustained release of these therapeutics. In this review, we discuss the most recently published applications of chitosan-based biomaterials for different tissue and organ regeneration as well as the delivery of various therapeutics.
Collapse
|
41
|
Sun W, Gregory DA, Zhao X. Designed peptide amphiphiles as scaffolds for tissue engineering. Adv Colloid Interface Sci 2023; 314:102866. [PMID: 36898186 DOI: 10.1016/j.cis.2023.102866] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023]
Abstract
Peptide amphiphiles (PAs) are peptide-based molecules that contain a peptide sequence as a head group covalently conjugated to a hydrophobic segment, such as lipid tails. They can self-assemble into well-ordered supramolecular nanostructures such as micelles, vesicles, twisted ribbons and nanofibers. In addition, the diversity of natural amino acids gives the possibility to produce PAs with different sequences. These properties along with their biocompatibility, biodegradability and a high resemblance to native extracellular matrix (ECM) have resulted in PAs being considered as ideal scaffold materials for tissue engineering (TE) applications. This review introduces the 20 natural canonical amino acids as building blocks followed by highlighting the three categories of PAs: amphiphilic peptides, lipidated peptide amphiphiles and supramolecular peptide amphiphile conjugates, as well as their design rules that dictate the peptide self-assembly process. Furthermore, 3D bio-fabrication strategies of PAs hydrogels are discussed and the recent advances of PA-based scaffolds in TE with the emphasis on bone, cartilage and neural tissue regeneration both in vitro and in vivo are considered. Finally, future prospects and challenges are discussed.
Collapse
Affiliation(s)
- Weizhen Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK
| | - David Alexander Gregory
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK; Department of Material Science and Engineering, University of Sheffield, Sheffield S3 7HQ, UK
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| |
Collapse
|
42
|
Guo X, Ma Y, Min Y, Sun J, Shi X, Gao G, Sun L, Wang J. Progress and prospect of technical and regulatory challenges on tissue-engineered cartilage as therapeutic combination product. Bioact Mater 2023; 20:501-518. [PMID: 35846847 PMCID: PMC9253051 DOI: 10.1016/j.bioactmat.2022.06.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/19/2022] [Accepted: 06/19/2022] [Indexed: 12/18/2022] Open
Abstract
Hyaline cartilage plays a critical role in maintaining joint function and pain. However, the lack of blood supply, nerves, and lymphatic vessels greatly limited the self-repair and regeneration of damaged cartilage, giving rise to various tricky issues in medicine. In the past 30 years, numerous treatment techniques and commercial products have been developed and practiced in the clinic for promoting defected cartilage repair and regeneration. Here, the current therapies and their relevant advantages and disadvantages will be summarized, particularly the tissue engineering strategies. Furthermore, the fabrication of tissue-engineered cartilage under research or in the clinic was discussed based on the traid of tissue engineering, that is the materials, seed cells, and bioactive factors. Finally, the commercialized cartilage repair products were listed and the regulatory issues and challenges of tissue-engineered cartilage repair products and clinical application would be reviewed. Tissue engineered cartilage, a promising strategy for articular cartilage repair. Nearly 20 engineered cartilage repair products in clinic based on clinical techniques. Combination product, the classification of tissue-engineered cartilage. Key regulatory compliance issues for combination products.
Collapse
Affiliation(s)
- Xiaolei Guo
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
- Corresponding author.
