1
|
Ge R, Liu C, Zhao Y, Wang K, Wang X. Endochondral Ossification for Spinal Fusion: A Novel Perspective from Biological Mechanisms to Clinical Applications. J Pers Med 2024; 14:957. [PMID: 39338212 PMCID: PMC11433020 DOI: 10.3390/jpm14090957] [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: 06/04/2024] [Revised: 08/07/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Degenerative scoliosis (DS), encompassing conditions like spondylolisthesis and spinal stenosis, is a common type of spinal deformity. Lumbar interbody fusion (LIF) stands as a conventional surgical intervention for this ailment, aiming at decompression, restoration of intervertebral height, and stabilization of motion segments. Despite its widespread use, the precise mechanism underlying spinal fusion remains elusive. In this review, our focus lies on endochondral ossification for spinal fusion, a process involving vertebral development and bone healing. Endochondral ossification is the key step for the successful vertebral fusion. Endochondral ossification can persist in hypoxic conditions and promote the parallel development of angiogenesis and osteogenesis, which corresponds to the fusion process of new bone formation in the hypoxic region between the vertebrae. The ideal material for interbody fusion cages should have the following characteristics: (1) Good biocompatibility; (2) Stable chemical properties; (3) Biomechanical properties similar to bone tissue; (4) Promotion of bone fusion; (5) Favorable for imaging observation; (6) Biodegradability. Utilizing cartilage-derived bone-like constructs holds promise in promoting bony fusion post-operation, thus warranting exploration in the context of spinal fusion procedures.
Collapse
Affiliation(s)
- Rile Ge
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong An Rd, Beijing 100050, China;
| | - Chenjun Liu
- Department of Spinal Surgery, Peking University People’s Hospital, 11th Xizhimen South Ave., Beijing 100044, China;
| | - Yuhong Zhao
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, China;
| | - Kaifeng Wang
- Department of Spinal Surgery, Peking University People’s Hospital, 11th Xizhimen South Ave., Beijing 100044, China;
| | - Xiluan Wang
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, China;
| |
Collapse
|
2
|
Raftery RM, Gonzalez Vazquez AG, Walsh DP, Chen G, Laiva AL, Keogh MB, O'Brien FJ. Mobilizing Endogenous Progenitor Cells Using pSDF1α-Activated Scaffolds Accelerates Angiogenesis and Bone Repair in Critical-Sized Bone Defects. Adv Healthc Mater 2024; 13:e2401031. [PMID: 38850118 DOI: 10.1002/adhm.202401031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/05/2024] [Indexed: 06/09/2024]
Abstract
Mobilizing endogenous progenitor cells to repair damaged tissue in situ has the potential to revolutionize the field of regenerative medicine, while the early establishment of a vascular network will ensure survival of newly generated tissue. In this study, a gene-activated scaffold containing a stromal derived factor 1α plasmid (pSDF1α), a pro-angiogenic gene that is also thought to be involved in the recruitment of mesenchymal stromal cells (MSCs) to sites of injury is described. It is shown that over-expression of SDF1α protein enhanced MSC recruitment and induced vessel-like structure formation by endothelial cells in vitro. When implanted subcutaneously, transcriptomic analysis reveals that endogenous MSCs are recruited and significant angiogenesis is stimulated. Just 1-week after implantation into a calvarial critical-sized bone defect, pSDF1α-activated scaffolds are recruited MSCs and rapidly activate angiogenic and osteogenic programs, upregulating Runx2, Dlx5, and Sp7. At the same time-point, pVEGF-activated scaffolds are recruited a variety of cell types, activating endochondral ossification. The early response induced by both scaffolds leads to complete bridging of the critical-sized bone defects within 4-weeks. The versatile cell-free gene-activated scaffold described in this study is capable of harnessing and enhancing the body's own regenerative capacity and has immense potential in a myriad of applications.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- iEd Hub and Department of Anatomy and Neuroscience, College of Medicine and Health, University College Cork, Cork, T12 CY82, Ireland
| | - Arlyng G Gonzalez Vazquez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| | - David P Walsh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
- Translational Research in Nanomedical Devices, School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
| | - Ashang L Laiva
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Michael B Keogh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Tisse Engineering Research Group, Royal College of Surgeons in Ireland - Medical University of Bahrain, Adliya, Bahrain
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 YN77, Ireland
- Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, D02 PN40, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, D02 YN77, Ireland
| |
Collapse
|
3
|
Tenkumo T, Koide R, Ogawa T, Yamaguchi H, Suzuki S, Miyashita M, Nakamura K, Wang H, Yoda N, Sasaki K. A triple growth factor strategy for optimizing bone augmentation in mice. J Biomed Mater Res B Appl Biomater 2024; 112:e35447. [PMID: 38997799 DOI: 10.1002/jbm.b.35447] [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: 12/05/2023] [Revised: 04/07/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024]
Abstract
With dental implant treatment becoming the gold standard, the need for effective bone augmentation prior to implantation has grown. This study aims to evaluate a bone augmentation strategy integrating three key growth factors: bone morphogenetic protein-2 (BMP-2), insulin-like growth factor 1 (IGF-1), and vascular endothelial growth factor (VEGF). Collagen scaffolds incorporating BMP-2, IGF-1, or VEGF were fabricated and categorized into five groups based on their content: scaffold alone; BMP-2 alone (BMP-2); BMP-2 and IGF-1 (BI); BMP-2, IGF-1, and VEGF (BIV); and BMP-2 and IGF-1 with an earlier release of VEGF (BI + V). The prepared scaffolds were surgically implanted into the calvarias of C57BL/6JJcl mice, and hard tissue formation was assessed after 10 and 28 days through histological, tomographic, and biochemical analyses. The combination of BMP-2 and IGF-1 induced a greater volume of hard tissue augmentation compared with that of BMP-2 alone, regardless of VEGF supplementation, and these groups had increased levels of cartilage compared with others. The volume of hard tissue formation was greatest in the BIV group. In contrast, the BI + V group exhibited a hard tissue volume similar to that of the BI group. While VEGF and CD31 levels were highest in the BIV group at 10 days, there was no correlation at the same time point between hard tissue formation and the quantity of M2 macrophages. In conclusion, the simultaneous release of BMP-2, IGF-1, and VEGF proved to be effective in promoting bone augmentation.
Collapse
Affiliation(s)
- Taichi Tenkumo
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Rie Koide
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Toru Ogawa
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Hirofumi Yamaguchi
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Shigeki Suzuki
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Makiko Miyashita
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Keisuke Nakamura
- Department of Advanced Free Radical Science, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Han Wang
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Nobuhiro Yoda
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate school of Dentistry, Sendai, Japan
| | - Keiichi Sasaki
- Tohoku University Graduate School of Dentistry, Sendai, Japan
| |
Collapse
|
4
|
He Z, Li H, Zhang Y, Gao S, Liang K, Su Y, Du Y, Wang D, Xing D, Yang Z, Lin J. Enhanced bone regeneration via endochondral ossification using Exendin-4-modified mesenchymal stem cells. Bioact Mater 2024; 34:98-111. [PMID: 38186959 PMCID: PMC10770633 DOI: 10.1016/j.bioactmat.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
Nonunions and delayed unions pose significant challenges in orthopedic treatment, with current therapies often proving inadequate. Bone tissue engineering (BTE), particularly through endochondral ossification (ECO), emerges as a promising strategy for addressing critical bone defects. This study introduces mesenchymal stem cells overexpressing Exendin-4 (MSC-E4), designed to modulate bone remodeling via their autocrine and paracrine functions. We established a type I collagen (Col-I) sponge-based in vitro model that effectively recapitulates the ECO pathway. MSC-E4 demonstrated superior chondrogenic and hypertrophic differentiation and enhanced the ECO cell fate in single-cell sequencing analysis. Furthermore, MSC-E4 encapsulated in microscaffold, effectively facilitated bone regeneration in a rat calvarial defect model, underscoring its potential as a therapeutic agent for bone regeneration. Our findings advocate for MSC-E4 within a BTE framework as a novel and potent approach for treating significant bone defects, leveraging the intrinsic ECO process.
Collapse
Affiliation(s)
- Zihao He
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Hui Li
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Shuang Gao
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Yiqi Su
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Du Wang
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Zhen Yang
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| | - Jianhao Lin
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Arthritis Institute, Peking University, Beijing, 100044, China
| |
Collapse
|
5
|
He L. Biomaterials for Regenerative Cranioplasty: Current State of Clinical Application and Future Challenges. J Funct Biomater 2024; 15:84. [PMID: 38667541 PMCID: PMC11050949 DOI: 10.3390/jfb15040084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Acquired cranial defects are a prevalent condition in neurosurgery and call for cranioplasty, where the missing or defective cranium is replaced by an implant. Nevertheless, the biomaterials in current clinical applications are hardly exempt from long-term safety and comfort concerns. An appealing solution is regenerative cranioplasty, where biomaterials with/without cells and bioactive molecules are applied to induce the regeneration of the cranium and ultimately repair the cranial defects. This review examines the current state of research, development, and translational application of regenerative cranioplasty biomaterials and discusses the efforts required in future research. The first section briefly introduced the regenerative capacity of the cranium, including the spontaneous bone regeneration bioactivities and the presence of pluripotent skeletal stem cells in the cranial suture. Then, three major types of biomaterials for regenerative cranioplasty, namely the calcium phosphate/titanium (CaP/Ti) composites, mineralised collagen, and 3D-printed polycaprolactone (PCL) composites, are reviewed for their composition, material properties, and findings from clinical trials. The third part discusses perspectives on future research and development of regenerative cranioplasty biomaterials, with a considerable portion based on issues identified in clinical trials. This review aims to facilitate the development of biomaterials that ultimately contribute to a safer and more effective healing of cranial defects.
