1
|
Shim DW, Lee KM, Lee D, Kim JS, Jung YS, Oh SS, Lee SW, Lee JW, Kim BS. Osteochondral Repair with Autologous Cartilage Transplantation with or without Bone Grafting: A Short Pilot Study in Mini-Pigs. Cartilage 2025; 16:61-70. [PMID: 37698092 PMCID: PMC11744595 DOI: 10.1177/19476035231199442] [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: 03/28/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023] Open
Abstract
OBJECTIVE Treatment strategies for osteochondral defects, for which particulated autologous cartilage transplantation (PACT) is an emerging treatment strategy, aim to restore the structure and function of the hyaline cartilage. Herein, we compared the efficacy of PACT with control or human transforming growth factor-β (rhTGF-β), and clarified the necessity of bone graft (BG) with PACT to treat shallow osteochondral defects in a porcine model. DESIGN Two skeletally mature male micropigs received 4 osteochondral defects in each knee. The 16 defects were randomized to (1) empty control, (2) PACT, (3) PACT with BG, or (4) rhTGF-β. Animals were euthanized after 2 months and histomorphometry, immunofluorescence analysis, semiquantitative evaluation (O'Driscoll score), and magnetic resonance observation of cartilage repair tissue (MOCART) score were performed. RESULTS Hyaline cartilages, glycosaminoglycan synthesis, and collagen type II staining were more abundant in the PACT than in the control and rhTGF-β groups. The O'Driscoll score was significantly different between groups (P < 0.001), with both PACT groups showing superiority (P = 0.002). PACT had the highest score (P = 0.002), with improved restoration of subchondral bone compared with PACT with BG. The MOCART score showed significant differences between groups (P = 0.021); MOCART and O'Driscoll scores showed high correlation (r = 0.847, P < 0.001). CONCLUSION Treatment of osteochondral defects with PACT improved tissue quality compared with that with control or rhTGF-β in a porcine model. BG, in addition to PACT, may be unnecessary for shallow osteochondral defects. Clinical Relevance. BG may not be necessary while performing PACT.
Collapse
Affiliation(s)
- Dong Woo Shim
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, Korea
- Department of Medicine, Inha Graduate School, Incheon, Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Donghyun Lee
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Jun Sik Kim
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Yeon Seop Jung
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Sung Suk Oh
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Si Wook Lee
- Department of Orthopaedic Surgery, School of Medicine, Dongsan Medical Center, Keimyung University, Daegu, Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Bom Soo Kim
- Department of Orthopaedic Surgery, College of Medicine, Inha University Hospital, Incheon, Korea
| |
Collapse
|
2
|
Yuan L, Liu J, Xiao S, Wei J, Liu H, Li Y, Zuo Y, Li Y, Wang J, Li J. EGCG-Modified Bioactive Core-Shell Fibers Modulate Oxidative Stress to Synergistically Promote Vascularized Bone Regeneration. ACS Biomater Sci Eng 2025; 11:543-555. [PMID: 39743979 DOI: 10.1021/acsbiomaterials.4c01906] [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: 01/04/2025]
Abstract
Oxidative stress induced by reactive oxygen species (ROS) can adversely affect tissue repair, whereas endowing biomaterials with antioxidant activity can improve the in vivo microenvironment, thereby promoting angiogenesis and osteogenesis. Accordingly, this study utilized epigallocatechin-3-gallate (EGCG), a material known for its reducing properties, oxidative self-polymerization capability, and strong binding characteristics, to modify a bioactive core-shell fibrous membrane (10RP-PG). Compared to the 10RP-PG fibrous membrane, the EGCG-modified fibrous membrane (E/10RP-PG) exhibited superior hydrophilicity, excellent cell adhesion, and compatibility. Moreover, the EGCG-modified fibrous membrane can effectively scavenge free radicals, ameliorate the local microenvironment, and foster angiogenesis (enhancing the expression of angiogenic genes in human umbilical vein endothelial cells (HUVECs) by 1.58 times and promoting vascular generation area upon subcutaneous implantation by 4.47 times). The enhancement of angiogenic activity of the E/10RP-PG fibrous membrane further promoted cartilage degeneration and absorption, as well as new bone formation, thus facilitating the repair of bone defects. This study provides a new strategy for promoting bone defect repair through the surface modification of biomaterials with an antioxidant agent, and the fabricated E/10RP-PG fibrous membranes show promise for guiding vascularized bone regeneration.
Collapse
Affiliation(s)
- Li Yuan
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Jiangshan Liu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Shiqi Xiao
- Clinical Medical College & Affiliated Hospital, Chengdu University, Chengdu 610081, PR China
| | - Jiawei Wei
- The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou 646000, PR China
| | - Huan Liu
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Yongzhi Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Yi Zuo
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Yubao Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610064, PR China
| | - Jidong Li
- Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610064, PR China
| |
Collapse
|
3
|
Feng Y, Jiang Z, Chen C, Hu L, Jiang Q, Wang Y, Cheng Z, Wang F, Yang G, Wang Y. Laminin expression profiles of osteogenic-and chondrogenic-induced dECM sheets. BIOMATERIALS ADVANCES 2024; 169:214127. [PMID: 39637724 DOI: 10.1016/j.bioadv.2024.214127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Decellularized extracellular matrix sheets (dECMSs) produced by stem cells have attracted attention because they preserve the natural biological activity of the ECM to direct lineage-specific differentiation with less immunogenicity. As a core ECM protein, laminin modulates cellular phenotype and differentiation. Nevertheless, no studies thus far have explored the distribution and abundance of laminins in diverse dECMSs. Herein, we first compared the differential expression of laminins among dECMSs in osteogenic-induced medium (OI-dECMS), chondrogenic-induced medium (CI-dECMS), and standard medium (dECMS), employing a defined mass spectrometry (MS)-based proteomic analysis. In vitro, dECMSs were verified to be successfully decellularized. Cluster analysis identified a marked fluctuation in the expression of 7 laminins and 17 laminin-associated proteins in OI-dECMS vs dECMS and CI-dECMS vs dECMS. Two significantly changed pathways were selected from the KEGG pathway enrichment analysis: the FAK/ERK pathway and the PI3K/AKT pathway. Moreover, Alkaline Phosphatase (ALP) activity, Alcian blue staining, and RT-qPCR results for recellularization showed that CI-dECMS promotes chondrogenesis while OI-dECMS inhibits osteogenesis compared with dECMS. In vivo experiments were conducted to implant dECMSs in a rat osteochondral defect, demonstrating that dECMS and CI-dECMS promoted bone and cartilage repair. Furthermore, the inhibitory analysis was performed to verify the function of specific laminin isoforms modulating osteogenesis and chondrogenesis, which might be related to FAK/ERK and PI3K/AKT pathways. In summary, this study constructed dECMS, OI-dECMS, and CI-dECMS and uncovered the internal comprehensive molecular regulatory network centralized by laminins, thus proposing a biomimetic substitute for bone and cartilage regeneration.
Collapse
Affiliation(s)
- Yuting Feng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Chaozhen Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Ling Hu
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou 311100, China
| | - Qifeng Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Yuchen Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhenxuan Cheng
- Affiliated Stomatology Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310059, China
| | - Fang Wang
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou 311100, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
4
|
ten Brink T, Damanik F, Rotmans JI, Moroni L. Unraveling and Harnessing the Immune Response at the Cell-Biomaterial Interface for Tissue Engineering Purposes. Adv Healthc Mater 2024; 13:e2301939. [PMID: 38217464 PMCID: PMC11468937 DOI: 10.1002/adhm.202301939] [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: 06/19/2023] [Revised: 12/14/2023] [Indexed: 01/15/2024]
Abstract
Biomaterials are defined as "engineered materials" and include a range of natural and synthetic products, designed for their introduction into and interaction with living tissues. Biomaterials are considered prominent tools in regenerative medicine that support the restoration of tissue defects and retain physiologic functionality. Although commonly used in the medical field, these constructs are inherently foreign toward the host and induce an immune response at the material-tissue interface, defined as the foreign body response (FBR). A strong connection between the foreign body response and tissue regeneration is suggested, in which an appropriate amount of immune response and macrophage polarization is necessary to trigger autologous tissue formation. Recent developments in this field have led to the characterization of immunomodulatory traits that optimizes bioactivity, the integration of biomaterials and determines the fate of tissue regeneration. This review addresses a variety of aspects that are involved in steering the inflammatory response, including immune cell interactions, physical characteristics, biochemical cues, and metabolomics. Harnessing the advancing knowledge of the FBR allows for the optimization of biomaterial-based implants, aiming to prevent damage of the implant, improve natural regeneration, and provide the tools for an efficient and successful in vivo implantation.
Collapse
Affiliation(s)
- Tim ten Brink
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Febriyani Damanik
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Joris I. Rotmans
- Department of Internal MedicineLeiden University Medical CenterAlbinusdreef 2Leiden2333ZAThe Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| |
Collapse
|
5
|
Zhao F, Qiu Y, Liu W, Zhang Y, Liu J, Bian L, Shao L. Biomimetic Hydrogels as the Inductive Endochondral Ossification Template for Promoting Bone Regeneration. Adv Healthc Mater 2024; 13:e2303532. [PMID: 38108565 DOI: 10.1002/adhm.202303532] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Repairing critical size bone defects (CSBD) is a major clinical challenge and requires effective intervention by biomaterial scaffolds. Inspired by the fact that the cartilaginous template-based endochondral ossification (ECO) process is crucial to bone healing and development, developing biomimetic biomaterials to promote ECO is recognized as a promising approach for repairing CSBD. With the unique highly hydrated 3D polymeric network, hydrogels can be designed to closely emulate the physiochemical properties of cartilage matrix to facilitate ECO. In this review, the various preparation methods of hydrogels possessing the specific physiochemical properties required for promoting ECO are introduced. The materiobiological impacts of the physicochemical properties of hydrogels, such as mechanical properties, topographical structures and chemical compositions on ECO, and the associated molecular mechanisms related to the BMP, Wnt, TGF-β, HIF-1α, FGF, and RhoA signaling pathways are further summarized. This review provides a detailed coverage on the materiobiological insights required for the design and preparation of hydrogel-based biomaterials to facilitate bone regeneration.
Collapse
Affiliation(s)
- Fujian Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yonghao Qiu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Wenjing Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Jia Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, 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
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, P. R. China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, P. R. China
| |
Collapse
|
6
|
Yang Y, Zhou X, Deng H, Chen L, Zhang X, Wu S, Song A, Liang F. The role of O-GlcNAcylation in bone metabolic diseases. Front Physiol 2024; 15:1416967. [PMID: 38915778 PMCID: PMC11194333 DOI: 10.3389/fphys.2024.1416967] [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: 04/13/2024] [Accepted: 05/20/2024] [Indexed: 06/26/2024] Open
Abstract
O-GlcNAcylation, as a post-translational modification, can modulate cellular activities such as kinase activity, transcription-translation, protein degradation, and insulin signaling by affecting the function of the protein substrate, including cellular localization of proteins, protein stability, and protein/protein interactions. Accumulating evidence suggests that dysregulation of O-GlcNAcylation is associated with disease progression such as cancer, neurodegeneration, and diabetes. Recent studies suggest that O-GlcNAcylation is also involved in the regulation of osteoblast, osteoclast and chondrocyte differentiation, which is closely related to the initiation and development of bone metabolic diseases such as osteoporosis, arthritis and osteosarcoma. However, the potential mechanisms by which O-GlcNAcylation regulates bone metabolism are not fully understood. In this paper, the literature related to the regulation of bone metabolism by O-GlcNAcylation was summarized to provide new potential therapeutic strategies for the treatment of orthopedic diseases such as arthritis and osteoporosis.
Collapse
Affiliation(s)
- Yajing Yang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
- School of Medicine, Xiamen University, Xiamen, China
| | - HuiLi Deng
- School of Medicine, Xiamen University, Xiamen, China
| | - Li Chen
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Xiaolin Zhang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Song Wu
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Aiqun Song
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| | - Fengxia Liang
- College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- University of Chinese Medicine (Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| |
Collapse
|
7
|
Shaikh S, Gupta S, Mishra A, Sheikh PA, Singh P, Kumar A. Laser-assisted synthesis of nano-hydroxyapatite and functionalization with bone active molecules for bone regeneration. Colloids Surf B Biointerfaces 2024; 237:113859. [PMID: 38547794 DOI: 10.1016/j.colsurfb.2024.113859] [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: 01/09/2024] [Revised: 02/27/2024] [Accepted: 03/17/2024] [Indexed: 04/08/2024]
Abstract
The main goal of bone tissue engineering research is to replace the allogenic and autologous bone graft substitutes that can promote bone repair. Owing to excellent biocompatibility and osteoconductivity, hydroxyapatite is in extensive research and high demand for both medical and non-medical applications. Although various methods have been developed for the synthesis of hydroxyapatite, in the present study we have shown the use of nanosecond laser energy in the wet precipitation method of nano-hydroxyapatite (nHAP) synthesis without using ammonium solution or any other chemicals for pH maintenance. Here, the present study aimed to fabricate the nanohydroxyapatite using a nanosecond laser. The X-ray diffraction and Fourier transform infrared spectroscopy have confirmed the hydroxyapatite formation under laser irradiation in less time without aging. A transmission electron microscopy confirmed the nano size of synthesized nHAP, which is comparable to conventional nHAP. The length and width of the laser-assisted nHAP were found to be in the range of 50-200 nm and 15-20 nm, respectively, at various laser parameters. The crystallite size obtained by Debye Scherrer formulae was found to be in the range of ∼ 16-36 nm. In addition, laser-assisted nHAP based composite cryogel (nanohydroxyapatite/gelatin/collagen I) was synthesized and impregnated with bioactive molecules (bone morphogenic protein and zoledronic acid) that demonstrated significant osteogenic potential both in vitro in cell experiment and in vivo rat muscle pouch model (abdomen and tibia muscles). Dual-energy X-ray analysis, micro-CT, and histological analysis confirmed ectopic bone regeneration. Micro-CT based histomorphometry showed a higher amount (more than 10-fold) of mineralization for animal groups implanted with composite cryogels loaded with bioactive molecules compared to only composite cryogels groups. Our findings thus demonstrate a controlled and rapid synthetic method for the synthesis of nHAP with various physical, chemical, and biological properties exhibited as comparable to conventionally synthesized nHAP.