| | - Yuan Ma
- State Key Laboratory of Tribology, Tsinghua University, Beijing, PR China
| | - Yue Min
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Jiayi Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Xinli Shi
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
- Corresponding author. Center for Medical Device Evaluation, National Medical Products Administration, Beijing, 100081, PR China
| | - Guobiao Gao
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Lei Sun
- Center for Medical Device Evaluation, National Medical Products Administration, Beijing, PR China
| | - Jiadao Wang
- State Key Laboratory of Tribology, Tsinghua University, Beijing, PR China
- Corresponding author. State Key Laboratory of Tribology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
43
|
Tolabi H, Davari N, Khajehmohammadi M, Malektaj H, Nazemi K, Vahedi S, Ghalandari B, Reis RL, Ghorbani F, Oliveira JM. Progress of Microfluidic Hydrogel-Based Scaffolds and Organ-on-Chips for the Cartilage Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2208852. [PMID: 36633376 DOI: 10.1002/adma.202208852] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/09/2022] [Indexed: 05/09/2023]
Abstract
Cartilage degeneration is among the fundamental reasons behind disability and pain across the globe. Numerous approaches have been employed to treat cartilage diseases. Nevertheless, none have shown acceptable outcomes in the long run. In this regard, the convergence of tissue engineering and microfabrication principles can allow developing more advanced microfluidic technologies, thus offering attractive alternatives to current treatments and traditional constructs used in tissue engineering applications. Herein, the current developments involving microfluidic hydrogel-based scaffolds, promising structures for cartilage regeneration, ranging from hydrogels with microfluidic channels to hydrogels prepared by the microfluidic devices, that enable therapeutic delivery of cells, drugs, and growth factors, as well as cartilage-related organ-on-chips are reviewed. Thereafter, cartilage anatomy and types of damages, and present treatment options are briefly overviewed. Various hydrogels are introduced, and the advantages of microfluidic hydrogel-based scaffolds over traditional hydrogels are thoroughly discussed. Furthermore, available technologies for fabricating microfluidic hydrogel-based scaffolds and microfluidic chips are presented. The preclinical and clinical applications of microfluidic hydrogel-based scaffolds in cartilage regeneration and the development of cartilage-related microfluidic chips over time are further explained. The current developments, recent key challenges, and attractive prospects that should be considered so as to develop microfluidic systems in cartilage repair are highlighted.
Collapse
Affiliation(s)
- Hamidreza Tolabi
- New Technologies Research Center (NTRC), Amirkabir University of Technology, Tehran, 15875-4413, Iran
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, 15875-4413, Iran
| | - Niyousha Davari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, 143951561, Iran
| | - Mehran Khajehmohammadi
- Department of Mechanical Engineering, Faculty of Engineering, Yazd University, Yazd, 89195-741, Iran
- Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, 8916877391, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg, 9220, Denmark
| | - Katayoun Nazemi
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Samaneh Vahedi
- Department of Material Science and Engineering, Faculty of Engineering, Imam Khomeini International University, Qazvin, 34149-16818, Iran
| | - Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| | - Farnaz Ghorbani
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058, Erlangen, Germany
| | - Joaquim Miguel Oliveira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4805-017, Portugal
| |
Collapse
|
44
|
Jorgensen AM, Mahajan N, Atala A, Murphy SV. Advances in Skin Tissue Engineering and Regenerative Medicine. J Burn Care Res 2023; 44:S33-S41. [PMID: 36567474 PMCID: PMC9790899 DOI: 10.1093/jbcr/irac126] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There are an estimated 500,000 patients treated with full-thickness wounds in the United States every year. Fire-related burn injuries are among the most common and devastating types of wounds that require advanced clinical treatment. Autologous split-thickness skin grafting is the clinical gold standard for the treatment of large burn wounds. However, skin grafting has several limitations, particularly in large burn wounds, where there may be a limited area of non-wounded skin to use for grafting. Non-cellular dermal substitutes have been developed but have their own challenges; they are expensive to produce, may require immunosuppression depending on design and allogenic cell inclusion. There is a need for more advanced treatments for devastating burns and wounds. This manuscript provides a brief overview of some recent advances in wound care, including the use of advanced biomaterials, cell-based therapies for wound healing, biological skin substitutes, biological scaffolds, spray on skin and skin bioprinting. Finally, we provide insight into the future of wound care and technological areas that need to be addressed to support the development and incorporation of these technologies.