Collapse
Affiliation(s)
- Lizhe He
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, China
| |
Collapse
|
6
|
Cheng P, Zhao X, Han M, Zhuang Y, Ning F, Hu Y, Lu W, Miao S, Zhao C, Jia L, Hao X, Sun M, Wang J, Chen F, Yang L, Jie Q. Periodic static compression of micro-strain pattern regulates endochondral bone formation. Front Bioeng Biotechnol 2024; 12:1356135. [PMID: 38600948 PMCID: PMC11004279 DOI: 10.3389/fbioe.2024.1356135] [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: 12/15/2023] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction: Developmental engineering based on endochondral ossification has been proposed as a potential strategy for repairing of critical bone defects. Bone development is driven by growth plate-mediated endochondral ossification. Under physiological conditions, growth plate chondrocytes undergo compressive forces characterized by micro-mechanics, but the regulatory effect of micro-mechanical loading on endochondral bone formation has not been investigated. Methods: In this study, a periodic static compression (PSC) model characterized by micro-strain (with 0.5% strain) was designed to clarify the effects of biochemical/mechanical cues on endochondral bone formation. Hydrogel scaffolds loaded with bone marrow mesenchymal stem cells (BMSCs) were incubated in proliferation medium or chondrogenic medium, and PSC was performed continuously for 14 or 28 days. Subsequently, the scaffold pretreated for 28 days was implanted into rat femoral muscle pouches and femoral condylar defect sites. The chondrogenesis and bone defect repair were evaluated 4 or 10 weeks post-operation. Results: The results showed that PSC stimulation for 14 days significantly increased the number of COL II positive cells in proliferation medium. However, the chondrogenic efficiency of BMSCs was significantly improved in chondrogenic medium, with or without PSC application. The induced chondrocytes (ichondrocytes) spontaneously underwent hypertrophy and maturation, but long-term mechanical stimulation (loading for 28 days) significantly inhibited hypertrophy and mineralization in ichondrocytes. In the heterotopic ossification model, no chondrocytes were found and no significant difference in terms of mineral deposition in each group; However, 4 weeks after implantation into the femoral defect site, all scaffolds that were subjected to biochemical/mechanical cues, either solely or synergistically, showed typical chondrocytes and endochondral bone formation. In addition, simultaneous biochemical induction/mechanical loading significantly accelerated the bone regeneration. Discussion: Our findings suggest that microstrain mechanics, biochemical cues, and in vivo microenvironment synergistically regulate the differentiation fate of BMSCs. Meanwhile, this study shows the potential of micro-strain mechanics in the treatment of critical bone defects.
Collapse
Affiliation(s)
- Pengzhen Cheng
- College of Life Sciences, Northwest University, Xi’an, China
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
- Xi’an Key Laboratory of Skeletal Developmental Deformity and Injury Repair, Xi’an, China
| | - Xueyi Zhao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Meige Han
- College of Life Sciences, Northwest University, Xi’an, China
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yaping Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fenru Ning
- Department of Neonatology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yaqian Hu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Weiguang Lu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Sheng Miao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Chengxiang Zhao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Liyuan Jia
- College of Life Sciences, Northwest University, Xi’an, China
| | - Xue Hao
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of Skeletal Developmental Deformity and Injury Repair, Xi’an, China
| | - Meng Sun
- College of Life Sciences, Northwest University, Xi’an, China
| | - Junxiang Wang
- College of Life Sciences, Northwest University, Xi’an, China
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Fulin Chen
- College of Life Sciences, Northwest University, Xi’an, China
- Xi’an Key Laboratory of Skeletal Developmental Deformity and Injury Repair, Xi’an, China
- Research Center for Skeletal Developmental Deformity and Injury Repair, School of Life Science and Medicine, Northwest University, Xi’an, China
| | - Liu Yang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Qiang Jie
- Pediatric Orthopaedic Hospital, Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
- Xi’an Key Laboratory of Skeletal Developmental Deformity and Injury Repair, Xi’an, China
- Research Center for Skeletal Developmental Deformity and Injury Repair, School of Life Science and Medicine, Northwest University, Xi’an, China
| |
Collapse
|
7
|
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
|
8
|
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: 6] [Impact Index Per Article: 6.0] [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
|
9
|
Hyaluronic acid hydrogels support to generate integrated bone formation through endochondral ossification in vivo using mesenchymal stem cells. PLoS One 2023; 18:e0281345. [PMID: 36730328 PMCID: PMC9894498 DOI: 10.1371/journal.pone.0281345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
Engineered cartilage tissue from differentiated mesenchymal stem cells (MSCs) can generate bone in vivo through endochondral ossification (ECO). This ECO-mediated approach has the potential to circumvent the severe problems associated with conventional MSC-based bone tissue engineering techniques that lack mechanisms to induce angiogenesis. Hyaluronic acid (HA) is a key component in the cartilage extracellular matrix. However, the ECO-supporting properties of HA remain largely unclear. This study aimed to compare the ability of HA and collagen hydrogels to support in vitro differentiation of MSC-based hypertrophic cartilage tissues and to promote endochondral bone formation in vivo. Following the chondrogenic and hypertrophic differentiation in vitro, both HA and collagen constructs accumulated sulfated glycosaminoglycan (sGAG) and type 1, type II, and type X collagen. However, HA hydrogels exhibited a more uniform distribution of sGAG, type 1 collagen, type X collagen, and osteocalcin proteins; in addition, the cells embedded in the hydrogels had more rounded cell morphologies than those in the collagen constructs. At week 5 of in vitro culture, two to three constructs were implanted into a subcutaneous pocket in nude mice and harvested after 4 and 8 weeks. Both HA and collagen constructs promoted endochondral bone formation with vascularization and bone marrow development; however, the HA constructs fused to form integrated bone tissues and the bone marrow developed along the space between the two adhered grafts in all implanted pockets (n = 5). In the collagen constructs, the integration was observed in 40% of the pockets (n = 5). Microcomputer CT analysis revealed that the bone volume of HA constructs was larger than that of collagen constructs. In conclusion, compared to collagen hydrogels, HA hydrogels had superior potential to generate integrated bone with vascularization and bone marrow development. This study provides valuable insights for applying ECO-mediated bone tissue engineering approaches for the repair of critical-sized bone defects.
Collapse
|
10
|
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: 19.0] [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
|
11
|
Wang H, Tenkumo T, Nemoto E, Kanda Y, Ogawa T, Sasaki K. Introduction of tenomodulin by gene transfection vectors for rat bone tissue regeneration. Regen Ther 2023; 22:99-108. [PMID: 36712960 PMCID: PMC9842804 DOI: 10.1016/j.reth.2022.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/17/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Periodontal ligament is regenerated in association with hard tissue regeneration. Tenomodulin (Tnmd) expression has been confirmed in periodontal ligament and it reportedly inhibits angiogenesis or is involved in collagen fibril maturation. The introduction of Tnmd by gene transfection in bone tissue regeneration therapy might inhibit topical hard tissue formation and induce the formation of dense fibrous tissue. Therefore, the effect of Tnmd introduction by gene transfection technique in vitro and in vivo was investigated in this study. Methods Osteogenesis- and chondrogenesis-related gene expression levels in osteoblastic cells (MC3T3E1) and rat bone marrow derived cells were detected using qPCR three days after gene transfection with plasmid DNA (Tnmd) using non-viral gene transfection vectors: a calcium phosphate-based gene transfection vector (CaP(Tnmd)) or a cationic polymer-based reagent (JetPEI (Tnmd)). Next, an atelocollagen scaffold with or without CaP (Tnmd) or JetPEI (Tnmd) was implanted into a rat calvaria bone defect, and the remaining bone defect volume and the tissue reaction at 28 days after surgery were evaluated. Results Runx 2 and SP7 mRNA was reduced by JetPEI (Tnmd) in both cells, but not in CaP(Tnmd). The volume of expressed Tnmd was at 9 ng/mL in both gene transfection vector. The remaining bone defect volume of JetPEI (Tnmd) was significantly bigger than that of the other groups and CaP (EGFP), and that of CaP (Tnmd) was significantly bigger than that of CaP (EGFP). Conclusions Tnmd introduction treatment inhibits bone formation in artificial bone defect, however, the effect of that was dependent on non-viral gene transfection vector.