Collapse
Affiliation(s)
- Shazia Shaikh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India; Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India.
| | - Sneha Gupta
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ankita Mishra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Parvaiz A Sheikh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Prerna Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India
| | - Ashok Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, India; Center for Environmental Science and Engineering, Indian Institute of Technology Kanpur, Kanpur 208016, India; Center for Nanosciences, Indian Institute of Technology Kanpur, Kanpur 208016, India; The Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, India; Center of Excellence for Materials in Medicine, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, UP 208016, India.
| |
Collapse
|
8
|
Spessot E, Passuello S, Shah LV, Maniglio D, Motta A. Nanocomposite Methacrylated Silk Fibroin-Based Scaffolds for Bone Tissue Engineering. Biomimetics (Basel) 2024; 9:218. [PMID: 38667229 PMCID: PMC11048339 DOI: 10.3390/biomimetics9040218] [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: 02/28/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The treatment of bone defects is a clinical challenge. Bone tissue engineering is gaining interest as an alternative to current treatments, with the development of 3D porous structures (scaffolds) helpful in promoting bone regeneration by ensuring temporary functional support. In this work, methacrylated silk fibroin (SilMA) sponges were investigated as scaffolds for bone tissue engineering by exploiting the combination of physical (induced by NaCl salt during particulate leaching) and chemical crosslinking (induced by UV-light exposure) techniques. A biomimetic approach was adopted to better simulate the extracellular matrix of the bone by introducing either natural (mussel shell-derived) or synthetic-origin hydroxyapatite nanoparticles into the SilMA sponges. The obtained materials were characterized in terms of pore size, water absorption capability and mechanical properties to understand both the effect of the inclusion of the two different types of nanoparticles and the effect of the photocrosslinking. Moreover, the SilMA sponges were tested for their bioactivity and suitability for bone tissue engineering purposes by using osteosarcoma cells, studying their metabolism by an AlamarBlue assay and their morphology by scanning electron microscopy. Results indicate that photocrosslinking helps in obtaining more regular structures with bimodal pore size distributions and in enhancing the stability of the constructs in water. Moreover, the addition of naturally derived hydroxyapatite was observed to be more effective at activating osteosarcoma cell metabolism than synthetic hydroxyapatite, showing a statistically significant difference in the AlamarBlue measurement on day 7 after seeding. The methacrylated silk fibroin/hydroxyapatite nanocomposite sponges developed in this work were found to be promising tools for targeting bone regeneration with a sustainable approach.
Collapse
Affiliation(s)
- Eugenia Spessot
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| | - Serena Passuello
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
| | - Lekha Vinod Shah
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| | - Devid Maniglio
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| | - Antonella Motta
- Department of Industrial Engineering and BIOtech Research Centre, University of Trento, Via Sommarive 9, 38123 Trento, Italy; (E.S.); (L.V.S.); (A.M.)
- European Institute of Excellence on Tissue Engineering and Regenerative Medicine Unit, Via delle Regole 101, 38123 Trento, Italy
| |
Collapse
|
9
|
Palomares Cabeza V, Fahy N, Kiernan CH, Lolli A, Witte-Bouma J, Fahmy Garcia S, Merino A, Kops N, Ridwan Y, Wolvius EB, Brama PAJ, Hoogduijn MJ, Farrell E. Bone formation by human paediatric marrow stromal cells in a functional allogeneic immune system. Biomaterials 2024; 306:122471. [PMID: 38377846 DOI: 10.1016/j.biomaterials.2024.122471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 02/22/2024]
Abstract
Allogeneic stem-cell based regenerative medicine is a promising approach for bone defect repair. The use of chondrogenically differentiated human marrow stromal cells (MSCs) has been shown to lead to bone formation by endochondral ossification in immunodeficient pre-clinical models. However, an insight into the interactions between the allogeneic immune system and the human MSC-derived bone grafts has not been fully achieved yet. The choice of a potent source of MSCs isolated from pediatric donors with consistent differentiation and high proliferation abilities, as well as low immunogenicity, could increase the chance of success for bone allografts. In this study, we employed an immunodeficient animal model humanised with allogeneic immune cells to study the immune responses towards chondrogenically differentiated human pediatric MSCs (ch-pMSCs). We show that ch-differentiated pMSCs remained non-immunogenic to allogeneic CD4 and CD8 T cells in an in vitro co-culture model. After subcutaneous implantation in mice, ch-pMSC-derived grafts were able to initiate bone mineralisation in the presence of an allogeneic immune system for 3 weeks without the onset of immune responses. Re-exposing the splenocytes of the humanised animals to pMSCs did not trigger further T cell proliferation, suggesting an absence of secondary immune responses. Moreover, ch-pMSCs generated mature bone after 8 weeks of implantation that persisted for up to 6 more weeks in the presence of an allogeneic immune system. These data collectively show that human allogeneic chondrogenically differentiated pediatric MSCs might be a safe and potent option for bone defect repair in the tissue engineering and regenerative medicine setting.
Collapse
Affiliation(s)
- Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Niamh Fahy
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Applied Science, Technological University of the Shannon: Midlands Midwest, Limerick, Ireland
| | - Caoimhe H Kiernan
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Shorouk Fahmy Garcia
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ana Merino
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Nicole Kops
- Department of Orthopedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yanto Ridwan
- AMIE Core Facility, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Martin J Hoogduijn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
10
|
Schott NG, Kaur G, Coleman R, Stegemann JP. Modular, Vascularized Hypertrophic Cartilage Constructs for Bone Tissue Engineering Applications. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582166. [PMID: 38464155 PMCID: PMC10925222 DOI: 10.1101/2024.02.26.582166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Insufficient vascularization is a main barrier to creating engineered bone grafts for treating large and ischemic defects. Modular tissue engineering approaches have promise in this application because of the ability to combine tissue types and to localize microenvironmental cues to drive desired cell function. In direct bone formation approaches, it is challenging to maintain sustained osteogenic activity, since vasculogenic cues can inhibit tissue mineralization. This study harnessed the physiological process of endochondral ossification to create multiphase tissues that allowed concomitant mineralization and vessel formation. Mesenchymal stromal cells in pellet culture were differentiated toward a cartilage phenotype, followed by induction to chondrocyte hypertrophy. Hypertrophic pellets exhibited increased alkaline phosphatase activity, calcium deposition, and osteogenic gene expression relative to chondrogenic pellets. In addition, hypertrophic pellets secreted and sequestered angiogenic factors, and supported new blood vessel formation by co-cultured endothelial cells and undifferentiated stromal cells. Multiphase constructs created by combining hypertrophic pellets and vascularizing microtissues and maintained in unsupplemented basal culture medium were shown to support robust vascularization and sustained tissue mineralization. These results demonstrate a new in vitro strategy to produce multiphase engineered constructs that concomitantly support the generation of mineralize and vascularized tissue in the absence of exogenous osteogenic or vasculogenic medium supplements.
Collapse
|
11
|
Wei Y, Pan H, Yang J, Zeng C, Wan W, Chen S. Aligned cryogel fibers incorporated 3D printed scaffold effectively facilitates bone regeneration by enhancing cell recruitment and function. SCIENCE ADVANCES 2024; 10:eadk6722. [PMID: 38324693 PMCID: PMC10849600 DOI: 10.1126/sciadv.adk6722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
Reconstructing extensive cranial defects represents a persistent clinical challenge. Here, we reported a hybrid three-dimensional (3D) printed scaffold with modification of QK peptide and KP peptide for effectively promoting endogenous cranial bone regeneration. The hybrid 3D printed scaffold consists of vertically aligned cryogel fibers that guide and promote cell penetration into the defect area in the early stages of bone repair. Then, the conjugated QK peptide and KP peptide further regulate the function of the recruited cells to promote vascularization and osteogenic differentiation in the defect area. The regenerated bone volume and surface coverage of the dual peptide-modified hybrid scaffold were significantly higher than the positive control group. In addition, the dual peptide-modified hybrid scaffold demonstrated sustained enhancement of bone regeneration and avoidance of bone resorption compared to the collagen sponge group. We expect that the design of dual peptide-modified hybrid scaffold will provide a promising strategy for bone regeneration.
Collapse
Affiliation(s)
- Yuxuan Wei
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, the Third Affiliated Hospital of Southern Medical University. Guangzhou, Guangdong 510630, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Hao Pan
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Department of Orthopaedic Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Jianqiu Yang
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Department of Orthopaedic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006 China
| | - Canjun Zeng
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, the Third Affiliated Hospital of Southern Medical University. Guangzhou, Guangdong 510630, China
| | - Wenbing Wan
- Department of Orthopaedic Surgery, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006 China
| | - Shixuan Chen
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| |
Collapse
|
12
|
Schaller R, Moya A, Zhang G, Chaaban M, Paillaud R, Bartoszek EM, Schaefer DJ, Martin I, Kaempfen A, Scherberich A. Engineered phalangeal grafts for children with symbrachydactyly: A proof of concept. J Tissue Eng 2024; 15:20417314241257352. [PMID: 38872920 PMCID: PMC11171439 DOI: 10.1177/20417314241257352] [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: 02/21/2024] [Accepted: 05/10/2024] [Indexed: 06/15/2024] Open
Abstract
Tissue engineering approaches hold great promise in the field of regenerative medicine, especially in the context of pediatric applications, where ideal grafts need to restore the function of the targeted tissue and consider growth. In the present study, we aimed to develop a protocol to engineer autologous phalangeal grafts of relevant size for children suffering from symbrachydactyly. This condition results in hands with short fingers and missing bones. A previously-described, developmentally-inspired strategy based on endochondral ossification (ECO)-the main pathway leading to bone and bone marrow development-and adipose derived-stromal cells (ASCs) as the source of chondroprogenitor was used. First, we demonstrated that pediatric ASCs associated with collagen sponges can generate hypertrophic cartilage tissues (HCTs) in vitro that remodel into bone tissue in vivo via ECO. Second, we developed and optimized an in vitro protocol to generate HCTs in the shape of small phalangeal bones (108-390 mm3) using freshly isolated adult cells from the stromal vascular fraction (SVF) of adipose tissue, associated with two commercially available large collagen scaffolds (Zimmer Plug® and Optimaix 3D®). We showed that after 12 weeks of in vivo implantation in an immunocompromised mouse model such upscaled grafts remodeled into bone organs (including bone marrow tissues) retaining the defined shape and size. Finally, we replicated similar outcome (albeit with a slight reduction in cartilage and bone formation) by using minimally expanded pediatric ASCs (3 × 106 cells per grafts) in the same in vitro and in vivo settings, thereby validating the compatibility of our pediatric phalanx engineering strategy with a clinically relevant scenario. Taken together, these results represent a proof of concept of an autologous approach to generate osteogenic phalangeal grafts of pertinent clinical size, using ASCs in children born with symbrachydactyly, despite a limited amount of tissue available from pediatric patients.
Collapse
Affiliation(s)
- Romain Schaller
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Adrien Moya
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Gangyu Zhang
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Mansoor Chaaban
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Robert Paillaud
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ewelina M Bartoszek
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandre Kaempfen
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
- Paediatric Orthopaedic, University Children’s Hospital Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
13
|
Al Maruf DSA, Xin H, Cheng K, Garcia AG, Mohseni-Dargah M, Ben-Sefer E, Tomaskovic-Crook E, Crook JM, Clark JR. Bioengineered cartilaginous grafts for repairing segmental mandibular defects. J Tissue Eng 2024; 15. [DOI: 10.1177/20417314241267017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Reconstructing critical-sized craniofacial bone defects is a global healthcare challenge. Current methods, like autologous bone transplantation, face limitations. Bone tissue engineering offers an alternative to autologous bone, with traditional approaches focusing on stimulating osteogenesis via the intramembranous ossification (IMO) pathway. However, IMO falls short in addressing larger defects, particularly in clinical scenarios where there is insufficient vascularisation. This review explores redirecting bone regeneration through endochondral ossification (ECO), a process observed in long bone healing stimulated by hypoxic conditions. Despite its promise, gaps exist in applying ECO to bone tissue engineering experiments, requiring the elucidation of key aspects such as cell sources, biomaterials and priming protocols. This review discusses various scaffold biomaterials and cellular sources for chondrogenesis and hypertrophic chondrocyte priming, mirroring the ECO pathway. The review highlights challenges in current endochondral priming and proposes alternative approaches. Emphasis is on segmental mandibular defect repair, offering insights for future research and clinical application. This concise review aims to advance bone tissue engineering by addressing critical gaps in ECO strategies.