Collapse
Affiliation(s)
- Adam M Jorgensen
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Naresh Mahajan
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
45
|
Mazzocchi T, Guarnera D, Trucco D, Restaino FR, Vannozzi L, Siliberto A, Lisignoli G, Zaffagnini S, Russo A, Ricotti L. A Novel Approach for Multiple Material Extrusion in Arthroscopic Knee Surgery. Ann Biomed Eng 2023; 51:550-565. [PMID: 36057760 PMCID: PMC9928818 DOI: 10.1007/s10439-022-03061-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/15/2022] [Indexed: 11/28/2022]
Abstract
Articular cartilage defects and degenerative diseases are pathological conditions that cause pain and the progressive loss of joint functionalities. The most severe cases are treated through partial or complete joint replacement with prostheses, even if the interest in cartilage regeneration and re-growth methods is steadily increasing. These methods consist of the targeted deposition of biomaterials. Only a few tools have been developed so far for performing these procedures in a minimally invasive way. This work presents an innovative device for the direct deposition of multiple biomaterials in an arthroscopic scenario. The tool is easily handleable and allows the extrusion of three different materials simultaneously. It is also equipped with a flexible tip to reach remote areas of the damaged cartilage. Three channels are arranged coaxially and a spring-based dip-coating approach allows the fabrication and assembly of a bendable polymeric tip. Experimental tests were performed to characterize the tip, showing the ability to bend it up to 90° (using a force of ~ 1.5 N) and to extrude three coaxial biomaterials at the same time with both tip straight and tip fully bent. Rheometric analysis and fluid-dynamic computational simulations were performed to analyze the fluids' behavior; the maximum shear stresses were observed in correspondence to the distal tip and the channel convergence chamber, but with values up to ~ 1.2 kPa, compatible with a safe extrusion of biomaterials, even laden with cells. The cells viability was assessed after the extrusion with Live/Dead assay, confirming the safety of the extrusion procedures. Finally, the tool was tested arthroscopically in a cadaveric knee, demonstrating its ability to deliver the biomaterial in different areas, even ones that are typically hard-to-reach with traditional tools.
Collapse
Affiliation(s)
- Tommaso Mazzocchi
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy ,Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy
| | - Daniele Guarnera
- The BioRobotics Institute, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy. .,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, Piazza Martiri della Liberta' 33, 56127, Pisa, Italy.
| | - Diego Trucco
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy ,Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy ,IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Francesco Rocco Restaino
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy ,Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy
| | - Lorenzo Vannozzi
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy ,Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy
| | - Alessio Siliberto
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy ,Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Stefano Zaffagnini
- IRCCS Istituto Ortopedico Rizzoli, Orthopaedic and Traumatologic Clinic, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Alessandro Russo
- IRCCS Istituto Ortopedico Rizzoli, Orthopaedic and Traumatologic Clinic, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy ,Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, Piazza Martiri della Liberta’ 33, 56127 Pisa, Italy
| |
Collapse
|
46
|
Nanoengineered hydrogels as 3D biomimetic extracellular matrix with injectable and sustained delivery capability for cartilage regeneration. Bioact Mater 2023; 19:487-498. [PMID: 35600973 PMCID: PMC9092603 DOI: 10.1016/j.bioactmat.2022.03.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022] Open
Abstract
The regeneration of articular cartilage remains a great challenge due to the difficulty in effectively enhancing spontaneous healing. Recently, the combination of implanted stem cells, suitable biomaterials and bioactive molecules has attracted attention for tissue regeneration. In this study, a novel injectable nanocomposite was rationally designed as a sustained release platform for enhanced cartilage regeneration through integration of a chitosan-based hydrogel, articular cartilage stem cells (ACSCs) and mesoporous SiO2 nanoparticles loaded with anhydroicaritin (AHI). The biocompatible engineered nanocomposite acting as a novel 3D biomimetic extracellular matrix exhibited a remarkable sustained release effect due to the synergistic regulation of the organic hydrogel framework and mesopore channels of inorganic mSiO2 nanoparticles (mSiO2 NPs). Histological assessment and biomechanical tests showed that the nanocomposites exhibited superior performance in inducing ACSCs proliferation and differentiation in vitro and promoting extracellular matrix (ECM) production and cartilage regeneration in vivo. Such a novel multifunctional biocompatible platform was demonstrated to significantly enhance cartilage regeneration based on the sustained release of AHI, an efficient bioactive natural small molecule for ACSCs chondrogenesis, within the hybrid matrix of hydrogel and mSiO2 NPs. Hence, the injectable nanocomposite holds great promise for use as a 3D biomimetic extracellular matrix for tissue regeneration in clinical diagnostics. The anhydroicaritin (AHI) was identified as a bioactive factor for promoting cartilage repair. The hydrogel was designed to achieve sustained AHI release and optimize the microenvironment of cartilage defect sites. The hydrogel exhibited superior advantages for chondrogenic differentiation and cartilage regeneration. The hydrogel holds a great promise for use as functional scaffold for tissue and organ regeneration in the future.