Collapse
Affiliation(s)
- Han Wang
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Taichi Tenkumo
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- Corresponding author. Fax.: (+81)(022)717-8371.
| | - Eiji Nemoto
- Division of Periodontology, Department of Oral Biology, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Yoshiaki Kanda
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Toru Ogawa
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Keiichi Sasaki
- Division of Advanced Prosthetic Dentistry, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|
12
|
Ranakoti L, Gangil B, Bhandari P, Singh T, Sharma S, Singh J, Singh S. Promising Role of Polylactic Acid as an Ingenious Biomaterial in Scaffolds, Drug Delivery, Tissue Engineering, and Medical Implants: Research Developments, and Prospective Applications. Molecules 2023; 28:485. [PMID: 36677545 PMCID: PMC9861437 DOI: 10.3390/molecules28020485] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023] Open
Abstract
In the present scenario, the research is now being focused on the naturally occurring polymers that can gradually replace the existing synthetic polymers for the development of bio composites having applications in medical surgeries and human implants. With promising mechanical properties and bio compatibility with human tissues, poly lactic acid (PLA) is now being viewed as a future bio material. In order to examine the applicability of PLA in human implants, the current article sheds light on the synthesis of PLA and its various copolymers used to alter its physical and mechanical properties. In the latter half, various processes used for the fabrication of biomaterials are discussed in detail. Finally, biomaterials that are currently in use in the field of biomedical (Scaffolding, drug delivery, tissue engineering, medical implants, derma, cosmetics, medical surgeries, and human implants) are represented with respective advantages in the sphere of biomaterials.
Collapse
Affiliation(s)
- Lalit Ranakoti
- Department of Mechanical Engineering, Graphic Era Deemed to be University, Dehradun 248002, Uttarakhand, India
| | - Brijesh Gangil
- Mechanical Engineering Department, SOET, HNB Garhwal University, Srinagar 246174, Uttarakhand, India
| | - Prabhakar Bhandari
- Mechanical Engineering Department, SOET, K. R. Mangalam University, Gurgaon 122103, Haryana, India
| | - Tej Singh
- Savaria Institute of Technology, Eötvös Loránd University, 9700 Szombathely, Hungary
| | - Shubham Sharma
- Mechanical Engineering Department, University Center for Research and Development, Chandigarh University, Mohali 140413, Punjab, India
- School of Mechanical and Automotive Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Jujhar Singh
- Department of Mechanical Engineering, IK Gujral Punjab Technical University, Kapurthala 144603, Punjab, India
| | - Sunpreet Singh
- Department of Mechanical Engineering, National University of Singapore, Singapore 117575, Singapore
| |
Collapse
|
13
|
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: 8] [Impact Index Per Article: 4.0] [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
|
14
|
Bernhard JC, Marolt Presen D, Li M, Monforte X, Ferguson J, Leinfellner G, Heimel P, Betti SL, Shu S, Teuschl-Woller AH, Tangl S, Redl H, Vunjak-Novakovic G. Effects of Endochondral and Intramembranous Ossification Pathways on Bone Tissue Formation and Vascularization in Human Tissue-Engineered Grafts. Cells 2022; 11:cells11193070. [PMID: 36231032 PMCID: PMC9564153 DOI: 10.3390/cells11193070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/24/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
Bone grafts can be engineered by differentiating human mesenchymal stromal cells (MSCs) via the endochondral and intramembranous ossification pathways. We evaluated the effects of each pathway on the properties of engineered bone grafts and their capacity to drive bone regeneration. Bone-marrow-derived MSCs were differentiated on silk scaffolds into either hypertrophic chondrocytes (hyper) or osteoblasts (osteo) over 5 weeks of in vitro cultivation, and were implanted subcutaneously for 12 weeks. The pathways' constructs were evaluated over time with respect to gene expression, composition, histomorphology, microstructure, vascularization and biomechanics. Hypertrophic chondrocytes expressed higher levels of osteogenic genes and deposited significantly more bone mineral and proteins than the osteoblasts. Before implantation, the mineral in the hyper group was less mature than that in the osteo group. Following 12 weeks of implantation, the hyper group had increased mineral density but a similar overall mineral composition compared with the osteo group. The hyper group also displayed significantly more blood vessel infiltration than the osteo group. Both groups contained M2 macrophages, indicating bone regeneration. These data suggest that, similar to the body's repair processes, endochondral pathway might be more advantageous when regenerating large defects, whereas intramembranous ossification could be utilized to guide the tissue formation pattern with a scaffold architecture.
Collapse
Affiliation(s)
- Jonathan C. Bernhard
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Darja Marolt Presen
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Ming Li
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xavier Monforte
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria
| | - James Ferguson
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Gabriele Leinfellner
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Patrick Heimel
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- School of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Susanna L. Betti
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sharon Shu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Andreas H. Teuschl-Woller
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Department of Life Science Engineering, University of Applied Sciences Technikum Wien, 1200 Vienna, Austria
| | - Stefan Tangl
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- School of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Correspondence: (H.R.); (G.V.-N.); Tel.: +43-(0)-59393-41961 (H.R.); +1-212-305-2304 (G.V.-N.); Fax: +43-(0)-59393-41982 (H.R.); +1-212-305-4692 (G.V.-N.)
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
- Department of Medicine, Columbia University, New York, NY 10032, USA
- College of Dental Medicine, Columbia University, New York, NY 10032, USA
- Correspondence: (H.R.); (G.V.-N.); Tel.: +43-(0)-59393-41961 (H.R.); +1-212-305-2304 (G.V.-N.); Fax: +43-(0)-59393-41982 (H.R.); +1-212-305-4692 (G.V.-N.)
| |
Collapse
|
15
|
Huang R, Fu R, Yan Y, Liu C, Yang J, Xie Y, Li Q. Engineering hypertrophic cartilage grafts from lipoaspirate for critical-sized calvarial bone defect reconstruction: An adipose tissue-based developmental engineering approach. Bioeng Transl Med 2022; 7:e10312. [PMID: 36176620 PMCID: PMC9472001 DOI: 10.1002/btm2.10312] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/17/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
Developmental engineering of living implants from different cell sources capable of stimulating bone regeneration by recapitulating endochondral ossification (ECO) is a promising strategy for large bone defect reconstruction. However, the clinical translation of these cell-based approaches is hampered by complex manufacturing procedures, poor cell differentiation potential, and limited predictive in vivo performance. We developed an adipose tissue-based developmental engineering approach to overcome these hurdles using hypertrophic cartilaginous (HyC) constructs engineered from lipoaspirate to repair large bone defects. The engineered HyC constructs were implanted into 4-mm calvarial defects in nude rats and compared with decellularized bone matrix (DBM) grafts. The DBM grafts induced neo-bone formation via the recruitment of host cells, while the HyC pellets supported bone regeneration via ECO, as evidenced by the presence of remaining cartilage analog and human NuMA-positive cells within the newly formed bone. However, the HyC pellets clearly showed superior regenerative capacity compared with that of the DBM grafts, yielding more new bone formation, higher blood vessel density, and better integration with adjacent native bone. We speculate that this effect arises from vascular endothelial growth factor and bone morphogenetic protein-2 secretion and mineral deposition in the HyC pellets before implantation, promoting increased vascularization and bone formation upon implantation. The results of this study demonstrate that adipose-derived HyC constructs can effectively heal large bone defects and present a translatable therapeutic option for bone defect repair.
Collapse
Affiliation(s)
- Ru‐Lin Huang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Rao Fu
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuxin Yan
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chuanqi Liu
- Department of Plastic and Burn SurgeryWest China Hospital, Sichuan UniversityChengduChina
| | - Jing Yang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yun Xie
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qingfeng Li
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
16
|
Ze Y, Li Y, Huang L, Shi Y, Li P, Gong P, Lin J, Yao Y. Biodegradable Inks in Indirect Three-Dimensional Bioprinting for Tissue Vascularization. Front Bioeng Biotechnol 2022; 10:856398. [PMID: 35402417 PMCID: PMC8990266 DOI: 10.3389/fbioe.2022.856398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/09/2022] [Indexed: 02/05/2023] Open
Abstract
Mature vasculature is important for the survival of bioengineered tissue constructs, both in vivo and in vitro; however, the fabrication of fully vascularized tissue constructs remains a great challenge in tissue engineering. Indirect three-dimensional (3D) bioprinting refers to a 3D printing technique that can rapidly fabricate scaffolds with controllable internal pores, cavities, and channels through the use of sacrificial molds. It has attracted much attention in recent years owing to its ability to create complex vascular network-like channels through thick tissue constructs while maintaining endothelial cell activity. Biodegradable materials play a crucial role in tissue engineering. Scaffolds made of biodegradable materials act as temporary templates, interact with cells, integrate with native tissues, and affect the results of tissue remodeling. Biodegradable ink selection, especially the choice of scaffold and sacrificial materials in indirect 3D bioprinting, has been the focus of several recent studies. The major objective of this review is to summarize the basic characteristics of biodegradable materials commonly used in indirect 3D bioprinting for vascularization, and to address recent advances in applying this technique to the vascularization of different tissues. Furthermore, the review describes how indirect 3D bioprinting creates blood vessels and vascularized tissue constructs by introducing the methodology and biodegradable ink selection. With the continuous improvement of biodegradable materials in the future, indirect 3D bioprinting will make further contributions to the development of this field.