Collapse
Affiliation(s)
- D S Abdullah Al Maruf
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Hai Xin
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Kai Cheng
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| | - Alejandro Garcia Garcia
- Cell, Tissue and Organ Engineering Laboratory, Biomedical Centre (BMC), Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, Lund, Sweden
| | - Masoud Mohseni-Dargah
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
| | - Eitan Ben-Sefer
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Eva Tomaskovic-Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jeremy Micah Crook
- Arto Hardy Biomedical Innovation Hub, Chris O`Brien Lifehouse, Camperdown, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Sarcoma and Surgical Research Centre, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- ARC Centre of Excellence for Electromaterials Science, The University of Wollongong, Wollongong, NSW, Australia
- Intelligent Polymer Research Institute, AIIM Facility, The University of Wollongong, Wollongong, NSW, Australia
| | - Jonathan Robert Clark
- Integrated Prosthetics and Reconstruction, Department of Head and Neck Surgery, Chris O’Brien Lifehouse, Camperdown, NSW, Australia
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Camperdown, NSW, Australia
| |
Collapse
|
14
|
Jeyachandran D, Murshed M, Haglund L, Cerruti M. A Bioglass-Poly(lactic-co-glycolic Acid) Scaffold@Fibrin Hydrogel Construct to Support Endochondral Bone Formation. Adv Healthc Mater 2023; 12:e2300211. [PMID: 37462089 PMCID: PMC11468889 DOI: 10.1002/adhm.202300211] [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: 01/19/2023] [Revised: 06/21/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
Bone tissue engineering using stem cells to build bone directly on a scaffold matrix often fails due to lack of oxygen at the injury site. This may be avoided by following the endochondral ossification route; herein, a cartilage template is promoted first, which can survive hypoxic environments, followed by its hypertrophy and ossification. However, hypertrophy is so far only achieved using biological factors. This work introduces a Bioglass-Poly(lactic-co-glycolic acid@fibrin (Bg-PLGA@fibrin) construct where a fibrin hydrogel infiltrates and encapsulates a porous Bg-PLGA. The hypothesis is that mesenchymal stem cells (MSCs) loaded in the fibrin gel and induced into chondrogenesis degrade the gel and become hypertrophic upon reaching the stiffer, bioactive Bg-PLGA core, without external induction factors. Results show that Bg-PLGA@fibrin induces hypertrophy, as well as matrix mineralization and osteogenesis; it also promotes a change in morphology of the MSCs at the gel/scaffold interface, possibly a sign of osteoblast-like differentiation of hypertrophic chondrocytes. Thus, the Bg-PLGA@fibrin construct can sequentially support the different phases of endochondral ossification purely based on material cues. This may facilitate clinical translation by decreasing in-vitro cell culture time pre-implantation and the complexity associated with the use of external induction factors.
Collapse
Affiliation(s)
| | - Monzur Murshed
- Faculty of DentistryDepartment of Medicineand Shriners Hospital for ChildrenMcGill UniversityMontrealQuebecH4A 0A9Canada
| | - Lisbet Haglund
- Experimental SurgeryMcGill UniversityMontrealH3G 2M1Canada
| | - Marta Cerruti
- Department of Mining and Materials EngineeringMcGill UniversityMontrealH3A 0C1Canada
| |
Collapse
|
15
|
Kolliopoulos V, Polanek M, Xu H, Harley B. Inflammatory Licensed hMSCs Exhibit Enhanced Immunomodulatory Capacity in a Biomaterial Mediated Manner. ACS Biomater Sci Eng 2023; 9:4916-4928. [PMID: 37390452 PMCID: PMC10600978 DOI: 10.1021/acsbiomaterials.3c00290] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
Craniomaxillofacial (CMF) bone injuries represent particularly challenging environments for regenerative healing due to their large sizes, irregular and unique defect shapes, angiogenic requirements, and mechanical stabilization needs. These defects also exhibit a heightened inflammatory environment that can complicate the healing process. This study investigates the influence of the initial inflammatory stance of human mesenchymal stem cells (hMSCs) on key osteogenic, angiogenic, and immunomodulatory criteria when cultured in a class of mineralized collagen scaffolds under development for CMF bone repair. We previously showed that changes in scaffold pore anisotropy and glycosaminoglycan content can significantly alter the regenerative activity of both MSCs and macrophages. While MSCs are known to adopt an immunomodulatory phenotype in response to inflammatory stimuli, here, we define the nature and persistence of MSC osteogenic, angiogenic, and immunomodulatory phenotypes in a 3D mineralized collagen environment, and further, whether changes to scaffold architecture and organic composition can blunt or accentuate this response as a function of inflammatory licensing. Notably, we found that a one-time licensing treatment of MSCs induced higher immunomodulatory potential compared to basal MSCs as observed by sustained immunomodulatory gene expression throughout the first 7 days as well as an increase in immunomodulatory cytokine (PGE2 and IL-6) expression throughout a 21-day culture period. Further, heparin scaffolds facilitated higher osteogenic cytokine secretion but lower immunomodulatory cytokine secretion compared to chondroitin-6-sulfate scaffolds. Anisotropic scaffolds facilitated higher secretion of both osteogenic protein OPG and immunomodulatory cytokines (PGE2 and IL-6) compared to isotropic scaffolds. These results highlight the importance of scaffold properties on the sustained kinetics of cell response to an inflammatory stimulus. The development of a biomaterial scaffold capable of interfacing with hMSCs to facilitate both immunomodulatory and osteogenic responses is an essential next step to determining the quality and kinetics of craniofacial bone repair.
Collapse
Affiliation(s)
- Vasiliki Kolliopoulos
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 600 S. Mathews Ave., Urbana, Illinois 61801, United States
| | - Maxwell Polanek
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 600 S. Mathews Ave., Urbana, Illinois 61801, United States
| | - Hui Xu
- Tumor Engineering and Phenotyping (TEP) Shared Resource, Cancer Center at Illinois University of Illinois Urbana-Champaign, 405 N. Mathews Ave., Urbana, Illinois 61801, United States
| | - Brendan Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 600 S. Mathews Ave., Urbana, Illinois 61801, United States
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, 405 N. Mathews Ave., Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 600 S. Mathews Ave., Urbana, Illinois 61801, United States
| |
Collapse
|
16
|
Tsou HK, Wu CH, Chan LY, Kataoka K, Itokazu N, Tsuzuki M, Hu H, Zhuo GY, Itaka K, Lin CY. Administration of mRNA-Nanomedicine-Augmented Calvarial Defect Healing via Endochondral Ossification. Pharmaceutics 2023; 15:1965. [PMID: 37514151 PMCID: PMC10383176 DOI: 10.3390/pharmaceutics15071965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Large-area craniofacial defects remain a challenge for orthopaedists, hastening the need to develop a facile and safe tissue engineering strategy; osteoconductive material and a combination of optimal growth factors and microenvironment should be considered. Faced with the unmet need, we propose that abundant cytokines and chemokines can be secreted from the bone defect, provoking the infiltration of endogenous stem cells to assist bone regeneration. We can provide a potent mRNA medicine cocktail to promptly initiate the formation of bone templates, osteogenesis, and subsequent bone matrix deposition via endochondral ossification, which may retard rapid fibroblast infiltration and prevent the formation of atrophic non-union. We explored the mutual interaction of BMP2 and TGFβ3 mRNA, both potent chondrogenic factors, on inducing endochondral ossification; examined the influence of in vitro the transcribed polyA tail length on mRNA stability; prepared mRNA nanomedicine using a PEGylated polyaspartamide block copolymer loaded in a gelatin sponge and grafted in a critical-sized calvarial defect; and evaluated bone regeneration using histological and μCT examination. The BMP2 and TGFβ3 composite mRNA nanomedicine resulted in over 10-fold new bone volume (BV) regeneration in 8 weeks than the BMP2 mRNA nanomedicine administration alone, demonstrating that the TGFβ3 mRNA nanomedicine synergistically enhances the bone's formation capability, which is induced by BMP2 mRNA nanomedicine. Our data demonstrated that mRNA-medicine-mediated endochondral ossification provides an alternative cell-free tissue engineering methodology for guiding craniofacial defect healing.
Collapse
Affiliation(s)
- Hsi-Kai Tsou
- Functional Neurosurgery Division, Neurological Institute, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County 35664, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- College of Health, National Taichung University of Science and Technology, Taichung 40303, Taiwan
| | - Cheng-Hsin Wu
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Long Yi Chan
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki 210-0821, Japan
| | - Nanae Itokazu
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Minoru Tsuzuki
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Hsuan Hu
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Guan-Yu Zhuo
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Keiji Itaka
- Department of Biofunction Research, Institute of Biomaterial and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| | - Chin-Yu Lin
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Biomedical Sciences and Engineering, Tzu Chi University, Hualien 97004, Taiwan
| |
Collapse
|
17
|
di Summa PG, Sapino G, Wagner D, Maruccia M, Guillier D, Burger H. Combined Free Flaps for Optimal Orthoplastic Lower Limb Reconstruction: A Retrospective Series. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050859. [PMID: 37241091 DOI: 10.3390/medicina59050859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023]
Abstract
Background and Objectives: Open fracture of the lower limb can lead to substantial bone and soft tissue damage, resulting in a challenging reconstructive scenarios, especially in presence of bone or periosteal loss, with a relevant risk of non-union. This work analyzes outcomes of using a double approach for orthoplastic reconstruction, adopting the free medial condyle flap to solve the bone defects, associated to a second free flap for specific soft tissue coverage. Indications, outcomes and reconstructive rationales are discussed. Materials and Methods: A retrospective investigation was performed on patients who underwent complex two-flap microsurgical reconstruction from January 2018 to January 2022. Inclusion criteria in this study were the use of a free femoral condyle periostal/bone flap together with a second skin-only flap. Only distal third lower limb reconstructions were included in order to help equalize our findings. Out of the total number of patients, only patients with complete pre- and post-operative follow-up (minimum 6 months) data were included in the study. Results: Seven patients were included in the study, with a total of 14 free flaps. The average age was 49. Among comorbidities, four patients were smokers and none suffered from diabetes. Etiology of the defect was acute trauma in four cases and septic non-union in three cases. No major complications occurred, and all flaps healed uneventfully with complete bone union. Conclusions: Combining a bone periosteal FMC to a second skin free flap for tailored defect coverage allowed achievement of bone union in all patients, despite the lack of initial bone vascularization or chronic infection. FMC is confirmed to be a versatile flap for small-to-medium bone defects, especially considering its use as a periosteal-only flap, with minimal donor site morbidity. Choosing a second flap for coverage allows for a higher inset freedom and tailored reconstruction, finally enhancing orthoplastic success.
Collapse
Affiliation(s)
- Pietro G di Summa
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
| | - Gianluca Sapino
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
| | - Daniel Wagner
- Department of Orthopedic Surgery, Centre Hospitalier Universitaire Vaudois (CHUV), 1011 Lausanne, Switzerland
| | - Michele Maruccia
- Department of Plastic Surgery and Reconstructive Surgery, University Hospital of Bari, 70100 Bari, Italy
| | - David Guillier
- Department of Plastic Reconstructive and Hand Surgery, Department of Oral and Maxillofacial Surgery-University Hospital, 21231 Dijon, France
| | - Heinz Burger
- Privat Clinic Maria Hilf, 9010 Klagenfurt, Austria
| |
Collapse
|
18
|
You J, Liu M, Li M, Zhai S, Quni S, Zhang L, Liu X, Jia K, Zhang Y, Zhou Y. The Role of HIF-1α in Bone Regeneration: A New Direction and Challenge in Bone Tissue Engineering. Int J Mol Sci 2023; 24:ijms24098029. [PMID: 37175732 PMCID: PMC10179302 DOI: 10.3390/ijms24098029] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
The process of repairing significant bone defects requires the recruitment of a considerable number of cells for osteogenesis-related activities, which implies the consumption of a substantial amount of oxygen and nutrients. Therefore, the limited supply of nutrients and oxygen at the defect site is a vital constraint that affects the regenerative effect, which is closely related to the degree of a well-established vascular network. Hypoxia-inducible factor (HIF-1α), which is an essential transcription factor activated in hypoxic environments, plays a vital role in vascular network construction. HIF-1α, which plays a central role in regulating cartilage and bone formation, induces vascular invasion and differentiation of osteoprogenitor cells to promote and maintain extracellular matrix production by mediating the adaptive response of cells to changes in oxygen levels. However, the application of HIF-1α in bone tissue engineering is still controversial. As such, clarifying the function of HIF-1α in regulating the bone regeneration process is one of the urgent issues that need to be addressed. This review provides insight into the mechanisms of HIF-1α action in bone regeneration and related recent advances. It also describes current strategies for applying hypoxia induction and hypoxia mimicry in bone tissue engineering, providing theoretical support for the use of HIF-1α in establishing a novel and feasible bone repair strategy in clinical settings.