Collapse
|
47
|
Guo X, Xi L, Yu M, Fan Z, Wang W, Ju A, Liang Z, Zhou G, Ren W. Regeneration of articular cartilage defects: Therapeutic strategies and perspectives. J Tissue Eng 2023; 14:20417314231164765. [PMID: 37025158 PMCID: PMC10071204 DOI: 10.1177/20417314231164765] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/03/2023] [Indexed: 04/03/2023] Open
Abstract
Articular cartilage (AC), a bone-to-bone protective device made of up to 80% water and populated by only one cell type (i.e. chondrocyte), has limited capacity for regeneration and self-repair after being damaged because of its low cell density, alymphatic and avascular nature. Resulting repair of cartilage defects, such as osteoarthritis (OA), is highly challenging in clinical treatment. Fortunately, the development of tissue engineering provides a promising method for growing cells in cartilage regeneration and repair by using hydrogels or the porous scaffolds. In this paper, we review the therapeutic strategies for AC defects, including current treatment methods, engineering/regenerative strategies, recent advances in biomaterials, and present emphasize on the perspectives of gene regulation and therapy of noncoding RNAs (ncRNAs), such as circular RNA (circRNA) and microRNA (miRNA).
Collapse
Affiliation(s)
- Xueqiang Guo
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Lingling Xi
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Mengyuan Yu
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Zhenlin Fan
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Weiyun Wang
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Andong Ju
- Abdominal Surgical Oncology, Xinxiang
Central Hospital, Institute of the Fourth Affiliated Hospital of Xinxiang Medical
University, Xinxiang, China
| | - Zhuo Liang
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
| | - Guangdong Zhou
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
- Department of Plastic and
Reconstructive Surgery, Shanghai Key Lab of Tissue Engineering, Shanghai 9th
People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
- Guangdong Zhou, Department of Plastic and
Reconstructive Surgery, Shanghai Key Lab of Tissue Engineering, Shanghai 9th
People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639
Shanghai Manufacturing Bureau Road, Shanghai 200011, China.
| | - Wenjie Ren
- Institutes of Health Central Plain, The
Third Affiliated Hospital of Xinxiang Medical University, Clinical Medical Center of
Tissue Engineering and Regeneration, Xinxiang Medical University, Xinxiang,
China
- Wenjie Ren, Institute of Regenerative
Medicine and Orthopedics, Institutes of Health Central Plain, Xinxiang Medical
University, 601 Jinsui Avenue, Hongqi District, Xinxiang 453003, Henan, China.
| |
Collapse
|
48
|
Ahmadian E, Eftekhari A, Janas D, Vahedi P. Nanofiber scaffolds based on extracellular matrix for articular cartilage engineering: A perspective. Nanotheranostics 2023; 7:61-69. [PMID: 36593799 PMCID: PMC9760364 DOI: 10.7150/ntno.78611] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/27/2022] [Indexed: 11/24/2022] Open
Abstract
Articular cartilage has a low self-repair capacity due to the lack of vessels and nerves. In recent times, nanofiber scaffolds have been widely used for this purpose. The optimum nanofiber scaffold should stimulate new tissue's growth and mimic the articular cartilage nature. Furthermore, the characteristics of the scaffold should match those of the cellular matrix components of the native tissue to best merge with the target tissue. Therefore, selective modification of prefabricated scaffolds based on the structure of the repaired tissues is commonly conducted to promote restoring the tissue. A thorough analysis is required to find out the architectural features of scaffolds that are essential to make the treatment successful. The current review aims to target this challenge. The article highlights different optimization approaches of nanofibrous scaffolds for improved cartilage tissue engineering. In this context, the influence of the architecture of nanoscaffolds on performance is discussed in detail. Finally, based on the gathered information, a future outlook is provided to catalyze development in this promising field.