Collapse
Affiliation(s)
- Yiting Ze
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxi Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyang Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixin Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
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
|
18
|
Baek I, Bello AB, Jeon J, Arai Y, Cha BH, Kim BJ, Lee SH. Therapeutic potential of epiphyseal growth plate cells for bone regeneration in an osteoporosis model. J Tissue Eng 2022; 13:20417314221116754. [PMID: 35983547 PMCID: PMC9379561 DOI: 10.1177/20417314221116754] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/14/2022] [Indexed: 12/04/2022] Open
Abstract
Bone growth occurs in the epiphyseal growth plate (EGP) and epiphyseal growth plate cells (EGPCs) exist in EGP. EGPCs, including skeletal stem cells (SSCs), are cells that induce bone growth and development through endochondral ossification. Recently, the superiority of bone regeneration through endochondral ossification has been reported. Our study compared EGPCs with bone marrow-derived mesenchymal stem cells (BM-MSCs) and suggested the therapeutic potential of new bone regeneration. In this study, we analyzed the characteristics between EGPCs and BM-MSCs based on morphological characteristics and molecular profiles. EGPCs expressed chondrogenic and osteogenic markers higher than BM-MSCs. Additionally, in co-culture with BM-MSCs, EGPCs induced an increase in chondrogenic, osteogenic, and hypertrophic markers of BM-MSCs. Finally, EGPCs induced higher bone regeneration than BM-MSCs in the osteoporosis model. Overall, we suggest the possibility of EGPCs as cell therapy for effective bone regeneration.
Collapse
Affiliation(s)
- Inho Baek
- Department of Medical Biotechnology, Dongguk University, Goyang, Gyeonggi, Republic of Korea
| | - Alvin Bacero Bello
- Department of Medical Biotechnology, Dongguk University, Goyang, Gyeonggi, Republic of Korea
| | - Jieun Jeon
- Department of Medical Biotechnology, Dongguk University, Goyang, Gyeonggi, Republic of Korea
| | - Yoshie Arai
- Department of Medical Biotechnology, Dongguk University, Goyang, Gyeonggi, Republic of Korea
| | - Byung-Hyun Cha
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | | | - Soo-Hong Lee
- Department of Medical Biotechnology, Dongguk University, Goyang, Gyeonggi, Republic of Korea
| |
Collapse
|
19
|
Cao C, Huang P, Prasopthum A, Parsons AJ, Ai F, Yang J. Characterisation of bone regeneration in 3D printed ductile PCL/PEG/hydroxyapatite scaffolds with high ceramic microparticle concentrations. Biomater Sci 2021; 10:138-152. [PMID: 34806738 DOI: 10.1039/d1bm01645h] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
3D printed bioactive glass or bioceramic particle reinforced composite scaffolds for bone tissue engineering currently suffer from low particle concentration (<50 wt%) hence low osteoconductivity. Meanwhile, composites with very high inorganic particle concentrations are very brittle. Scaffolds combining high particle content and ductility are urgently required for bone tissue engineering. Herein, 3D printed PCL/hydroxyapatite (HA) scaffolds with high ceramic concentration (up to 90 wt%) are made ductile (>100% breaking strain) by adding poly(ethylene glycol) which is biocompatible and FDA approved. The scaffolds require no post-printing washing to remove hazardous components. More exposure of HA microparticles on strut surfaces is enabled by incorporating higher HA concentrations. Compared to scaffolds with 72 wt% HA, scaffolds with higher HA content (90 wt%) enhance matrix formation but not new bone volume after 12 weeks implantation in rat calvarial defects. Histological analyses demonstrate that bone regeneration within the 3D printed scaffolds is via intramembranous ossification and starts in the central region of pores. Fibrous tissue that resembles non-union tissue within bone fractures is formed within pores that do not have new bone. The amount of blood vessels is similar between scaffolds with mainly fibrous tissue and those with more bone tissue, suggesting vascularization is not a deciding factor for determining the type of tissues regenerated within the pores of 3D printed scaffolds. Multinucleated immune cells are commonly present in all scaffolds surrounding the struts, suggesting a role of managing inflammation in bone regeneration within 3D printed scaffolds.
Collapse
Affiliation(s)
- Chuanliang Cao
- School of Mechatronic Engineering, Nanchang University, Nanchang, Jiangxi, China 330031.
| | - Pengren Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China 330006
| | - Aruna Prasopthum
- Biodiscovery Institute, University of Nottingham, Nottingham, UK NG7 2RD.
| | - Andrew J Parsons
- Composites Research Group, Faculty of Engineering, University of Nottingham, Nottingham, UK NG7 2RD
| | - Fanrong Ai
- School of Mechatronic Engineering, Nanchang University, Nanchang, Jiangxi, China 330031.
| | - Jing Yang
- Biodiscovery Institute, University of Nottingham, Nottingham, UK NG7 2RD.
| |
Collapse
|
20
|
Dalisson B, Charbonnier B, Aoude A, Gilardino M, Harvey E, Makhoul N, Barralet J. Skeletal regeneration for segmental bone loss: Vascularised grafts, analogues and surrogates. Acta Biomater 2021; 136:37-55. [PMID: 34626818 DOI: 10.1016/j.actbio.2021.09.053] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 02/08/2023]
Abstract
Massive segmental bone defects (SBD) are mostly treated by removing the fibula and transplanting it complete with blood supply. While revolutionary 50 years ago, this remains the standard treatment. This review considers different strategies to repair SBD and emerging potential replacements for this highly invasive procedure. Prior to the technical breakthrough of microsurgery, researchers in the 1960s and 1970s had begun to make considerable progress in developing non autologous routes to repairing SBD. While the breaktthrough of vascularised bone transplantation solved the immediate problem of a lack of reliable repair strategies, much of their prior work is still relevant today. We challenge the assumption that mimicry is necessary or likely to be successful and instead point to the utility of quite crude (from a materials technology perspective), approaches. Together there are quite compelling indications that the body can regenerate entire bone segments with few or no exogenous factors. This is important, as there is a limit to how expensive a bone repair can be and still be widely available to all patients since cost restraints within healthcare systems are not likely to diminish in the near future. STATEMENT OF SIGNIFICANCE: This review is significant because it is a multidisciplinary view of several surgeons and scientists as to what is driving improvement in segmental bone defect repair, why many approaches to date have not succeeded and why some quite basic approaches can be as effective as they are. While there are many reviews of the literature of grafting and bone repair the relative lack of substantial improvement and slow rate of progress in clinical translation is often overlooked and we seek to challenge the reader to consider the issue more broadly.
Collapse
|
21
|
Andrés Sastre E, Nossin Y, Jansen I, Kops N, Intini C, Witte-Bouma J, van Rietbergen B, Hofmann S, Ridwan Y, Gleeson JP, O'Brien FJ, Wolvius EB, van Osch GJVM, Farrell E. A new semi-orthotopic bone defect model for cell and biomaterial testing in regenerative medicine. Biomaterials 2021; 279:121187. [PMID: 34678648 DOI: 10.1016/j.biomaterials.2021.121187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
In recent decades, an increasing number of tissue engineered bone grafts have been developed. However, expensive and laborious screenings in vivo are necessary to assess the safety and efficacy of their formulations. Rodents are the first choice for initial in vivo screens but their size limits the dimensions and number of the bone grafts that can be tested in orthotopic locations. Here, we report the development of a refined murine subcutaneous model for semi-orthotopic bone formation that allows the testing of up to four grafts per mouse one order of magnitude greater in volume than currently possible in mice. Crucially, these defects are also "critical size" and unable to heal within the timeframe of the study without intervention. The model is based on four bovine bone implants, ring-shaped, where the bone healing potential of distinct grafts can be evaluated in vivo. In this study we demonstrate that promotion and prevention of ossification can be assessed in our model. For this, we used a semi-automatic algorithm for longitudinal micro-CT image registration followed by histological analyses. Taken together, our data supports that this model is suitable as a platform for the real-time screening of bone formation, and provides the possibility to study bone resorption, osseointegration and vascularisation.