Collapse
Affiliation(s)
- Jiaqian You
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Manxuan Liu
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Minghui Li
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Shaobo Zhai
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Sezhen Quni
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Lu Zhang
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Xiuyu Liu
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Kewen Jia
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Yidi Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
- School of Stomatology, Jilin University, Changchun 130021, China
| | - Yanmin Zhou
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, China
- School of Stomatology, Jilin University, Changchun 130021, China
| |
Collapse
|
19
|
Ji E, Leijsten L, Witte-Bouma J, Rouchon A, Di Maggio N, Banfi A, van Osch GJVM, Farrell E, Lolli A. In Vitro Mineralisation of Tissue-Engineered Cartilage Reduces Endothelial Cell Migration, Proliferation and Tube Formation. Cells 2023; 12:cells12081202. [PMID: 37190110 DOI: 10.3390/cells12081202] [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: 02/15/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Tissue engineering bone via endochondral ossification requires the generation of a cartilage template which undergoes vascularisation and remodelling. While this is a promising route for bone repair, achieving effective cartilage vascularisation remains a challenge. Here, we investigated how mineralisation of tissue-engineered cartilage affects its pro-angiogenic potential. To generate in vitro mineralised cartilage, human mesenchymal stromal cell (hMSC)-derived chondrogenic pellets were treated with β-glycerophosphate (BGP). After optimising this approach, we characterised the changes in matrix components and pro-angiogenic factors by gene expression analysis, histology and ELISA. Human umbilical vein endothelial cells (HUVECs) were exposed to pellet-derived conditioned media, and migration, proliferation and tube formation were assessed. We established a reliable strategy to induce in vitro cartilage mineralisation, whereby hMSC pellets are chondrogenically primed with TGF-β for 2 weeks and BGP is added from week 2 of culture. Cartilage mineralisation determines loss of glycosaminoglycans, reduced expression but not protein abundance of collagen II and X, and decreased VEGFA production. Finally, the conditioned medium from mineralised pellets showed a reduced ability to stimulate endothelial cell migration, proliferation and tube formation. The pro-angiogenic potential of transient cartilage is thus stage-dependent, and this aspect must be carefully considered in the design of bone tissue engineering strategies.
Collapse
Affiliation(s)
- Encheng Ji
- Department of Oral and Maxillofacial Surgery, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Lieke Leijsten
- Department of Oral and Maxillofacial Surgery, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Adelin Rouchon
- Department of Biomedicine, Basel University Hospital, University of Basel, 4031 Basel, Switzerland
| | - Nunzia Di Maggio
- Department of Biomedicine, Basel University Hospital, University of Basel, 4031 Basel, Switzerland
| | - Andrea Banfi
- Department of Biomedicine, Basel University Hospital, University of Basel, 4031 Basel, Switzerland
| | - Gerjo J V M van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Otorhinolaryngology, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
- Department of Biomechanical Engineering, University of Technology Delft, 2628 CD Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
20
|
Li T, Ma Z, Zhang Y, Yang Z, Li W, Lu D, Liu Y, Qiang L, Wang T, Ren Y, Wang W, He H, Zhou X, Mao Y, Zhu J, Wang J, Chen X, Dai K. Regeneration of Humeral Head Using a 3D Bioprinted Anisotropic Scaffold with Dual Modulation of Endochondral Ossification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205059. [PMID: 36755334 PMCID: PMC10131811 DOI: 10.1002/advs.202205059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Indexed: 06/18/2023]
Abstract
Tissue engineering is theoretically thought to be a promising method for the reconstruction of biological joints, and thus, offers a potential treatment alternative for advanced osteoarthritis. However, to date, no significant progress is made in the regeneration of large biological joints. In the current study, a biomimetic scaffold for rabbit humeral head regeneration consisting of heterogeneous porous architecture, various bioinks, and different hard supporting materials in the cartilage and bone regions is designed and fabricated in one step using 3D bioprinting technology. Furthermore, orchestrated dynamic mechanical stimulus combined with different biochemical cues (parathyroid hormone [PTH] and chemical component hydroxyapatite [HA] in the outer and inner region, respectively) are used for dual regulation of endochondral ossification. Specifically, dynamic mechanical stimulus combined with growth factor PTH in the outer region inhibits endochondral ossification and results in cartilage regeneration, whereas dynamic mechanical stimulus combined with HA in the inner region promotes endochondral ossification and results in efficient subchondral bone regeneration. The strategy established in this study with the dual modulation of endochondral ossification for 3D bioprinted anisotropic scaffolds represents a versatile and scalable approach for repairing large joints.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Zhengjiang Ma
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Yuxin Zhang
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Zezheng Yang
- Department of OrthopedicsThe Fifth People's Hospital of ShanghaiFudan UniversityMinhang DistrictShanghai200240P. R. China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Dezhi Lu
- School of MedicineShanghai UniversityJing An DistrictShanghai200444China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Lei Qiang
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Tianchang Wang
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Ya Ren
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Wenhao Wang
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Hongtao He
- The Third Ward of Department of OrthopedicsThe Second Hospital of Dalian Medical UniversityNo. 467, Zhongshan Road, Shahekou DistrictDalianLiaoning Province116000P. R. China
| | - Xiaojun Zhou
- College of Biological Science and Medical EngineeringState Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsDonghua UniversityShanghai201620P. R. China
| | - Yuanqing Mao
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Junfeng Zhu
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Xiaodong Chen
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| |
Collapse
|
21
|
Pitacco P, Sadowska JM, O'Brien FJ, Kelly DJ. 3D bioprinting of cartilaginous templates for large bone defect healing. Acta Biomater 2023; 156:61-74. [PMID: 35907556 DOI: 10.1016/j.actbio.2022.07.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 01/18/2023]
Abstract
Damaged or diseased bone can be treated using autografts or a range of different bone grafting biomaterials, however limitations with such approaches has motivated increased interest in developmentally inspired bone tissue engineering (BTE) strategies that seek to recapitulate the process of endochondral ossification (EO) as a means of regenerating critically sized defects. The clinical translation of such strategies will require the engineering of scaled-up, geometrically defined hypertrophic cartilage grafts that can be rapidly vascularised and remodelled into bone in mechanically challenging defect environments. The goal of this study was to 3D bioprint mechanically reinforced cartilaginous templates and to assess their capacity to regenerate critically sized femoral bone defects. Human mesenchymal stem/stromal cells (hMSCs) were incorporated into fibrin based bioinks and bioprinted into polycaprolactone (PCL) frameworks to produce mechanically reinforced constructs. Chondrogenic priming of such hMSC laden constructs was required to support robust vascularisation and graft mineralisation in vivo following their subcutaneous implantation into nude mice. With a view towards maximising their potential to support endochondral bone regeneration, we next explored different in vitro culture regimes to produce chondrogenic and early hypertrophic engineered grafts. Following their implantation into femoral bone defects within transiently immunosuppressed rats, such bioprinted constructs were rapidly remodelled into bone in vivo, with early hypertrophic constructs supporting higher levels of vascularisation and bone formation compared to the chondrogenic constructs. Such early hypertrophic bioprinted constructs also supported higher levels of vascularisation and spatially distinct patterns of new formation compared to BMP-2 loaded collagen scaffolds (here used as a positive control). In conclusion, this study demonstrates that fibrin based bioinks support chondrogenesis of hMSCs in vitro, which enables the bioprinting of mechanically reinforced hypertrophic cartilaginous templates capable of supporting large bone defect regeneration. These results support the use of 3D bioprinting as a strategy to scale-up the engineering of developmentally inspired templates for BTE. STATEMENT OF SIGNIFICANCE: Despite the promise of developmentally inspired tissue engineering strategies for bone regeneration, there are still challenges that need to be addressed to enable clinical translation. This work reports the development and assessment (in vitro and in vivo) of a 3D bioprinting strategy to engineer mechanically-reinforced cartilaginous templates for large bone defect regeneration using human MSCs. Using distinct in vitro priming protocols, it was possible to generate cartilage grafts with altered phenotypes. More hypertrophic grafts, engineered in vitro using TGF-β3 and BMP-2, supported higher levels of blood vessel infiltration and accelerated bone regeneration in vivo. This study also identifies some of the advantages and disadvantages of such endochondral bone TE strategies over the direct delivery of BMP-2 from collagen-based scaffolds.
Collapse
Affiliation(s)
- Pierluca Pitacco
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.
| | - Joanna M Sadowska
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland, Ireland.
| | - Fergal J O'Brien
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland, Ireland.
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine Royal College of Surgeons in Ireland, Ireland.
| |
Collapse
|
22
|
A composite, off-the-shelf osteoinductive material for large, vascularized bone flap prefabrication. Acta Biomater 2022; 154:641-649. [PMID: 36261107 DOI: 10.1016/j.actbio.2022.10.023] [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: 06/08/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
We previously described an immortalized, genetically-engineered human Mesenchymal stromal cell line to generate BMP2-enriched Chondrogenic Matrices (MB-CM), which after devitalization and storage could efficiently induce ectopic bone tissue by endochondral ossification. In order to increase the efficiency of MB-CM utilization towards engineering scaled-up bone structures, here we hypothesized that MB-CM can retain osteoinductive properties when combined with an osteoconductive material. We first tested different volumetric ratios of MB-CM:SmartBone® (as clinically used, osteoconductive reference material) and assessed the bone formation capacity of the resulting composites following ectopic mouse implantation. After 8 weeks, as little as 25% of MB-CM was sufficient to induce bone formation and fusion across SmartBone® granules, generating large interconnected bony structures. The same composite percentage was then further assessed in a scaled-up model, namely within an axially-vascularized, confined, ectopically prefabricated flap (0.8 cm3) in rats. The material efficiently induced the formation of new bone (31% of the cross-sectional area after 8 weeks), including bone marrow and vascular elements, throughout the flap volume. Our findings outline a strategy for efficient use of MB-CM as part of a composite material, thereby reducing the amount required to fill large spaces and enabling utilization in critically sized grafts, to address challenging clinical scenarios in bone reconstruction. STATEMENT OF SIGNIFICANCE: Most bone repair strategies rely either on osteconductive properties of ceramics and devitalized bone, or osteoinductive properties of growth factors and extracellular matrices (ECM). Here we designed a composite material made of a clinically accepted osteoconductive material and an off-the-shelf tissue engineered human cartilage ECM with strong osteoinductive properties. We showed that low amount of osteoinductive ECM potentiated host cells recruitment to form large vascularized bone structures in two different animal models, one being a challenging prefabricated bone-flap model targeting challenging clinical bone repair. Overall, this study highlights the use of a promising human off-the-shelf material for accelerated healing towards clinical applications.
Collapse
|
23
|
Zhang H, Li Q, Xu X, Zhang S, Chen Y, Yuan T, Zeng Z, Zhang Y, Mei Z, Yan S, Zhang L, Wei S. Functionalized Microscaffold-Hydrogel Composites Accelerating Osteochondral Repair through Endochondral Ossification. ACS APPLIED MATERIALS & INTERFACES 2022; 14:52599-52617. [PMID: 36394998 DOI: 10.1021/acsami.2c12694] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Osteochondral regeneration remains a key challenge because of the limited self-healing ability of the bone and its complex structure and composition. Biomaterials based on endochondral ossification (ECO) are considered an attractive candidate to promote bone repair because they can effectively address the difficulties in establishing vascularization and poor bone regeneration via intramembranous ossification (IMO). However, its clinical application is limited by the complex cellular behavior of ECO and the long time required for induction of the cell cycle. Herein, functionalized microscaffold-hydrogel composites are developed to accelerate the developmental bone growth process via recapitulating ECO. The design comprises arginine-glycine-aspartic acid (RGD)-peptide-modified microscaffolds loaded with kartogenin (KGN) and wrapped with a layer of RGD- and QK-peptide-comodified alginate hydrogel. These microscaffolds enhance the proliferation and aggregation behavior of the human bone marrow mesenchymal stem cells (hBMSCs); the controlled release of kartogenin induces the differentiation of hBMSCs into chondrocytes; and the hydrogel grafted with RGD and QK peptide facilitates chondrocyte hypertrophy, which creates a vascularized niche for osteogenesis and finally accelerates osteochondral repair in vivo. The findings provide an efficient bioengineering approach by sequentially modulating cellular ECO behavior for osteochondral defect repair.
Collapse
Affiliation(s)
- He Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Qian Li
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Xiangliang Xu
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Siqi Zhang
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Yang Chen
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| | - Tao Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Tumor Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ziqian Zeng
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Yifei Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Zi Mei
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Shuang Yan
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Lei Zhang
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
| | - Shicheng Wei
- Central Laboratory and Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Peking University, Beijing 100081, P.R. China
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, P.R. China
| |
Collapse
|
24
|
Nadine S, Fernandes IJ, Correia CR, Mano JF. Close-to-native bone repair via tissue-engineered endochondral ossification approaches. iScience 2022; 25:105370. [PMID: 36339269 PMCID: PMC9626746 DOI: 10.1016/j.isci.2022.105370] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In order to solve the clinical challenges related to bone grafting, several tissue engineering (TE) strategies have been proposed to repair critical-sized defects. Generally, the classical TE approaches are designed to promote bone repair via intramembranous ossification. Although promising, strategies that direct the osteogenic differentiation of mesenchymal stem/stromal cells are usually characterized by a lack of functional vascular supply, often resulting in necrotic cores. A less explored alternative is engineering bone constructs through a cartilage-mediated approach, resembling the embryological process of endochondral ossification. The remodeling of an intermediary hypertrophic cartilaginous template triggers vascular invasion and bone tissue deposition. Thus, employing this knowledge can be a promising direction for the next generation of bone TE constructs. This review highlights the most recent biomimetic strategies for applying endochondral ossification in bone TE while discussing the plethora of cell types, culture conditions, and biomaterials essential to promote a successful bone regeneration process.