Collapse
Affiliation(s)
- Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| | - Dawid Janas
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100, Gliwice, Poland,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| | - Parviz Vahedi
- Department of Anatomical Sciences, Maragheh University of Medical Sciences, Maragheh 78151-55158, Iran,✉ Corresponding authors: Aziz Eftekhari (), Dawid Janas (), Parviz Vahedi ()
| |
Collapse
|
49
|
Kutaish H, Tscholl PM, Cosset E, Bengtsson L, Braunersreuther V, Mor FM, Laedermann J, Furfaro I, Stafylakis D, Hannouche D, Gerstel E, Krause KH, Assal M, Menetrey J, Tieng V. Articular Cartilage Repair After Implantation of Hyaline Cartilage Beads Engineered From Adult Dedifferentiated Chondrocytes: Cartibeads Preclinical Efficacy Study in a Large Animal Model. Am J Sports Med 2023; 51:237-249. [PMID: 36592016 DOI: 10.1177/03635465221138099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Chondrocyte-based cell therapy to repair cartilage has been used for >25 years despite current limitations. This work presents a new treatment option for cartilage lesions. HYPOTHESIS High-quality hyaline cartilage microtissues called Cartibeads are capable of treating focal chondral lesions once implanted in the defect, by complete fusion of Cartibeads among themselves and their integration with the surrounding native cartilage and subchondral bone. STUDY DESIGN Controlled laboratory study. METHODS Cartibeads were first produced from human donors and characterized using histology (safranin O staining of glycosaminoglycan [GAG] and immunohistochemistry of collagen I and II) and GAG dosage. Cartibeads from 6 Göttingen minipigs were engineered and implanted in an autologous condition in the knee (4 or 5 lesions per knee). One group was followed up for 3 months and the other for 6 months. Feasibility and efficacy were measured using histological analysis and macroscopic and microscopic scores. RESULTS Cartibeads revealed hyaline features with strong staining of GAG and collagen II. High GAG content was obtained: 24.6-µg/mg tissue (wet weight), 15.52-µg/mg tissue (dry weight), and 35 ± 3-µg GAG/bead (mean ± SD). Histological analysis of Göttingen minipigs showed good integration of Cartibeads grafts at 3 and 6 months after implantation. The Bern Score of the histological assay comparing grafted versus empty lesions was significant at 3 months (grafted, n = 10; nongrafted, n = 4; score, 3.3 and 5.3, respectively) and 6 months (grafted, n = 11; nongrafted, n = 3; score, 1.6 and 5.1). CONCLUSION We developed an innovative 3-step method allowing, for the first time, the use of fully dedifferentiated adult chondrocytes with a high number of cell passage (owing to the extensive amplification in culture). Cartibeads engineered from chondrocytes hold potential as an advanced therapy medicinal product for treating cartilage lesions with established efficacy. CLINICAL RELEVANCE This successful preclinical study, combined with standardized manufacturing of Cartibeads according to good manufacturing practice guidelines, led to the approval of first-in-human clinical trial by the ethics committee and local medical authority. The generated data highlighted a promising therapy to treat cartilage lesions from a small amount of starting biopsy specimen. With our innovative cell amplification technology, very large lesions can be treated, and older active patients can benefit from it.