Collapse
Affiliation(s)
- E Andrés Sastre
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Y Nossin
- Department of Otorhinolaryngology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - I Jansen
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands; Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - N Kops
- Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C Intini
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - J Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - B van Rietbergen
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Department of Orthopaedic Surgery, Research School CAPHRI, Maastricht University Medical Center, Maastricht, the Netherlands
| | - S Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Y Ridwan
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J P Gleeson
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - F J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; SFI Advanced Materials and Bioengineering Research (AMBER) Center, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland; Trinity Center for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - E B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - G J V M van Osch
- Department of Otorhinolaryngology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Orthopaedics and Sports Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Delft, the Netherlands
| | - E Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
22
|
Fahy N, Palomares Cabeza V, Lolli A, Witte-Bouma J, Merino A, Ridwan Y, Wolvius EB, Hoogduijn MJ, Farrell E, Brama PAJ. Chondrogenically Primed Human Mesenchymal Stem Cells Persist and Undergo Early Stages of Endochondral Ossification in an Immunocompetent Xenogeneic Model. Front Immunol 2021; 12:715267. [PMID: 34659205 PMCID: PMC8515138 DOI: 10.3389/fimmu.2021.715267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
Tissue engineering approaches using progenitor cells such as mesenchymal stromal cells (MSCs) represent a promising strategy to regenerate bone. Previous work has demonstrated the potential of chondrogenically primed human MSCs to recapitulate the process of endochondral ossification and form mature bone in vivo, using immunodeficient xenogeneic models. To further the translation of such MSC-based approaches, additional investigation is required to understand the impact of interactions between human MSC constructs and host immune cells upon the success of MSC-mediated bone formation. Although human MSCs are considered hypoimmunogenic, the potential of chondrogenically primed human MSCs to induce immunogenic responses in vivo, as well as the efficacy of MSC-mediated ectopic bone formation in the presence of fully competent immune system, requires further elucidation. Therefore, the aim of this study was to investigate the capacity of chondrogenically primed human MSC constructs to persist and undergo the process of endochondral ossification in an immune competent xenogeneic model. Chondrogenically differentiated human MSC pellets were subcutaneously implanted to wild-type BALB/c mice and retrieved at 2 and 12 weeks post-implantation. The percentages of CD4+ and CD8+ T cells, B cells, and classical/non-classical monocyte subsets were not altered in the peripheral blood of mice that received chondrogenic MSC constructs compared to sham-operated controls at 2 weeks post-surgery. However, MSC-implanted mice had significantly higher levels of serum total IgG compared to sham-operated mice at this timepoint. Flow cytometric analysis of retrieved MSC constructs identified the presence of T cells and macrophages at 2 and 12 weeks post-implantation, with low levels of immune cell infiltration to implanted MSC constructs detected by CD45 and CD3 immunohistochemical staining. Despite the presence of immune cells in the tissue, MSC constructs persisted in vivo and were not degraded/resorbed. Furthermore, constructs became mineralised, with longitudinal micro-computed tomography imaging revealing an increase in mineralised tissue volume from 4 weeks post-implantation until the experimental endpoint at 12 weeks. These findings indicate that chondrogenically differentiated human MSC pellets can persist and undergo early stages of endochondral ossification following subcutaneous implantation in an immunocompetent xenogeneic model. This scaffold-free model may be further extrapolated to provide mechanistic insight to osteoimmunological processes regulating bone regeneration and homeostasis.
Collapse
Affiliation(s)
- Niamh Fahy
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Orthopaedics and Sports Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands.,Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ana Merino
- Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Yanto Ridwan
- Department of Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
23
|
Tam WL, Freitas Mendes L, Chen X, Lesage R, Van Hoven I, Leysen E, Kerckhofs G, Bosmans K, Chai YC, Yamashita A, Tsumaki N, Geris L, Roberts SJ, Luyten FP. Human pluripotent stem cell-derived cartilaginous organoids promote scaffold-free healing of critical size long bone defects. Stem Cell Res Ther 2021; 12:513. [PMID: 34563248 PMCID: PMC8466996 DOI: 10.1186/s13287-021-02580-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 08/20/2021] [Indexed: 12/16/2022] Open
Abstract
Background Bones have a remarkable capacity to heal upon fracture. Yet, in large defects or compromised conditions healing processes become impaired, resulting in delayed or non-union. Current therapeutic approaches often utilize autologous or allogeneic bone grafts for bone augmentation. However, limited availability of these tissues and lack of predictive biological response result in limitations for clinical demands. Tissue engineering using viable cell-based implants is a strategic approach to address these unmet medical needs. Methods Herein, the in vitro and in vivo cartilage and bone tissue formation potencies of human pluripotent stem cells were investigated. The induced pluripotent stem cells were specified towards the mesodermal lineage and differentiated towards chondrocytes, which subsequently self-assembled into cartilaginous organoids. The tissue formation capacity of these organoids was then challenged in an ectopic and orthotopic bone formation model. Results The derived chondrocytes expressed similar levels of collagen type II as primary human articular chondrocytes and produced stable cartilage when implanted ectopically in vivo. Upon targeted promotion towards hypertrophy and priming with a proinflammatory mediator, the organoids mediated successful bridging of critical size long bone defects in immunocompromised mice. Conclusions These results highlight the promise of induced pluripotent stem cell technology for the creation of functional cartilage tissue intermediates that can be explored for novel bone healing strategies. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02580-7.
Collapse
Affiliation(s)
- Wai Long Tam
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, Onderwijs en Navorsing 8th floor, bus 813, 3000, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium
| | - Luís Freitas Mendes
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium
| | - Xike Chen
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium
| | - Raphaëlle Lesage
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Biomechmanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3000, Leuven, Belgium
| | - Inge Van Hoven
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, Onderwijs en Navorsing 8th floor, bus 813, 3000, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium
| | - Elke Leysen
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, Onderwijs en Navorsing 8th floor, bus 813, 3000, Leuven, Belgium.,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium
| | - Greet Kerckhofs
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Institute of Mechanics, Materials, and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium.,Institute of Experimental and Clinical Research, UCLouvain, Woluwé-Saint-Lambert, Belgium.,Department of Materials Engineering, KU Leuven, Leuven, Belgium
| | - Kathleen Bosmans
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium
| | - Yoke Chin Chai
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium.,Department of Development and Regeneration, Stem Cell Institute, KU Leuven, O&N4, Herestraat 49, 3000, Leuven, Belgium
| | - Akihiro Yamashita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kawahara-cho 53, Kyoto, 606-8507, Japan
| | - Noriyuki Tsumaki
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kawahara-cho 53, Kyoto, 606-8507, Japan
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium.,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium.,Biomechmanics Section, KU Leuven, Celestijnenlaan 300C (2419), 3000, Leuven, Belgium.,GIGA In Silico Medicine, Quartier Hôpital, Avenue de l'Hôpital 11 B34, 4000, Liège, Belgium
| | - Scott J Roberts
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, Onderwijs en Navorsing 8th floor, bus 813, 3000, Leuven, Belgium.,Department of Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Frank P Luyten
- Laboratory for Developmental and Stem Cell Biology (DSB), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, Onderwijs en Navorsing 8th floor, bus 813, 3000, Leuven, Belgium. .,Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, O&N 1 Herestraat 49 Bus 813, 3000, Leuven, Belgium. .,Laboratory for Tissue Engineering (TE), Skeletal Biology and Engineering Research Center (SBE), KU Leuven, O&N1, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
24
|
Kim HD, Hong X, An YH, Park MJ, Kim DG, Greene AK, Padwa BL, Hwang NS, Lin RZ, Melero-Martin JM. A Biphasic Osteovascular Biomimetic Scaffold for Rapid and Self-Sustained Endochondral Ossification. Adv Healthc Mater 2021; 10:e2100070. [PMID: 33882194 PMCID: PMC8273143 DOI: 10.1002/adhm.202100070] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/23/2021] [Indexed: 12/14/2022]
Abstract
Regeneration of large bones remains a challenge in surgery. Recent developmental engineering efforts aim to recapitulate endochondral ossification (EO), a critical step in bone formation. However, this process entails the condensation of mesenchymal stem cells (MSCs) into cartilaginous templates, which requires long-term cultures and is challenging to scale up. Here, a biomimetic scaffold is developed that allows rapid and self-sustained EO without initial hypertrophic chondrogenesis. The design comprises a porous chondroitin sulfate cryogel decorated with whitlockite calcium phosphate nanoparticles, and a soft hydrogel occupying the porous space. This composite scaffold enables human endothelial colony-forming cells (ECFCs) and MSCs to rapidly assemble into osteovascular niches in immunodeficient mice. These niches contain ECFC-lined blood vessels and perivascular MSCs that differentiate into RUNX2+ OSX+ pre-osteoblasts after one week in vivo. Subsequently, multiple ossification centers are formed, leading to de novo bone tissue formation by eight weeks, including mature human OCN+ OPN+ osteoblasts, collagen-rich mineralized extracellular matrix, hydroxyapatite, osteoclast activity, and gradual mechanical competence. The early establishment of blood vessels is essential, and grafts that do not contain ECFCs fail to produce osteovascular niches and ossification centers. The findings suggest a novel bioengineering approach to recapitulate EO in the context of human bone regeneration.