Collapse
|
25
|
Hara K, Hellem E, Yamada S, Sariibrahimoglu K, Mølster A, Gjerdet NR, Hellem S, Mustafa K, Yassin MA. Efficacy of treating segmental bone defects through endochondral ossification: 3D printed designs and bone metabolic activities. Mater Today Bio 2022; 14:100237. [PMID: 35280332 PMCID: PMC8914554 DOI: 10.1016/j.mtbio.2022.100237] [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: 12/20/2021] [Revised: 02/15/2022] [Accepted: 03/05/2022] [Indexed: 10/25/2022] Open
Abstract
Three-dimensional printing (3D printing) is a promising technique for producing scaffolds for bone tissue engineering applications. Porous scaffolds can be printed directly, and the design, shape and porosity can be controlled. 3D synthetic biodegradable polymeric scaffolds intended for in situ bone regeneration must meet stringent criteria, primarily appropriate mechanical properties, good 3D design, adequate biocompatibility and the ability to enhance bone formation. In this study, healing of critical-sized (5 mm) femur defects of rats was enhanced by implanting two different designs of 3D printed poly(l-lactide-co-ε-caprolactone) (poly(LA-co-CL)) scaffolds seeded with rat bone marrow mesenchymal stem cells (rBMSC), which had been pre-differentiated in vitro into cartilage-forming chondrocytes. Depending on the design, the scaffolds had an interconnected porous structure of 300-500 μm and porosity of 50-65%. According to a computational simulation, the internal force distribution was consistent with scaffold designs and comparable between the two designs. Moreover, the defects treated with 3D-printed scaffolds seeded with chondrocyte-like cells exhibited significantly increased bone formation up to 15 weeks compared with empty defects. In all experimental animals, bone metabolic activity was monitored by positron emission tomography 1, 3, 5, 7, 11 and 14 weeks after surgery. This demonstrated a time-dependent relationship between scaffold design and metabolic activity. This confirmed that successful regeneration was highly reproducible. The in vitro and in vivo data indicated that the experimental setups had promising outcomes and could facilitate new bone formation through endochondral ossification.
Collapse
Affiliation(s)
- Kenji Hara
- Centre of Translational Oral Research (TOR) - Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
- Department of Oral and Maxillofacial Surgery, Fujieda Heisei Memorial Hospital, Japan
| | - Endre Hellem
- Centre of Translational Oral Research (TOR) - Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Shuntaro Yamada
- Centre of Translational Oral Research (TOR) - Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Kemal Sariibrahimoglu
- Centre of Translational Oral Research (TOR) - Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Anders Mølster
- Department of Clinical Medicine University of Bergen, Bergen, Norway
| | - Nils R Gjerdet
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Sølve Hellem
- Centre of Translational Oral Research (TOR) - Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Kamal Mustafa
- Centre of Translational Oral Research (TOR) - Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Mohammed A Yassin
- Centre of Translational Oral Research (TOR) - Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| |
Collapse
|
26
|
Complicated Mandible Fracture Treatment with Xenogenic Bone Graft. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12052384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The problem of filling bone cavities remains relevant in maxillofacial and oral surgery. There is a large selection of osteotropic materials, of various natures, for filling bone defects of different etiologies. The aim of our research was to improve the outcome of surgical treatment in a patient with a complicated mandibular fracture, with the use of a collagenic xenograft during osteosynthesis. In this article, we share our experience of the treatment of a patient with a complicated mandibular angle fracture, in combination with a follicular cyst. The obligate steps of treatment included stabilization of the bone fragments, decreasing the risk of fracture line malposition, using titan mini-plates, and shortening the time of bone regeneration, by filling the bone defect with osteotropic material. This approach allowed us to reduce the rehabilitation period and further prosthetic treatment after 4–5 months, without additional bone grafting manipulations. Thus, the use of collagen osteotropic materials, possessing osteoconductive properties, can improve the treatment of patients with mandibular fractures.
Collapse
|
27
|
On the horizon: Hedgehog signaling to heal broken bones. Bone Res 2022; 10:13. [PMID: 35165260 PMCID: PMC8844053 DOI: 10.1038/s41413-021-00184-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022] Open
Abstract
Uncovering the molecular pathways that drive skeletal repair has been an ongoing challenge. Initial efforts have relied on in vitro assays to identify the key signaling pathways that drive cartilage and bone differentiation. While these assays can provide some clues, assessing specific pathways in animal models is critical. Furthermore, definitive proof that a pathway is required for skeletal repair is best provided using genetic tests. Stimulating the Hh (Hedgehog) pathway can promote cartilage and bone differentiation in cell culture assays. In addition, the application of HH protein or various pathway agonists in vivo has a positive influence on bone healing. Until recently, however, genetic proof that the Hh pathway is involved in bone repair has been lacking. Here, we consider both in vitro and in vivo studies that examine the role of Hh in repair and discuss some of the challenges inherent in their interpretation. We also identify needed areas of study considering a new appreciation for the role of cartilage during repair, the variety of cell types that may have differing roles in repair, and the recent availability of powerful lineage tracing techniques. We are optimistic that emerging genetic tools will make it possible to precisely define when and in which cells promoting Hh signaling can best promote skeletal repair, and thus, the clinical potential for targeting the Hh pathway can be realized.
Collapse
|
28
|
Longoni A, Utomo L, Robinson A, Levato R, Rosenberg AJWP, Gawlitta D. Acceleration of Bone Regeneration Induced by a Soft-Callus Mimetic Material. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103284. [PMID: 34962103 PMCID: PMC8867155 DOI: 10.1002/advs.202103284] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/12/2021] [Indexed: 06/14/2023]
Abstract
Clinical implementation of endochondral bone regeneration (EBR) would benefit from the engineering of devitalized cartilaginous constructs of allogeneic origins. Nevertheless, development of effective devitalization strategies that preserves extracellular matrix (ECM) is still challenging. The aim of this study is to investigate EBR induced by devitalized, soft callus-mimetic spheroids. To challenge the translatability of this approach, the constructs are generated using an allogeneic cell source. Neo-bone formation is evaluated in an immunocompetent rat model, subcutaneously and in a critical size femur defect. Living spheroids are used as controls. Also, the effect of spheroid maturation towards hypertrophy is evaluated. The devitalization procedure successfully induces cell death without affecting ECM composition or bioactivity. In vivo, a larger amount of neo-bone formation is observed for the devitalized chondrogenic group both ectopically and orthotopically. In the femur defect, accelerated bone regeneration is observed in the devitalized chondrogenic group, where defect bridging is observed 4 weeks post-implantation. The authors' results show, for the first time, a dramatic increase in the rate of bone formation induced by devitalized soft callus-mimetics. These findings pave the way for the development of a new generation of allogeneic, "off-the-shelf" products for EBR, which are suitable for the treatment of every patient.
Collapse
Affiliation(s)
- Alessia Longoni
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht UniversityG05.222, PO Box 85500Utrecht3508 GAThe Netherlands
- Regenerative Medicine Center UtrechtUtrecht3584 CTThe Netherlands
| | - Lizette Utomo
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht UniversityG05.222, PO Box 85500Utrecht3508 GAThe Netherlands
- Regenerative Medicine Center UtrechtUtrecht3584 CTThe Netherlands
- Department of Clinical SciencesFaculty of Veterinary MedicineUtrecht UniversityYalelaan 108Utrecht3584CMThe Netherlands
| | - Abbie Robinson
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht UniversityG05.222, PO Box 85500Utrecht3508 GAThe Netherlands
- Regenerative Medicine Center UtrechtUtrecht3584 CTThe Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center UtrechtUtrecht3584 CTThe Netherlands
- Department of Clinical SciencesFaculty of Veterinary MedicineUtrecht UniversityYalelaan 108Utrecht3584CMThe Netherlands
- Department of OrthopaedicsUniversity Medical Center UtrechtUtrecht UniversityUtrecht3508 GAThe Netherlands
| | - Antoine J. W. P. Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht UniversityG05.222, PO Box 85500Utrecht3508 GAThe Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental CareUniversity Medical Center UtrechtUtrecht UniversityG05.222, PO Box 85500Utrecht3508 GAThe Netherlands
- Regenerative Medicine Center UtrechtUtrecht3584 CTThe Netherlands
| |
Collapse
|
29
|
Erickson CB, Hill R, Pascablo D, Kazakia G, Hansen K, Bahney C. A timeseries analysis of the fracture callus extracellular matrix proteome during bone fracture healing. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2021; 3:1-30. [PMID: 35765657 PMCID: PMC9236279 DOI: 10.36069/jols/20220601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
While most bones fully self-heal, certain diseases require bone allograft to assist with fracture healing. Bone allografts offer promise as treatments for such fractures due to their osteogenic properties. However, current bone allografts made of decellularized bone extracellular matrix (ECM) have high failure rates, and thus grafts which improve fracture healing outcomes are needed. Understanding specific changes to the ECM proteome during normal fracture healing would enable the identification of key proteins that could be used enhance osteogenicity of bone allograft. Here, we performed a timeseries analysis of the fracture callus in mice to investigate proteomic and mineralization changes to the ECM at key stages of fracture healing. We found that changes to the ECM proteome largely coincide with the distinct phases of fracture healing. Basement membrane proteins (AGRN, COL4, LAMA), cartilage proteins (COL2A1, ACAN), and collagen crosslinking enzymes (LOXL, PLOD, ITIH) were initially upregulated, followed by bone specific proteoglycans and collagens (IBSP, COL1A1). Various tissue proteases (MMP2, 9, 13, 14; CTSK, CTSG, ELANE) were expressed at different levels throughout fracture healing. These changes coordinated with mineralization of the fracture callus, which increased steeply during the initial stages of healing. Interestingly the later timepoint was characterized by a response to wound healing and high expression of clotting factors (F2, 7, 9, 10). We identified ELANE and ITIH2 as tissue remodeling enzymes having no prior known involvement with fracture healing. This data can be further mined to identify regenerative proteins for enhanced bone graft design.
Collapse
Affiliation(s)
- Christopher B. Erickson
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Ryan Hill
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Donna Pascablo
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA
| | - Galateia Kazakia
- Department of Radiology and Biomedical Imaging, University of California, San Francisco (UCSF), San Francisco, CA
| | - Kirk Hansen
- Department of Biochemistry and Molecular Genetics,University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Chelsea Bahney
- Stedman Philippon Research Institute (SPRI), Center for Regenerative and Personalized Medicine. Vail, CO
- Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA
| |
Collapse
|
30
|
Christen T, Krähenbühl SM, Müller CT, Durand S. Periosteal medial femoral condyle free flap for metacarpal nonunion. Microsurgery 2021; 42:226-230. [PMID: 34636060 PMCID: PMC9292408 DOI: 10.1002/micr.30826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/07/2021] [Accepted: 09/28/2021] [Indexed: 11/12/2022]
Abstract
Background Metacarpal nonunion is a rare condition. The osteogenic capacity of periosteal free flap was investigated in five patients with metacarpal nonunion and impaired bone vascularization. Patients and methods Surgery was performed between 64 and 499 days after the initial bone osteosynthesis. The average age was 27.6 (range 16–32) years. Nonunion was caused by excessive periosteal removal in two patients, extensive open trauma in three. Four nonunions were diaphyseal, one metaphyseal. A periosteal medial femoral condyle free flap was raised on the descending genicular artery for four patients, the superomedial genicular artery for one. After osteosynthesis with a plate, the flap was wrapped around the metacarpal, overlapping the bone proximally and distally. The recipient vessel were the dorsal branch of the radial artery and a vena comitans in the anatomical snuffbox in four patients, at the base of the first webspace in one. Results The flap size ranged from 5 × 3.5 cm to 8 × 4 cm. No postoperative complication occurred. Radiological bone union was achieved 3 to 8 months after surgery. One patient had a full range of motion, one a slight extension lag of the proximal interphalangeal joint, two moderate joint stiffness of the proximal interphalangeal or metacarpophalangeal joint (one requiring plate removal and extensor tenolysis), one severe stiffness that allowed using a hook grip which was the aim of the surgery. Conclusion In case of metacarpal nonunion with impaired bone vascularization, the periosteal medial femoral condyle free flap provides an effective and biomimetic approach to bone healing.
Collapse
Affiliation(s)
- Thierry Christen
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Swenn M Krähenbühl
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Camillo T Müller
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Sébastien Durand
- Department of Plastic and Hand Surgery, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
31
|
Arakura M, Lee SY, Fukui T, Oe K, Takahara S, Matsumoto T, Hayashi S, Matsushita T, Kuroda R, Niikura T. Endochondral Bone Tissue Engineering Using Human Induced Pluripotent Stem Cells. Tissue Eng Part A 2021; 28:184-195. [PMID: 34309415 DOI: 10.1089/ten.tea.2021.0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
There has been great interest in the use of induced pluripotent stem cells (iPSCs) in bone regenerative strategies for bone defects. In the present study, we investigated whether the implantation of chondrogenically differentiated iPSC-derived mesenchymal stem cells (iMSCs) could lead to the successful regeneration of bone defects in nude mice. Two clones of human iPSCs (201B7 and 454E2) were used. After the generation of iMSCs, chondrogenic differentiation was achieved using a three-dimensional pellet culture. Then, a 2-mm defect was created in the radius of nude mice and chondrogenically differentiated iMSC pellets were placed in the defect. Micro-computed tomography (μ-CT) imaging analysis was performed 8 weeks after transplantation to assess bone regeneration. Eleven out of 11 (100%) radii in the 201B7 cell-derived-pellet transplantation group and 7 out of 10 (70%) radii in the 454E2 cell-derived-pellet transplantation group showed bone union. On the other hand, only 2 out of 11 radii (18%) in the control group showed bone union. Therefore, the bone union rates in the experimental groups were significantly higher than that in the control group (p < 0.05). Histological analysis 2 weeks post-implantation in the experimental groups revealed hypertrophic chondrocytes within grafted iMSC pellets, and the formation of woven bone around them; this hypertrophic chondrocyte transitioning to the newly formed bone suggests that the cartilaginous template can trigger the process of endochondral bone ossification (ECO). Four weeks post-implantation, the cartilage template was reduced in size; newly formed woven bone predominated at the defect site. New vessels were surrounded by a matrix of woven bone and the hypertrophic chondrocytes transitioning to the newly formed bone indicated the progression of ECO. Eight weeks post-implantation, the pellets were completely resorbed and replaced by bone; complete bone union was overall observed. Dense mature bone developed with evidence of lamellar-like bone formation. Collectively, our results suggest that iMSC-based cartilage grafts recapitulating the morphogenetic process of ECO in the context of embryonic skeletogenesis are a novel and promising strategy for the repair of large bone defects.