Collapse
Affiliation(s)
- Halah Kutaish
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland. University Medical Center, University of Geneva, Geneva, Switzerland. Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Philippe Matthias Tscholl
- Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Erika Cosset
- University Medical Center, University of Geneva, Geneva, Switzerland. Laboratory of Tumor Immunology, Oncology Department, Center for Translational Research in Onco- Hematology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Laura Bengtsson
- University Medical Center, University of Geneva, Geneva, Switzerland. Vanarix SA, Lausanne, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Vincent Braunersreuther
- Service of Clinical Pathology, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Flavio Maurizio Mor
- Tissue Engineering Laboratory, HEPIA/HES-SO, University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Jeremy Laedermann
- Wyss Center for Bio and Neuroengineering, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Ivan Furfaro
- Laboratory for Soft Bioelectronic Interfaces, Institute of Microengineering, Institute of Bioengineering, Centre for Neuroprosthetics, École Polytechnique Fédeérale de Lausanne (EPFL), Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Dimitrios Stafylakis
- Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Didier Hannouche
- University Medical Center, University of Geneva, Geneva, Switzerland. Department of Orthopaedics Surgery, Geneva University Hospitals, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Eric Gerstel
- University Medical Center, University of Geneva, Geneva, Switzerland. Clinique la Colline, Hirslanden, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Medical School, University of Geneva, Geneva, Switzerland. University Medical Center, University of Geneva, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Mathieu Assal
- University Medical Center, University of Geneva, Geneva, Switzerland. Foot and Ankle Surgery Centre, Centre Assal, Clinique La Colline, Hirslanden Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Menetrey
- University Medical Center, University of Geneva, Geneva, Switzerland. Centre for Sports Medicine and Exercise, Clinique la Colline, Hirslanden, Geneva, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| | - Vannary Tieng
- University Medical Center, University of Geneva, Geneva, Switzerland. Vanarix SA, Lausanne, Switzerland.,Investigation performed at the Faculty of Medicine, University of Geneva, in collaboration with Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
50
|
Mao Z, Bi X, Wu C, Zheng Y, Shu X, Wu S, Guan J, Ritchie RO. A Cell-Free Silk Fibroin Biomaterial Strategy Promotes In Situ Cartilage Regeneration Via Programmed Releases of Bioactive Molecules. Adv Healthc Mater 2023; 12:e2201588. [PMID: 36314425 DOI: 10.1002/adhm.202201588] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/11/2022] [Indexed: 02/03/2023]
Abstract
In situ tissue regeneration using cell-free biofunctional scaffolds has been extensively studied as a promising alternative strategy to promote cartilage repair. In this study, a cartilage-biomimetic silk fibroin (SF)-based scaffold with controlled sequential release of two bioactive molecules is developed. Transforming growth factor-β1 (TGF-β1) is initially loaded onto the SF scaffolds by physical absorption, which are then successively functionalized with bone marrow mesenchymal stem cells (BMSCs)-specific-affinity peptide (E7) via gradient degradation coating of Silk fibroin Methacryloyl (SilMA)/Hyaluronic acid Methacryloyl (HAMA). Such SF-based scaffolds exhibit excellent structural stability and catilage-like mechanical properties, thus providing a desirable 3D microenvironment for cartilage reconstruction. Furthermore, rapid initial release of E7 during the first few days, followed by slow and sustained release of TGF-β1 for as long as few weeks, synergistically induced the recruitment of BMSCs and chondrogenic differentiation of them in vitro. Finally, in vivo studies indicate that the implantation of the biofunctional scaffold markedly promote in situ cartilage regeneration in a rabbit cartilage defect model. It is believed that this cartilage-biomimetic biofunctional SF-based scaffold with sequential controlled release of E7 and TGF-β1 may have a promising potential for improved cartilage tissue engineering.
Collapse
Affiliation(s)
- Zhinan Mao
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing, 100191, China.,School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xuewei Bi
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, China
| | - Chengai Wu
- Beijing Jishuitan Hospital, Beijing Research Institute of Orthopedics and Traumatology, Beijing, 100035, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xiong Shu
- Beijing Jishuitan Hospital, Beijing Research Institute of Orthopedics and Traumatology, Beijing, 100035, China
| | - Sujun Wu
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing, 100191, China
| | - Juan Guan
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing, 100191, China.,Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, China
| | - Robert O Ritchie
- Department of Materials Science and Engineering, University of California, Berkeley, CA, 94720, USA
| |
Collapse
|