Collapse
Affiliation(s)
- Hwan D. Kim
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea (H.D.K current address)
| | - Xuechong Hong
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Young-Hyeon An
- School of Chemical and Biological Engineering, BioMAX Institute, Institute of Chemical Processes, Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Mihn Jeong Park
- School of Chemical and Biological Engineering, BioMAX Institute, Institute of Chemical Processes, Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Do-Gyoon Kim
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH 43210, USA
| | - Arin K. Greene
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Bonnie L. Padwa
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Department of Plastic and Oral Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Nathaniel S. Hwang
- School of Chemical and Biological Engineering, BioMAX Institute, Institute of Chemical Processes, Institute of Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Juan M. Melero-Martin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
25
|
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: 9] [Impact Index Per Article: 3.0] [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
|
26
|
Schott NG, Friend NE, Stegemann JP. Coupling Osteogenesis and Vasculogenesis in Engineered Orthopedic Tissues. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:199-214. [PMID: 32854589 PMCID: PMC8349721 DOI: 10.1089/ten.teb.2020.0132] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Inadequate vascularization of engineered tissue constructs is a main challenge in developing a clinically impactful therapy for large, complex, and recalcitrant bone defects. It is well established that bone and blood vessels form concomitantly during development, as well as during repair after injury. Endothelial cells (ECs) and mesenchymal stromal cells (MSCs) are known to be key players in orthopedic tissue regeneration and vascularization, and these cell types have been used widely in tissue engineering strategies to create vascularized bone. Coculture studies have demonstrated that there is crosstalk between ECs and MSCs that can lead to synergistic effects on tissue regeneration. At the same time, the complexity in fabricating, culturing, and characterizing engineered tissue constructs containing multiple cell types presents a challenge in creating multifunctional tissues. In particular, the timing, spatial distribution, and cell phenotypes that are most conducive to promoting concurrent bone and vessel formation are not well understood. This review describes the processes of bone and vascular development, and how these have been harnessed in tissue engineering strategies to create vascularized bone. There is an emphasis on interactions between ECs and MSCs, and the culture systems that can be used to understand and control these interactions within a single engineered construct. Developmental engineering strategies to mimic endochondral ossification are discussed as a means of generating vascularized orthopedic tissues. The field of tissue engineering has made impressive progress in creating tissue replacements. However, the development of larger, more complex, and multifunctional engineered orthopedic tissues will require a better understanding of how osteogenesis and vasculogenesis are coupled in tissue regeneration. Impact statement Vascularization of large engineered tissue volumes remains a challenge in developing new and more biologically functional bone grafts. A better understanding of how blood vessels develop during bone formation and regeneration is needed. This knowledge can then be applied to develop new strategies for promoting both osteogenesis and vasculogenesis during the creation of engineered orthopedic tissues. This article summarizes the processes of bone and blood vessel development, with a focus on how endothelial cells and mesenchymal stromal cells interact to form vascularized bone both during development and growth, as well as tissue healing. It is meant as a resource for tissue engineers who are interested in creating vascularized tissue, and in particular to those developing cell-based therapies for large, complex, and recalcitrant bone defects.
Collapse
Affiliation(s)
- Nicholas G. Schott
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
27
|
Brennan MÁ, Barilani M, Rusconi F, de Lima J, Vidal L, Lavazza C, Lazzari L, Giordano R, Layrolle P. Chondrogenic and BMP-4 primings confer osteogenesis potential to human cord blood mesenchymal stromal cells delivered with biphasic calcium phosphate ceramics. Sci Rep 2021; 11:6751. [PMID: 33762629 PMCID: PMC7991626 DOI: 10.1038/s41598-021-86147-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/24/2021] [Indexed: 12/15/2022] Open
Abstract
Bone marrow mesenchymal stem/stromal cells (BMSCs) show great promise for bone repair, however they are isolated by an invasive bone marrow harvest and their regenerative potential decreases with age. Conversely, cord blood can be collected non-invasively after birth and contains MSCs (CBMSCs) that can be stored for future use. However, whether CBMSCs can replace BMSCs targeting bone repair is unknown. This study evaluates the in vitro osteogenic potential of unprimed, osteogenically primed, or chondrogenically primed CBMSCs and BMSCs and their in vivo bone forming capacity following ectopic implantation on biphasic calcium phosphate ceramics in nude mice. In vitro, alkaline phosphatase (intracellular, extracellular, and gene expression), and secretion of osteogenic cytokines (osteoprotegerin and osteocalcin) was significantly higher in BMSCs compared with CBMSCs, while CBMSCs demonstrated superior chondrogenic differentiation and secretion of interleukins IL-6 and IL-8. BMSCs yielded significantly more cell engraftment and ectopic bone formation compared to CBMSCs. However, priming of CBMSCs with either chondrogenic or BMP-4 supplements led to bone formation by CBMSCs. This study is the first direct quantification of the bone forming abilities of BMSCs and CBMSCs in vivo and, while revealing the innate superiority of BMSCs for bone repair, it provides avenues to induce osteogenesis by CBMSCs.
Collapse
Affiliation(s)
- Meadhbh Á Brennan
- Inserm, UMR 1238, PHY-OS Laboratory, Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France
- National University of Ireland (NUIG), Galway, Ireland
| | - Mario Barilani
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Rusconi
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Julien de Lima
- Inserm, UMR 1238, PHY-OS Laboratory, Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Luciano Vidal
- Inserm, UMR 1238, PHY-OS Laboratory, Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France
- Rapid Manufacturing Platform, GEM Laboratory, Centrale Nantes, Nantes, France
| | - Cristiana Lavazza
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Rosaria Giordano
- Laboratory of Regenerative Medicine-Cell Factory, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Pierre Layrolle
- Inserm, UMR 1238, PHY-OS Laboratory, Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France.
| |
Collapse
|
28
|
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: 8] [Impact Index Per Article: 2.7] [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
|
29
|
Fu R, Liu C, Yan Y, Li Q, Huang RL. Bone defect reconstruction via endochondral ossification: A developmental engineering strategy. J Tissue Eng 2021; 12:20417314211004211. [PMID: 33868628 PMCID: PMC8020769 DOI: 10.1177/20417314211004211] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 02/05/2023] Open
Abstract
Traditional bone tissue engineering (BTE) strategies induce direct bone-like matrix formation by mimicking the embryological process of intramembranous ossification. However, the clinical translation of these clinical strategies for bone repair is hampered by limited vascularization and poor bone regeneration after implantation in vivo. An alternative strategy for overcoming these drawbacks is engineering cartilaginous constructs by recapitulating the embryonic processes of endochondral ossification (ECO); these constructs have shown a unique ability to survive under hypoxic conditions as well as induce neovascularization and ossification. Such developmentally engineered constructs can act as transient biomimetic templates to facilitate bone regeneration in critical-sized defects. This review introduces the concept and mechanism of developmental BTE, explores the routes of endochondral bone graft engineering, highlights the current state of the art in large bone defect reconstruction via ECO-based strategies, and offers perspectives on the challenges and future directions of translating current knowledge from the bench to the bedside.
Collapse
Affiliation(s)
- Rao Fu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Yan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Mahmoud E, Sayed M, El-Kady AM, Elsayed H, Naga S. In vitro and in vivo study of naturally derived alginate/hydroxyapatite bio composite scaffolds. Int J Biol Macromol 2020; 165:1346-1360. [DOI: 10.1016/j.ijbiomac.2020.10.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/19/2020] [Accepted: 10/01/2020] [Indexed: 12/25/2022]
|
31
|
González-Vázquez A, Raftery RM, Günbay S, Chen G, Murray DJ, O'Brien FJ. Accelerating bone healing in vivo by harnessing the age-altered activation of c-Jun N-terminal kinase 3. Biomaterials 2020; 268:120540. [PMID: 33307368 DOI: 10.1016/j.biomaterials.2020.120540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/10/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023]
Abstract
We have recently demonstrated that c-Jun N-terminal kinase 3 (JNK3) is a key modulator of the enhanced osteogenic potential of stem cells derived from children when compared to those derived from adults. In this study, we formulated a JNK3-activator nanoparticle (JNK3*) that recapitulates the immense osteogenic potential of juvenile cells in adult stem cells by facilitating JNK3 activation. Moreover, we aimed to functionalize a collagen-based scaffold by incorporating the JNK3* in order to develop an advanced platform capable of accelerating bone healing by recruitment of host stem cells. Our data, in vitro and in vivo, demonstrated that the immense osteogenic potential of juvenile cells could be recapitulated in adult stem cells by facilitating JNK3 activation. Moreover, our results revealed that the JNK3* functionalized 3D scaffold induced the fastest bone healing and greatest blood vessel infiltration when implanted in critical-size rat calvarial defects in vivo. JNK3*scaffold fastest bone healing in vivo was associated with its capacity to recruit host stem cells to the site of injury and promote angiogenic-osteogenic coupling (e.g. Vegfa, Tie1, Runx2, Alp and Igf2 upregulation). In summary, this study has demonstrated the potential of harnessing knowledge of age-altered stem cell mechanobiology in order to develop a materials-based functionalization approach for the repair of large tissue defects.