Collapse
Affiliation(s)
- Michio Arakura
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Sang Yang Lee
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan.,Department of Orthopaedic Surgery, Showa University School of Medicine, Shinagawa-ku, Tokyo, Japan;
| | - Tomoaki Fukui
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Keisuke Oe
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Shunsuke Takahara
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Tomoyuki Matsumoto
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Shinya Hayashi
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Takehiko Matsushita
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Ryosuke Kuroda
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Hyogo, Japan;
| | - Takahiro Niikura
- Kobe University Graduate School of Medicine, Department of Orthopaedic Surgery, Kobe, Japan;
| |
Collapse
|
32
|
Effects of chondrogenic priming duration on mechanoregulation of engineered cartilage. J Biomech 2021; 125:110580. [PMID: 34198021 DOI: 10.1016/j.jbiomech.2021.110580] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/21/2022]
Abstract
Chondrocyte maturation during cartilage development occurs under diverse and dynamic mechanical environments. Mechanical stimulation through bioreactor culture may mimic these conditions to direct cartilage tissue engineering in vitro. Mechanical cues can promote chondrocyte homeostasis or hypertrophy and mineralization, depending potentially on the timing of load application. Here, we tested the effects of chondrogenic priming duration on the response of engineered human cartilage constructs to dynamic mechanical compression. We cultured human bone marrow stromal cells (hMSCs) in fibrin hydrogels under chondrogenic priming conditions for periods of 0, 2, 4, or 6 weeks prior to two weeks of either static culture or dynamic compression. We measured construct mechanical properties, cartilage matrix composition, and gene expression. Dynamic compression increased the equilibrium and dynamic modulus of the engineered tissue, depending on the duration of chondrogenic priming. For priming times of 2 weeks or greater, dynamic compression enhanced COL2A1 and AGGRECAN mRNA expression at the end of the loading period, but did not alter total collagen or glycosaminoglycan matrix deposition. Load initiation at priming times of 4 weeks or less repressed transient osteogenic signaling (RUNX2, OPN) and expression of CYR61, a YAP/TAZ-TEAD-target gene. No suppression of osteogenic gene expression was observed if loading was initiated after 6 weeks of in vitro priming, when mechanical stimulation was observed to increase the expression of type X collagen. Taken together, these data demonstrate that the duration of in vitro chondrogenic priming regulates the cell response to dynamic mechanical compression and suggests that early loading may preserve chondrocyte homeostasis while delayed loading may support cartilage maturation.
Collapse
|
33
|
Nulty J, Burdis R, Kelly DJ. Biofabrication of Prevascularised Hypertrophic Cartilage Microtissues for Bone Tissue Engineering. Front Bioeng Biotechnol 2021; 9:661989. [PMID: 34169064 PMCID: PMC8218548 DOI: 10.3389/fbioe.2021.661989] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Bone tissue engineering (TE) has the potential to transform the treatment of challenging musculoskeletal pathologies. To date, clinical translation of many traditional TE strategies has been impaired by poor vascularisation of the implant. Addressing such challenges has motivated research into developmentally inspired TE strategies, whereby implants mimicking earlier stages of a tissue's development are engineered in vitro and then implanted in vivo to fully mature into the adult tissue. The goal of this study was to engineer in vitro tissues mimicking the immediate developmental precursor to long bones, specifically a vascularised hypertrophic cartilage template, and to then assess the capacity of such a construct to support endochondral bone formation in vivo. To this end, we first developed a method for the generation of large numbers of hypertrophic cartilage microtissues using a microwell system, and encapsulated these microtissues into a fibrin-based hydrogel capable of supporting vasculogenesis by human umbilical vein endothelial cells (HUVECs). The microwells supported the formation of bone marrow derived stem/stromal cell (BMSC) aggregates and their differentiation toward a hypertrophic cartilage phenotype over 5 weeks of cultivation, as evident by the development of a matrix rich in sulphated glycosaminoglycan (sGAG), collagen types I, II, and X, and calcium. Prevascularisation of these microtissues, undertaken in vitro 1 week prior to implantation, enhanced their capacity to mineralise, with significantly higher levels of mineralised tissue observed within such implants after 4 weeks in vivo within an ectopic murine model for bone formation. It is also possible to integrate such microtissues into 3D bioprinting systems, thereby enabling the bioprinting of scaled-up, patient-specific prevascularised implants. Taken together, these results demonstrate the development of an effective strategy for prevascularising a tissue engineered construct comprised of multiple individual microtissue "building blocks," which could potentially be used in the treatment of challenging bone defects.
Collapse
Affiliation(s)
- Jessica Nulty
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Ross Burdis
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
34
|
Liu Y, Yang Z, Wang L, Sun L, Kim BYS, Jiang W, Yuan Y, Liu C. Spatiotemporal Immunomodulation Using Biomimetic Scaffold Promotes Endochondral Ossification-Mediated Bone Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100143. [PMID: 34105266 PMCID: PMC8188258 DOI: 10.1002/advs.202100143] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Indexed: 05/16/2023]
Abstract
Biomaterials play an important role in treating bone defects by promoting direct osteogenic healing through intramembranous ossification (IO). However, majority of the body's bones form via cartilaginous intermediates by endochondral ossification (EO), a process that has not been well mimicked by engineered scaffolds, thus limiting their clinical utility in treating large segmental bone defects. Here, by entrapping corticosteroid dexamethasone within biomimetic recombinant human bone morphogenetic protein (rhBMP)-loaded porous mesoporous bioglass scaffolds and regulating their release kinetics, significant degree of ectopic bone formation through endochondral ossification is achieved. By regulating the recruitment and polarization of immune suppressive macrophage phenotypes, the scaffold promotes rapid chondrogenesis by activating Hif-3α signaling pathway in mesenchymal stem cells, which upregulates the expression of downstream chondrogenic genes. Inhibition of Hif-3α signaling reverses the endochondral ossification phenotype. Together, these results reveal a strategy to facilitate developmental bone growth process using immune modulating biomimetic scaffolds, thus providing new opportunities for developing biomaterials capable of inducing natural tissue regeneration.
Collapse
Affiliation(s)
- Yutong Liu
- Key Laboratory for Ultrafine Materials of Ministry of EducationSchool of Materials Science and Engineering, and Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| | - Zhaogang Yang
- Department of Radiation OncologyUniversity of Texas Southwestern Medical CenterDallasTX75390USA
| | - Lixuan Wang
- Key Laboratory for Ultrafine Materials of Ministry of EducationSchool of Materials Science and Engineering, and Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| | - Lili Sun
- Key Laboratory for Ultrafine Materials of Ministry of EducationSchool of Materials Science and Engineering, and Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| | - Betty Y. S. Kim
- Department of NeurosurgeryThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Wen Jiang
- Department of Radiation OncologyUniversity of Texas Southwestern Medical CenterDallasTX75390USA
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of EducationSchool of Materials Science and Engineering, and Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of EducationSchool of Materials Science and Engineering, and Engineering Research Center for Biomedical Materials of Ministry of EducationEast China University of Science and TechnologyShanghai200237P. R. China
| |
Collapse
|
35
|
Dewey MJ, Harley BAC. Biomaterial design strategies to address obstacles in craniomaxillofacial bone repair. RSC Adv 2021; 11:17809-17827. [PMID: 34540206 PMCID: PMC8443006 DOI: 10.1039/d1ra02557k] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022] Open
Abstract
Biomaterial design to repair craniomaxillofacial defects has largely focused on promoting bone regeneration, while there are many additional factors that influence this process. The bone microenvironment is complex, with various mechanical property differences between cortical and cancellous bone, a unique porous architecture, and multiple cell types that must maintain homeostasis. This complex environment includes a vascular architecture to deliver cells and nutrients, osteoblasts which form new bone, osteoclasts which resorb excess bone, and upon injury, inflammatory cells and bacteria which can lead to failure to repair. To create biomaterials able to regenerate these large missing portions of bone on par with autograft materials, design of these materials must include methods to overcome multiple obstacles to effective, efficient bone regeneration. These obstacles include infection and biofilm formation on the biomaterial surface, fibrous tissue formation resulting from ill-fitting implants or persistent inflammation, non-bone tissue formation such as cartilage from improper biomaterial signals to cells, and voids in bone infill or lengthy implant degradation times. Novel biomaterial designs may provide approaches to effectively induce osteogenesis and new bone formation, include design motifs that facilitate surgical handling, intraoperative modification and promote conformal fitting within complex defect geometries, induce a pro-healing immune response, and prevent bacterial infection. In this review, we discuss the bone injury microenvironment and methods of biomaterial design to overcome these obstacles, which if unaddressed, may result in failure of the implant to regenerate host bone.
Collapse
Affiliation(s)
- Marley J. Dewey
- Dept of Materials Science and Engineering, University of Illinois at Urbana-ChampaignUrbanaIL 61801USA
| | - Brendan A. C. Harley
- Dept of Materials Science and Engineering, University of Illinois at Urbana-ChampaignUrbanaIL 61801USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-ChampaignUrbanaIL 61801USA
- Dept of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory600 S. Mathews AveUrbanaIL 61801USA+1-217-333-5052+1-217-244-7112
| |
Collapse
|
36
|
Xing Z, Jiang X, Si Q, Finne-Wistrand A, Liu B, Xue Y, Mustafa K. Endochondral Ossification Induced by Cell Transplantation of Endothelial Cells and Bone Marrow Stromal Cells with Copolymer Scaffold Using a Rat Calvarial Defect Model. Polymers (Basel) 2021; 13:polym13091521. [PMID: 34065081 PMCID: PMC8125936 DOI: 10.3390/polym13091521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
It has been recently reported that, in a rat calvarial defect model, adding endothelial cells (ECs) to a culture of bone marrow stromal cells (BMSCs) significantly enhanced bone formation. The aim of this study is to further investigate the ossification process of newly formed osteoid and host response to the poly(L-lactide-co-1,5-dioxepan-2-one) [poly(LLA-co-DXO)] scaffolds based on previous research. Several different histological methods and a PCR Array were applied to evaluate newly formed osteoid after 8 weeks after implantation. Histological results showed osteoid formed in rat calvarial defects and endochondral ossification-related genes, such as dentin matrix acidic phosphoprotein 1 (Dmp1) and collagen type II, and alpha 1 (Col2a1) exhibited greater expression in the CO (implantation with BMSC/EC/Scaffold constructs) than the BMSC group (implantation with BMSC/Scaffold constructs) as demonstrated by PCR Array. It was important to notice that cartilage-like tissue formed in the pores of the copolymer scaffolds. In addition, multinucleated giant cells (MNGCs) were observed surrounding the scaffold fragments. It was concluded that the mechanism of ossification might be an endochondral ossification process when the copolymer scaffolds loaded with co-cultured ECs/BMSCs were implanted into rat calvarial defects. MNGCs were induced by the poly(LLA-co-DXO) scaffolds after implantation, and more specific in vivo studies are needed to gain a better understanding of host response to copolymer scaffolds.
Collapse
Affiliation(s)
- Zhe Xing
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway;
| | - Xiaofeng Jiang
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
| | - Qingzong Si
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Teknikringen 56-58, SE 100-44 Stockholm, Sweden;
| | - Bin Liu
- School of Stomatology, Lanzhou University, Lanzhou 730000, China; (Z.X.); (X.J.); (Q.S.)
- Correspondence: (B.L.); (Y.X.); Tel.: +86-9318915051 (B.L.); +47-55586519 (Y.X.)
| | - Ying Xue
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway;
- Correspondence: (B.L.); (Y.X.); Tel.: +86-9318915051 (B.L.); +47-55586519 (Y.X.)
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, 5009 Bergen, Norway;
| |
Collapse
|
37
|
Burdis R, Kelly DJ. Biofabrication and bioprinting using cellular aggregates, microtissues and organoids for the engineering of musculoskeletal tissues. Acta Biomater 2021; 126:1-14. [PMID: 33711529 DOI: 10.1016/j.actbio.2021.03.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022]
Abstract
The modest clinical impact of musculoskeletal tissue engineering (TE) can be attributed, at least in part, to a failure to recapitulate the structure, composition and functional properties of the target tissue. This has motivated increased interest in developmentally inspired TE strategies, which seek to recapitulate key events that occur during embryonic and post-natal development, as a means of generating truly biomimetic grafts to replace or regenerate damaged tissues and organs. Such TE strategies can be substantially enabled by emerging biofabrication and bioprinting strategies, and in particular the use of cellular aggregates, microtissues and organoids as 'building blocks' for the development of larger tissues and/or organ precursors. Here, the application of such biological building blocks for the engineering of musculoskeletal tissues, from vascularised bone to zonally organised articular cartilage, will be reviewed. The importance of first scaling-down to later scale-up will be discussed, as this is viewed as a key component of engineering functional grafts using cellular aggregates or microtissues. In the context of engineering anatomically accurate tissues of scale suitable for tissue engineering and regenerative medicine applications, novel bioprinting modalities and their application in controlling the process by which cellular aggregates or microtissues fuse and self-organise will be reviewed. Throughout the paper, we will highlight some of the key challenges facing this emerging field. STATEMENT OF SIGNIFICANCE: The field of bioprinting has grown substantially in recent years, but despite the hype and excitement it has generated, there are relatively few examples of bioprinting strategies producing implants with superior regenerative potential to that achievable with more traditional tissue engineering approaches. This paper provides an up-to-date review of emerging biofabrication and bioprinting strategies which use cellular aggregates and microtissues as 'building blocks' for the development of larger musculoskeletal tissues and/or organ precursors - a field of research that can potentially enable functional regeneration of damaged and diseased tissues. The application of cellular aggregates and microtissues for the engineering of musculoskeletal tissues, from vascularised bone to zonally organised articular cartilage, will be reviewed. In the context of engineering anatomically accurate tissues of scale, novel bioprinting modalities and their application in controlling the process by which cellular aggregates or microtissues self-organise is addressed, as well as key challenges facing this emerging field.