Collapse
Affiliation(s)
- Arlyng González-Vázquez
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin 2 D02 YN77, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), RCSI and TCD, Dublin 2 D02 PN40, Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin 2 D02 YN77, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), RCSI and TCD, Dublin 2 D02 PN40, Ireland
| | - Suzan Günbay
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin 2 D02 YN77, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2 D02 PN40, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, RCSI, Dublin 2 D02 YN77, Ireland
| | - Dylan J Murray
- National Paediatric Craniofacial Centre, Children's Health Ireland at Temple Street, Temple Street, Rotunda, Dublin 1 D01 XD99, Ireland
| | - Fergal J O'Brien
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin (TCD), Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), RCSI and TCD, Dublin 2 D02 PN40, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin 2 D02 YN77, Ireland.
| |
Collapse
|
32
|
Mohamed-Ahmed S, Yassin MA, Rashad A, Espedal H, Idris SB, Finne-Wistrand A, Mustafa K, Vindenes H, Fristad I. Comparison of bone regenerative capacity of donor-matched human adipose-derived and bone marrow mesenchymal stem cells. Cell Tissue Res 2020; 383:1061-1075. [PMID: 33242173 PMCID: PMC7960590 DOI: 10.1007/s00441-020-03315-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 09/28/2020] [Indexed: 12/22/2022]
Abstract
Adipose-derived stem cells (ASC) have been used as an alternative to bone marrow mesenchymal stem cells (BMSC) for bone tissue engineering. However, the efficacy of ASC in bone regeneration in comparison with BMSC remains debatable, since inconsistent results have been reported. Comparing ASC with BMSC obtained from different individuals might contribute to this inconsistency in results. Therefore, this study aimed to compare the bone regenerative capacity of donor-matched human ASC and BMSC seeded onto poly(l-lactide-co-ε-caprolactone) scaffolds using calvarial bone defects in nude rats. First, donor-matched ASC and BMSC were seeded onto the co-polymer scaffolds to evaluate their in vitro osteogenic differentiation. Seeded scaffolds and scaffolds without cells (control) were then implanted in calvarial defects in nude rats. The expression of osteogenesis-related genes was examined after 4 weeks. Cellular activity was investigated after 4 and 12 weeks. Bone formation was evaluated radiographically and histologically after 4, 12, and 24 weeks. In vitro, ASC and BMSC demonstrated mineralization. However, BMSC showed higher alkaline phosphatase activity than ASC. In vivo, human osteogenesis–related genes Runx2 and collagen type I were expressed in defects with scaffold/cells. Defects with scaffold/BMSC had higher cellular activity than defects with scaffold/ASC. Moreover, bone formation in defects with scaffold/BMSC was greater than in defects with scaffold/ASC, especially at the early time-point. These results suggest that although ASC have the potential to regenerate bone, the rate of bone regeneration with ASC may be slower than with BMSC. Accordingly, BMSC are more suitable for bone regenerative applications.
Collapse
Affiliation(s)
- Samih Mohamed-Ahmed
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Mohammed A Yassin
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Ahmad Rashad
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Heidi Espedal
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Shaza B Idris
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Hallvard Vindenes
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway.,Department for Plastic, Hand and Reconstructive Surgery, National Fire Damage Center, Bergen, Norway
| | - Inge Fristad
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
33
|
Freeman FE, Brennan MÁ, Browe DC, Renaud A, De Lima J, Kelly DJ, McNamara LM, Layrolle P. A Developmental Engineering-Based Approach to Bone Repair: Endochondral Priming Enhances Vascularization and New Bone Formation in a Critical Size Defect. Front Bioeng Biotechnol 2020; 8:230. [PMID: 32296687 PMCID: PMC7137087 DOI: 10.3389/fbioe.2020.00230] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/05/2020] [Indexed: 12/17/2022] Open
Abstract
There is a distinct clinical need for new therapies that provide an effective treatment for large bone defect repair. Herein we describe a developmental approach, whereby constructs are primed to mimic certain aspects of bone formation that occur during embryogenesis. Specifically, we directly compared the bone healing potential of unprimed, intramembranous, and endochondral primed MSC-laden polycaprolactone (PCL) scaffolds. To generate intramembranous constructs, MSC-seeded PCL scaffolds were exposed to osteogenic growth factors, while endochondral constructs were exposed to chondrogenic growth factors to generate a cartilage template. Eight weeks after implantation into a cranial critical sized defect in mice, there were significantly more vessels present throughout defects treated with endochondral constructs compared to intramembranous constructs. Furthermore, 33 and 50% of the animals treated with the intramembranous and endochondral constructs respectively, had full bone union along the sagittal suture line, with significantly higher levels of bone healing than the unprimed group. Having demonstrated the potential of endochondral priming but recognizing that only 50% of animals completely healed after 8 weeks, we next sought to examine if we could further accelerate the bone healing capacity of the constructs by pre-vascularizing them in vitro prior to implantation. The addition of endothelial cells alone significantly reduced the healing capacity of the constructs. The addition of a co-culture of endothelial cells and MSCs had no benefit to either the vascularization or mineralization potential of the scaffolds. Together, these results demonstrate that endochondral priming alone is enough to induce vascularization and subsequent mineralization in a critical-size defect.
Collapse
Affiliation(s)
- Fiona E Freeman
- 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.,Biomechanics Research Centre (BMEC), Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Meadhbh Á Brennan
- INSERM, UMR 1238, PHY-OS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - David C Browe
- 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
| | - Audrey Renaud
- INSERM, UMR 1238, PHY-OS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - Julien De Lima
- INSERM, UMR 1238, PHY-OS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - 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.,Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Laoise M McNamara
- Biomechanics Research Centre (BMEC), Biomedical Engineering, National University of Ireland Galway, Galway, Ireland
| | - Pierre Layrolle
- INSERM, UMR 1238, PHY-OS, Laboratory of Bone Sarcomas and Remodelling of Calcified Tissues, Faculty of Medicine, University of Nantes, Nantes, France
| |
Collapse
|
34
|
Neves SC, Moroni L, Barrias CC, Granja PL. Leveling Up Hydrogels: Hybrid Systems in Tissue Engineering. Trends Biotechnol 2020; 38:292-315. [DOI: 10.1016/j.tibtech.2019.09.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022]
|
35
|
Kronemberger GS, Matsui RAM, Miranda GDASDCE, Granjeiro JM, Baptista LS. Cartilage and bone tissue engineering using adipose stromal/stem cells spheroids as building blocks. World J Stem Cells 2020; 12:110-122. [PMID: 32184936 PMCID: PMC7062040 DOI: 10.4252/wjsc.v12.i2.110] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/19/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
Scaffold-free techniques in the developmental tissue engineering area are designed to mimic in vivo embryonic processes with the aim of biofabricating, in vitro, tissues with more authentic properties. Cell clusters called spheroids are the basis for scaffold-free tissue engineering. In this review, we explore the use of spheroids from adult mesenchymal stem/stromal cells as a model in the developmental engineering area in order to mimic the developmental stages of cartilage and bone tissues. Spheroids from adult mesenchymal stromal/stem cells lineages recapitulate crucial events in bone and cartilage formation during embryogenesis, and are capable of spontaneously fusing to other spheroids, making them ideal building blocks for bone and cartilage tissue engineering. Here, we discuss data from ours and other labs on the use of adipose stromal/stem cell spheroids in chondrogenesis and osteogenesis in vitro. Overall, recent studies support the notion that spheroids are ideal "building blocks" for tissue engineering by “bottom-up” approaches, which are based on tissue assembly by advanced techniques such as three-dimensional bioprinting. Further studies on the cellular and molecular mechanisms that orchestrate spheroid fusion are now crucial to support continued development of bottom-up tissue engineering approaches such as three-dimensional bioprinting.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
| | - Renata Akemi Morais Matsui
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
| | - Guilherme de Almeida Santos de Castro e Miranda
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Duque de Caxias, RJ 25250-020, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói 25255-030 Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
36
|
Kronemberger GS, Dalmônico GML, Rossi AL, Leite PEC, Saraiva AM, Beatrici A, Silva KR, Granjeiro JM, Baptista LS. Scaffold- and serum-free hypertrophic cartilage tissue engineering as an alternative approach for bone repair. Artif Organs 2020; 44:E288-E299. [PMID: 31950507 DOI: 10.1111/aor.13637] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/25/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
Abstract
Human adipose stem/stromal cell (ASC) spheroids were used as a serum-free in vitro model to recapitulate the molecular events and extracellular matrix organization that orchestrate a hypertrophic cartilage phenotype. Induced-ASC spheroids (ø = 450 µm) showed high cell viability throughout the period of culture. The expression of collagen type X alpha 1 chain (COLXA1) and matrix metallopeptidase 13 (MMP-13) was upregulated at week 2 in induced-ASC spheroids compared with week 5 (P < .001) evaluated by quantitative real-time PCR. In accordance, secreted levels of IL-6 (P < .0001), IL-8 (P < .0001), IL-10 (P < .0001), bFGF (P < .001), VEGF (P < .0001), and RANTES (P < .0001) were the highest at week 2. Strong in situ staining for collagen type X and low staining for TSP-1 was associated with the increase of hypertrophic genes expression at week 2 in induced-ASC spheroids. Collagen type I, osteocalcin, biglycan, and tenascin C were detected at week 5 by in situ staining, in accordance with the highest expression of alkaline phosphatase (ALPL) gene and the presence of calcium deposits as evaluated by Alizarin Red O staining. Induced-ASC spheroids showed a higher force required to compression at week 2 (P < .0001). The human ASC spheroids under serum-free inducer medium and normoxic culture conditions were induced to a hypertrophic cartilage phenotype, opening a new perspective to recapitulate endochondral ossification in vivo.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Duque de Caxias, Brazil.,Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil
| | | | | | - Paulo Emílio Correa Leite
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Antonio M Saraiva
- Laboratory of Macromolecules, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Anderson Beatrici
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Scientific and Technological Metrology Division (Dimci), National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - Karina Ribeiro Silva
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói, Brazil
| | - Leandra Santos Baptista
- Nucleus of Multidisciplinary Research in Biology (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ), Duque de Caxias, Brazil.,Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil.,Post-graduation Program of Translational Biomedicine (Biotrans), Unigranrio, Duque de Caxias, Brazil.,Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Duque de Caxias, Brazil
| |
Collapse
|
37
|
Wang Z, Tang J, Li Y, Wang Y, Guo Y, Tu Q, Chen J, Wang C. AdipoRon promotes diabetic fracture repair through endochondral ossification-based bone repair by enhancing survival and differentiation of chondrocytes. Exp Cell Res 2019; 387:111757. [PMID: 31838062 DOI: 10.1016/j.yexcr.2019.111757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022]
Abstract
Diabetic bone defects may exhibit impaired endochondral ossification (ECO) leading to delayed bone repair. AdipoRon, a receptor agonist of adiponectin polymers, can ameliorate diabetes and related complications, as well as overcome the disadvantages of the unstable structure of artificial adiponectin polymers. Here, the effects of AdipoRon on the survival and differentiation of chondrocytes in a diabetic environment were explored focusing on related mechanisms in gene and protein levels. In vivo, AdipoRon was applied to diet-induced-obesity (DIO) mice, a model of obesity and type 2 diabetes, with femoral fracture. Sequential histological evaluations and micro-CT were examined for further verification. We found that AdipoRon could ameliorate cell viability, apoptosis, and reactive oxygen species (ROS) production and promote mRNA expression of chondrogenic markers and cartilaginous matrix production of ATDC5 cells in high glucose medium via activating ERK1/2 pathway. Additionally, DIO mice with intragastric AdipoRon administration had more neocartilage and accelerated new bone formation. These data suggest that AdipoRon could stimulate bone regeneration via ECO in diabetes.