Collapse
|
38
|
Teng B, Zhang S, Pan J, Zeng Z, Chen Y, Hei Y, Fu X, Li Q, Ma M, Sui Y, Wei S. A chondrogenesis induction system based on a functionalized hyaluronic acid hydrogel sequentially promoting hMSC proliferation, condensation, differentiation, and matrix deposition. Acta Biomater 2021; 122:145-159. [PMID: 33444801 DOI: 10.1016/j.actbio.2020.12.054] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Hydrogel scaffolds are widely used in cartilage tissue engineering as a natural stem cell niche. In particular, hydrogels based on multiple biological signals can guide behaviors of mesenchymal stem cells (MSCs) during neo-chondrogenesis. In the first phase of this study, we showed that functionalized hydrogels with grafted arginine-glycine-aspartate (RGD) peptides and lower degree of crosslinking can promote the proliferation of human mesenchymal stem cells (hMSCs) and upregulate the expression of cell receptor proteins. Moreover, grafted RGD and histidine-alanine-valine (HAV) peptides in hydrogel scaffolds can regulate the adhesion of the intercellular at an early stage. In the second phase, we confirmed that simultaneous use of HAV and RGD peptides led to greater chondrogenic differentiation compared to the blank control and single-peptide groups. Furthermore, the controlled release of kartogenin (KGN) can better facilitate cell chondrogenesis compared to other groups. Interestingly, with longer culture time, cell condensation was clearly observed in the groups with RGD and HAV peptide. In all groups with RGD peptide, significant matrix deposition was observed, accompanied by glycosaminoglycan (GAG) and collagen (Coll) production. Through in vitro and in vivo experiments, this study confirmed that our hydrogel system can sequentially promote the proliferation, adhesion, condensation, chondrogenic differentiation of hMSCs, by mimicking the cell microenvironment during neo-chondrogenesis.
Collapse
|
39
|
Decellularized bone extracellular matrix in skeletal tissue engineering. Biochem Soc Trans 2021; 48:755-764. [PMID: 32369551 DOI: 10.1042/bst20190079] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
Bone possesses an intrinsic regenerative capacity, which can be compromised by aging, disease, trauma, and iatrogenesis (e.g. tumor resection, pharmacological). At present, autografts and allografts are the principal biological treatments available to replace large bone segments, but both entail several limitations that reduce wider use and consistent success. The use of decellularized extracellular matrices (ECM), often derived from xenogeneic sources, has been shown to favorably influence the immune response to injury and promote site-appropriate tissue regeneration. Decellularized bone ECM (dbECM), utilized in several forms - whole organ, particles, hydrogels - has shown promise in both in vitro and in vivo animal studies to promote osteogenic differentiation of stem/progenitor cells and enhance bone regeneration. However, dbECM has yet to be investigated in clinical studies, which are needed to determine the relative efficacy of this emerging biomaterial as compared with established treatments. This mini-review highlights the recent exploration of dbECM as a biomaterial for skeletal tissue engineering and considers modifications on its future use to more consistently promote bone regeneration.
Collapse
|
40
|
Papantoniou I, Nilsson Hall G, Loverdou N, Lesage R, Herpelinck T, Mendes L, Geris L. Turning Nature's own processes into design strategies for living bone implant biomanufacturing: a decade of Developmental Engineering. Adv Drug Deliv Rev 2021; 169:22-39. [PMID: 33290762 PMCID: PMC7839840 DOI: 10.1016/j.addr.2020.11.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 11/20/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022]
Abstract
A decade after the term developmental engineering (DE) was coined to indicate the use of developmental processes as blueprints for the design and development of engineered living implants, a myriad of proof-of-concept studies demonstrate the potential of this approach in small animal models. This review provides an overview of DE work, focusing on applications in bone regeneration. Enabling technologies allow to quantify the distance between in vitro processes and their developmental counterpart, as well as to design strategies to reduce that distance. By embedding Nature's robust mechanisms of action in engineered constructs, predictive large animal data and subsequent positive clinical outcomes can be gradually achieved. To this end, the development of next generation biofabrication technologies should provide the necessary scale and precision for robust living bone implant biomanufacturing.
Collapse
Affiliation(s)
- Ioannis Papantoniou
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology - Hellas (FORTH), Stadiou street, 26504 Patras, Greece; Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Gabriella Nilsson Hall
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Niki Loverdou
- Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium; GIGA in silico medicine, University of Liège, Avenue de l'Hôpital 11 (B34), 4000 Liège, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3001 Leuven, Belgium.
| | - Raphaelle Lesage
- Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3001 Leuven, Belgium.
| | - Tim Herpelinck
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Luis Mendes
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium.
| | - Liesbet Geris
- Skeletal Biology & Engineering Research Center, KU Leuven, Herestraat 49 (813), 3000 Leuven, Belgium; GIGA in silico medicine, University of Liège, Avenue de l'Hôpital 11 (B34), 4000 Liège, Belgium; Prometheus, The KU Leuven R&D Division for Skeletal Tissue Engineering, Herestraat 49 (813), 3000 Leuven, Belgium; Biomechanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3001 Leuven, Belgium.
| |
Collapse
|
41
|
Panda M, Tripathi SK, Biswal BK. SOX9: An emerging driving factor from cancer progression to drug resistance. Biochim Biophys Acta Rev Cancer 2021; 1875:188517. [PMID: 33524528 DOI: 10.1016/j.bbcan.2021.188517] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Dysregulation of transcription factors is one of the common problems in the pathogenesis of human cancer. Among them, SOX9 is one of the critical transcription factors involved in various diseases, including cancer. The expression of SOX9 is regulated by microRNAs (miRNAs), methylation, phosphorylation, and acetylation. Interestingly, SOX9 acts as a proto-oncogene or tumor suppressor gene, relying upon kinds of cancer. Recent studies have reported the critical role of SOX9 in the regulation of the tumor microenvironment (TME). Additionally, activation of SOX9 signaling or SOX9 regulated signaling pathways play a crucial role in cancer development and progression. Accumulating evidence also suggests that SOX9 acquires stem cell features to induce epithelial-mesenchymal transition (EMT). Moreover, SOX9 has been broadly studied in the field of cancer stem cell (CSC) and EMT in the last decades. However, the link between SOX9 and cancer drug resistance has only recently been discovered. Furthermore, its differential expression could be a potential biomarker for tumor prognosis and progression. This review outlined the various biological implications of SOX9 in cancer progression and cancer drug resistance and elucidated its signaling network, which could be a potential target for designing novel anticancer drugs.
Collapse
Affiliation(s)
- Munmun Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Surya Kant Tripathi
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India.
| |
Collapse
|
42
|
Sun L, Ma Y, Niu H, Liu Y, Yuan Y, Liu C. Recapitulation of In Situ Endochondral Ossification Using an Injectable Hypoxia‐Mimetic Hydrogel. ADVANCED FUNCTIONAL MATERIALS 2021; 31. [DOI: 10.1002/adfm.202008515] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Indexed: 01/06/2025]
Abstract
AbstractDue to the limited ability for perfusion, traditional intramembranous ossification (IMO) often fails to recapitulate the natural regeneration process of most long bones and craniofacial bones. Alternatively, endochondral ossification (ECO) strategy has emerged and has been evidenced to circumvent the drawbacks in the routine application of IMO. Here, an injectable, poly(glycerol sebacate)‐co‐poly (ethylene glycol)/polyacrylic acid (PEGS/PAA) hydrogels are successfully developed to induce a hypoxia‐mimicking environment and subsequently recapitulate ECO via in situ iron chelation. With the incorporation of PAA, these hydrogels present remarkable viscoelasticity and high efficacy of iron ion‐chelating after injection, giving rise to the activation of HIF‐1α signaling pathway and suppression of inflammatory responses, and thereby improving chondrogenic differentiation in the early stage and facilitating vascularization in the later stage, which consequently trigger typical ECO. More importantly, through sustained and stable expression of HIF‐1α regulated by PEGS/PAA hydrogels throughout the regeneration, a harmonious chondrogenic/osteogenic balance can be struck and thereby accelerating the progress of ECO compared to the PEGS. The findings provide an efficient strategy to achieve in situ ECO via biomaterial‐based iron ion‐chelating and ensuing hypoxia‐mimicking, representing a novel and promising concept for future application in bone regeneration.
Collapse
Affiliation(s)
- Lili Sun
- Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yifan Ma
- Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Haoyi Niu
- Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yutong Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Yuan Yuan
- Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering East China University of Science and Technology Shanghai 200237 P. R. China
- Engineering Research Center for Biomedical Materials of Ministry of Education East China University of Science and Technology Shanghai 200237 P. R. China
| |
Collapse
|
43
|
Ma R, Liu S, Qiao T, Li D, Zhang R, Guo X. Fluoride Inhibits Longitudinal Bone Growth by Acting Directly at the Growth Plate in Cultured Neonatal Rat Metatarsal Bones. Biol Trace Elem Res 2020; 197:522-532. [PMID: 31838736 DOI: 10.1007/s12011-019-01997-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/27/2019] [Indexed: 02/03/2023]
Abstract
Excessive intake of fluoride inhibits bone growth in both humans and animals. It is unknown whether fluoride acts directly on the growth plate to inhibit longitudinal bone growth, and its mechanism of action has not been elucidated. In this study, we used an organ culture system and SW1353 cells to evaluate the effects of fluoride on endochondral ossification. Neonatal rat metatarsal bones were dissected and cultured with or without fluoride for 7 days. The total length and width of the metatarsal rudiments and the length of the calcification zone were measured. Chondrocyte proliferation, differentiation, and apoptosis were analyzed by immunohistochemistry and TUNEL assay in sectioned bones. The apoptosis was detected by flow cytometry, and the expression of apoptosis-related proteins Bax, Bcl-2, and Caspase-3 were detected by western blotting in SW1353 cells. Linear measurements demonstrated that fluoride induced a biphasic effect on longitudinal bone growth in organ culture, with a significant growth inhibition at a high concentration (10-4 M) and a stimulatory action at low concentration (10-6 M) of fluoride. Histomorphometrical analysis of growth plate from fluoride-exposed metatarsal rudiments showed a significant reduction in the height of the proliferative and hypertrophic chondrocyte zones. Analysis of the Col2α1 and Col10α1 expression by immunohistochemistry revealed fluoride-suppressed metatarsal growth plate chondrocyte proliferation and differentiation. In addition, fluoride increased the number of apoptotic chondrocytes in the metatarsal growth plate. Western blotting showed an up-regulated expression of Caspase-3 and Bax and down-regulated expression of anti-apoptotic protein Bcl-2 after treatment with 5 × 10-4 M fluoride in SW1353 cells. Our findings indicated that fluoride inhibited longitudinal bone growth by acting directly at the growth plate in cultured neonatal rat metatarsal bones. Such growth inhibition was mediated by suppressing proliferation and differentiation, increasing apoptosis of resting chondrocytes and causing premature cell senescence in the growth plate.
Collapse
Affiliation(s)
- Rui Ma
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, People's Republic of China
| | - Shuang Liu
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, People's Republic of China
| | - Tingting Qiao
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, People's Republic of China
| | - Demin Li
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, People's Republic of China
| | - Ruixue Zhang
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, People's Republic of China
| | - Xiaoying Guo
- Department of Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, People's Republic of China.
| |
Collapse
|
44
|
Pigeot S, Bourgine PE, Claude J, Scotti C, Papadimitropoulos A, Todorov A, Epple C, Peretti GM, Martin I. Orthotopic Bone Formation by Streamlined Engineering and Devitalization of Human Hypertrophic Cartilage. Int J Mol Sci 2020; 21:ijms21197233. [PMID: 33008121 PMCID: PMC7582540 DOI: 10.3390/ijms21197233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/26/2020] [Indexed: 12/25/2022] Open
Abstract
Most bones of the human body form and heal through endochondral ossification, whereby hypertrophic cartilage (HyC) is formed and subsequently remodeled into bone. We previously demonstrated that HyC can be engineered from human mesenchymal stromal cells (hMSC), and subsequently devitalized by apoptosis induction. The resulting extracellular matrix (ECM) tissue retained osteoinductive properties, leading to ectopic bone formation. In this study, we aimed at engineering and devitalizing upscaled quantities of HyC ECM within a perfusion bioreactor, followed by in vivo assessment in an orthotopic bone repair model. We hypothesized that the devitalized HyC ECM would outperform a clinical product currently used for bone reconstructive surgery. Human MSC were genetically engineered with a gene cassette enabling apoptosis induction upon addition of an adjuvant. Engineered hMSC were seeded, differentiated, and devitalized within a perfusion bioreactor. The resulting HyC ECM was subsequently implanted in a 10-mm rabbit calvarial defect model, with processed human bone (Maxgraft®) as control. Human MSC cultured in the perfusion bioreactor generated a homogenous HyC ECM and were efficiently induced towards apoptosis. Following six weeks of in vivo implantation, microcomputed tomography and histological analyses of the defects revealed an increased bone formation in the defects filled with HyC ECM as compared to Maxgraft®. This work demonstrates the suitability of engineered devitalized HyC ECM as a bone substitute material, with a performance superior to a state-of-the-art commercial graft. Streamlined generation of the devitalized tissue transplant within a perfusion bioreactor is relevant towards standardized and automated manufacturing of a clinical product.
Collapse
Affiliation(s)
- Sébastien Pigeot
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Paul Emile Bourgine
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Jaquiery Claude
- Department of Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (J.C.); (C.E.)
| | - Celeste Scotti
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland;
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
| | - Adam Papadimitropoulos
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Atanas Todorov
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
| | - Christian Epple
- Department of Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (J.C.); (C.E.)
| | - Giuseppe M. Peretti
- IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy;
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (S.P.); (P.E.B.); (A.P.); (A.T.)
- Department of Surgery, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; (J.C.); (C.E.)
- Correspondence:
| |
Collapse
|
45
|
Ko FC, Sumner DR. How faithfully does intramembranous bone regeneration recapitulate embryonic skeletal development? Dev Dyn 2020; 250:377-392. [PMID: 32813296 DOI: 10.1002/dvdy.240] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/19/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Postnatal intramembranous bone regeneration plays an important role during a wide variety of musculoskeletal regeneration processes such as fracture healing, joint replacement and dental implant surgery, distraction osteogenesis, stress fracture healing, and repair of skeletal defects caused by trauma or resection of tumors. The molecular basis of intramembranous bone regeneration has been interrogated using rodent models of most of these conditions. These studies reveal that signaling pathways such as Wnt, TGFβ/BMP, FGF, VEGF, and Notch are invoked, reminiscent of embryonic development of membranous bone. Discoveries of several skeletal stem cell/progenitor populations using mouse genetic models also reveal the potential sources of postnatal intramembranous bone regeneration. The purpose of this review is to compare the underlying molecular signals and progenitor cells that characterize embryonic development of membranous bone and postnatal intramembranous bone regeneration.
Collapse
Affiliation(s)
- Frank C Ko
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - D Rick Sumner
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
46
|
Longoni A, Pennings I, Cuenca Lopera M, van Rijen MHP, Peperzak V, Rosenberg AJWP, Levato R, Gawlitta D. Endochondral Bone Regeneration by Non-autologous Mesenchymal Stem Cells. Front Bioeng Biotechnol 2020; 8:651. [PMID: 32733861 PMCID: PMC7363768 DOI: 10.3389/fbioe.2020.00651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/27/2020] [Indexed: 12/31/2022] Open
Abstract
Mimicking endochondral bone formation is a promising strategy for bone regeneration. To become a successful therapy, the cell source is a crucial translational aspect. Typically, autologous cells are used. The use of non-autologous mesenchymal stromal cells (MSCs) represents an interesting alternative. Nevertheless, non-autologous, differentiated MSCs may trigger an undesired immune response, hampering bone regeneration. The aim of this study was to unravel the influence of the immune response on endochondral bone regeneration, when using xenogeneic (human) or allogeneic (Dark Agouti) MSCs. To this end, chondrogenically differentiated MSCs embedded in a collagen carrier were implanted in critical size femoral defects of immunocompetent Brown Norway rats. Control groups were included with syngeneic/autologous (Brown Norway) MSCs or a cell-free carrier. The amount of neo-bone formation was proportional to the degree of host-donor relatedness, as no full bridging of the defect was observed in the xenogeneic group whereas 2/8 and 7/7 bridges occurred in the allogeneic and the syngeneic group, respectively. One week post-implantation, the xenogeneic grafts were invaded by pro-inflammatory macrophages, T lymphocytes, which persisted after 12 weeks, and anti-human antibodies were developed. The immune response toward the allogeneic graft was comparable to the one evoked by the syngeneic implants, aside from an increased production of alloantibodies, which might be responsible for the more heterogeneous bone formation. Our results demonstrate for the first time the feasibility of using non-autologous MSC-derived chondrocytes to elicit endochondral bone regeneration in vivo. Nevertheless, the pronounced immune response and the limited bone formation observed in the xenogeneic group undermine the clinical relevance of this group. On the contrary, although further research on how to achieve robust bone formation with allogeneic cells is needed, they may represent an alternative to autologous transplantation.
Collapse
Affiliation(s)
- Alessia Longoni
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - I Pennings
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - Marta Cuenca Lopera
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - M H P van Rijen
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Victor Peperzak
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - A J W P Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
47
|
Kim HS, Mandakhbayar N, Kim HW, Leong KW, Yoo HS. Protein-reactive nanofibrils decorated with cartilage-derived decellularized extracellular matrix for osteochondral defects. Biomaterials 2020; 269:120214. [PMID: 32736808 DOI: 10.1016/j.biomaterials.2020.120214] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/06/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022]
Abstract
Cartilage defect is difficult to heal due to its avascular properties. Implantation of mesenchymal stem cell is one of the most promising approach for regenerating cartilage defects. Here we prepared polymeric nanofibrils decorated with cartilage-derived decellularized extracellular matrix (dECM) as a chondroinductive scaffold material for cartilage repair. To fabricate nanofibrils, eletrospun PCL nanofibers were fragmented by subsequent mechanical and chemical process. The nanofibrils were surface-modified with poly(glycidyl methacrylate) (PGMA@NF) via surface-initiated atom transfer radical polymerization (SI-ATRP). The epoxy groups of PGMA@NF were subsequently reacted with dECM prepared from bovine articular cartilage. Therefore, the cartilage-dECM-decorated nanofibrils structurally and biochemically mimic cartilage-specific microenvironment. Once adipose-derived stem cells (ADSCs) were self-assembled with the cartilage-dECM-decorated nanofibrils by cell-directed association, they exhibited differentiation hallmarks of chondrogenesis without additional biologic additives. ADSCs in the nanofibril composites significantly increased expression of chondrogenic gene markers in comparison to those in pellet culture. Furthermore, ADSC-laden nanofibril composites filled the osteochondral defects compactly due to their clay-like texture. Thus, the ADSC-laden nanofibril composites supported the long-term regeneration of 12 weeks without matrix loss during joint movement. The defects treated with the ADSC-laden PGMA@NF significantly facilitated reconstruction of their cartilage and subchondral bone ECM matrices compared to those with ADSC-laden nanofibrils, non-specifically adsorbing cartilage-dECM without surface decoration of PGMA.
Collapse
Affiliation(s)
- Hye Sung Kim
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomateials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Hyuk Sang Yoo
- Department of Biomedical Materials Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute of Molecular Science and Fusion Technology, Kangwon National University, Republic of Korea.
| |
Collapse
|
48
|
Abstract
The modification of implant devices with biocompatible coatings has become necessary as a consequence of premature loosening of prosthesis. This is caused mainly by chronic inflammation or allergies that are triggered by implant wear, production of abrasion particles, and/or release of metallic ions from the implantable device surface. Specific to the implant tissue destination, it could require coatings with specific features in order to provide optimal osseointegration. Pulsed laser deposition (PLD) became a well-known physical vapor deposition technology that has been successfully applied to a large variety of biocompatible inorganic coatings for biomedical prosthetic applications. Matrix assisted pulsed laser evaporation (MAPLE) is a PLD-derived technology used for depositions of thin organic material coatings. In an attempt to surpass solvent related difficulties, when different solvents are used for blending various organic materials, combinatorial MAPLE was proposed to grow thin hybrid coatings, assembled in a gradient of composition. We review herein the evolution of the laser technological process and capabilities of growing thin bio-coatings with emphasis on blended or multilayered biomimetic combinations. These can be used either as implant surfaces with enhanced bioactivity for accelerating orthopedic integration and tissue regeneration or combinatorial bio-platforms for cancer research.
Collapse
|
49
|
Raftery RM, Gonzalez Vazquez AG, Chen G, O'Brien FJ. Activation of the SOX-5, SOX-6, and SOX-9 Trio of Transcription Factors Using a Gene-Activated Scaffold Stimulates Mesenchymal Stromal Cell Chondrogenesis and Inhibits Endochondral Ossification. Adv Healthc Mater 2020; 9:e1901827. [PMID: 32329217 DOI: 10.1002/adhm.201901827] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/18/2020] [Indexed: 02/02/2023]
Abstract
Current treatments for articular cartilage defects relieve symptoms but often only delay cartilage degeneration. Mesenchymal stem cells (MSCs) have shown chondrogenic potential but tend to undergo endochondral ossification when implanted in vivo. Harnessing factors governing joint development to functionalize biomaterial scaffolds, termed developmental engineering, might allow to prime host MSCs to regenerate mature articular cartilage in situ without requiring cell isolation or ex vivo expansion. Therefore, the aim of this study is to develop a gene-activated scaffold capable of delivering developmental cues to host MSCs, thus priming MSCs for articular cartilage differentiation and inhibiting endochondral ossification. It is shown that delivery of the SOX-Trio induced MSCs to over-express COL2A1 and ACAN and deposit a sulfated and collagen type II rich extracellular matrix while hypertrophic gene expression and collagen type X deposition is inhibited. When cell-free SOX-Trio-activated scaffolds are implanted ectopically in vivo, they induced spontaneous chondrogenesis without evidence of hypertrophy. MSCs pre-cultured on SOX-Trio-activated scaffolds prior to implantation differentiate into phenotypically stable chondrocytes as evidenced by a lack of collagen X expression or vascular invasion. This SOX-trio-activated scaffold represents a potent, single treatment, developmentally inspired strategy to prime MSCs in situ for articular cartilage defect repair.
Collapse
Affiliation(s)
- Rosanne M. Raftery
- Tissue Engineering Research GroupDepartment of Anatomy and Regenerative MedicineRoyal College of Surgeons in Ireland Dublin D02 YN77 Ireland
- Trinity Centre for Biomedical Engineering (TCBE)Trinity College Dublin Dublin 2 Dublin D02 R590 Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)RCSI and TCD Dublin D02 YN77 Ireland
| | - Arlyng G. Gonzalez Vazquez
- Tissue Engineering Research GroupDepartment of Anatomy and Regenerative MedicineRoyal College of Surgeons in Ireland Dublin D02 YN77 Ireland
- Trinity Centre for Biomedical Engineering (TCBE)Trinity College Dublin Dublin 2 Dublin D02 R590 Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)RCSI and TCD Dublin D02 YN77 Ireland
| | - Gang Chen
- Department of Physiology and Medical PhysicsCentre for the Study of Neurological DisordersMicrosurgical Research and Training Facility (MRTF)Royal College of Surgeons in Ireland Dublin D02 YN77 Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research GroupDepartment of Anatomy and Regenerative MedicineRoyal College of Surgeons in Ireland Dublin D02 YN77 Ireland
- Trinity Centre for Biomedical Engineering (TCBE)Trinity College Dublin Dublin 2 Dublin D02 R590 Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)RCSI and TCD Dublin D02 YN77 Ireland
| |
Collapse
|
50
|
Dewey MJ, Nosatov AV, Subedi K, Harley B. Anisotropic mineralized collagen scaffolds accelerate osteogenic response in a glycosaminoglycan-dependent fashion. RSC Adv 2020; 10:15629-15641. [PMID: 32655857 PMCID: PMC7351350 DOI: 10.1039/d0ra01336f] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Regeneration of critically-sized craniofacial bone defects requires a template to promote cell activity and bone remodeling. However, induced regeneration becomes more challenging with increasing defect size. Methods of repair using allografts and autografts have inconsistent results, attributed to age-related regenerative capabilities of bone. We are developing a mineralized collagen scaffold to promote craniomaxillofacial bone regeneration as an alternative to repair. Here, we hypothesize modifying the pore anisotropy and glycosaminoglycan content of the scaffold will improve cell migration, viability, and subsequent bone formation. Using anisotropic and isotropic scaffold variants, we test the role of pore orientation on human mesenchymal stem cell (MSC) activity. We subsequently explore the role of glycosaminoglycan content, notably chondroitin-6-sulfate, chondroitin-4-sulfate, and heparin sulfate on mineralization. We find that while short term MSC migration and activity was not affected by pore orientation, increased bone mineral synthesis was observed in anisotropic scaffolds. Further, while scaffold glycosaminoglycan content did not impact cell viability, heparin sulfate and chondroitin-6-sulfate containing variants increased mineral formation at the late stage of in vitro culture, respectively. Overall, these findings show scaffold microstructural and proteoglycan modifications represent a powerful tool to improve MSC osteogenic activity. Mineralized collagen scaffolds were modified to include anisotropic pore architecture and one of three glycosaminoglycans in order to improve bone mineral formation in vitro.![]()
Collapse
Affiliation(s)
| | | | | | - Brendan Harley
- Dept. of Materials Science and Engineering, USA.,School of Chemical Sciences, USA.,Dept. Chemical and Biomolecular Engineering, USA.,Dept. of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory 600 S. Mathews Ave., Urbana, IL 61801, USA
| |
Collapse
|