Collapse
Affiliation(s)
- Zhongyi Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Jinxin Tang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Ying Li
- Department of Stomatology, Jinan Central Hospital Affiliated to Shandong University, Jinan, 250013, China
| | - Yu Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Yanyang Guo
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Qisheng Tu
- Tufts School of Dental Medicine, Sackler School of Graduate Biomedical Sciences, Tufts School of Medicine, Boston, 02111, USA
| | - Jake Chen
- Tufts School of Dental Medicine, Sackler School of Graduate Biomedical Sciences, Tufts School of Medicine, Boston, 02111, USA.
| | - Chen Wang
- Jiangsu Key Laboratory of Oral Diseases, Department of Prosthodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
38
|
Novais A, Lesieur J, Sadoine J, Slimani L, Baroukh B, Saubaméa B, Schmitt A, Vital S, Poliard A, Hélary C, Rochefort GY, Chaussain C, Gorin C. Priming Dental Pulp Stem Cells from Human Exfoliated Deciduous Teeth with Fibroblast Growth Factor-2 Enhances Mineralization Within Tissue-Engineered Constructs Implanted in Craniofacial Bone Defects. Stem Cells Transl Med 2019; 8:844-857. [PMID: 31016898 PMCID: PMC6646701 DOI: 10.1002/sctm.18-0182] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022] Open
Abstract
The craniofacial area is prone to trauma or pathologies often resulting in large bone damages. One potential treatment option is the grafting of a tissue-engineered construct seeded with adult mesenchymal stem cells (MSCs). The dental pulp appears as a relevant source of MSCs, as dental pulp stem cells display strong osteogenic properties and are efficient at bone formation and repair. Fibroblast growth factor-2 (FGF-2) and/or hypoxia primings were shown to boost the angiogenesis potential of dental pulp stem cells from human exfoliated deciduous teeth (SHED). Based on these findings, we hypothesized here that these primings would also improve bone formation in the context of craniofacial bone repair. We found that both hypoxic and FGF-2 primings enhanced SHED proliferation and osteogenic differentiation into plastically compressed collagen hydrogels, with a much stronger effect observed with the FGF-2 priming. After implantation in immunodeficient mice, the tissue-engineered constructs seeded with FGF-2 primed SHED mediated faster intramembranous bone formation into critical size calvarial defects than the other groups (no priming and hypoxia priming). The results of this study highlight the interest of FGF-2 priming in tissue engineering for craniofacial bone repair. Stem Cells Translational Medicine 2019;8:844&857.
Collapse
Affiliation(s)
- Anita Novais
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
- AP‐HP Département d'OdontologieHôpitaux Universitaires PNVS, Charles Foix et Henri MondorIle de FranceFrance
| | - Julie Lesieur
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
| | - Jérémy Sadoine
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
| | - Lotfi Slimani
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
| | - Brigitte Baroukh
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
| | - Bruno Saubaméa
- Cellular and Molecular Imaging FacilityInserm US25, CNRS UMS 3612, Faculté de Pharmacie de Paris, Université Paris Descartes Sorbonne Paris CitéParisFrance
| | - Alain Schmitt
- Cochin Institute, Transmission Electron Microscopy Platform, INSERM U1016, CNRS UMR8104Université Paris Descartes Sorbonne Paris CitéParisFrance
| | - Sibylle Vital
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
- AP‐HP Département d'OdontologieHôpitaux Universitaires PNVS, Charles Foix et Henri MondorIle de FranceFrance
| | - Anne Poliard
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
| | - Christophe Hélary
- Laboratoire de Chimie de la Matière Condensée de ParisSorbonne Universités, CNRS, Collège de FranceParisFrance
| | - Gaël Y. Rochefort
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
| | - Catherine Chaussain
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
- AP‐HP Département d'OdontologieHôpitaux Universitaires PNVS, Charles Foix et Henri MondorIle de FranceFrance
| | - Caroline Gorin
- EA 2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant (PIV)Dental School, Université Paris Descartes Sorbonne Paris CitéMontrougeFrance
- AP‐HP Département d'OdontologieHôpitaux Universitaires PNVS, Charles Foix et Henri MondorIle de FranceFrance
| |
Collapse
|
39
|
Larson BL, Yu SN, Park H, Estes BT, Moutos FT, Bloomquist CJ, Wu PB, Welter JF, Langer R, Guilak F, Freed LE. Chondrogenic, hypertrophic, and osteochondral differentiation of human mesenchymal stem cells on three-dimensionally woven scaffolds. J Tissue Eng Regen Med 2019; 13:1453-1465. [PMID: 31115161 DOI: 10.1002/term.2899] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022]
Abstract
The development of mechanically functional cartilage and bone tissue constructs of clinically relevant size, as well as their integration with native tissues, remains an important challenge for regenerative medicine. The objective of this study was to assess adult human mesenchymal stem cells (MSCs) in large, three-dimensionally woven poly(ε-caprolactone; PCL) scaffolds in proximity to viable bone, both in a nude rat subcutaneous pouch model and under simulated conditions in vitro. In Study I, various scaffold permutations-PCL alone, PCL-bone, "point-of-care" seeded MSC-PCL-bone, and chondrogenically precultured Ch-MSC-PCL-bone constructs-were implanted in a dorsal, ectopic pouch in a nude rat. After 8 weeks, only cells in the Ch-MSC-PCL constructs exhibited both chondrogenic and osteogenic gene expression profiles. Notably, although both tissue profiles were present, constructs that had been chondrogenically precultured prior to implantation showed a loss of glycosaminoglycan (GAG) as well as the presence of mineralization along with the formation of trabecula-like structures. In Study II of the study, the GAG loss and mineralization observed in Study I in vivo were recapitulated in vitro by the presence of either nearby bone or osteogenic culture medium additives but were prevented by a continued presence of chondrogenic medium additives. These data suggest conditions under which adult human stem cells in combination with polymer scaffolds synthesize functional and phenotypically distinct tissues based on the environmental conditions and highlight the potential influence that paracrine factors from adjacent bone may have on MSC fate, once implanted in vivo for chondral or osteochondral repair.
Collapse
Affiliation(s)
- Benjamin L Larson
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Sarah N Yu
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Hyoungshin Park
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | | | | | | | - Patrick B Wu
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Jean F Welter
- Skeletal Research Center and Case Center for Multimodal Evaluation of Engineered Cartilage, Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Robert Langer
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| | - Farshid Guilak
- Cytex Therapeutics, Inc., Durham, NC.,Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO.,Shriners Hospitals for Children-St. Louis, St. Louis, MO.,Center of Regenerative Medicine, Washington University in St. Louis, St. Louis, MO
| | - Lisa E Freed
- Institute for Medical Engineering and Science, David H. Koch Institute for Integrative Cancer Research, and Media Lab, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
40
|
Wagner DR, Karnik S, Gunderson ZJ, Nielsen JJ, Fennimore A, Promer HJ, Lowery JW, Loghmani MT, Low PS, McKinley TO, Kacena MA, Clauss M, Li J. Dysfunctional stem and progenitor cells impair fracture healing with age. World J Stem Cells 2019; 11:281-296. [PMID: 31293713 PMCID: PMC6600851 DOI: 10.4252/wjsc.v11.i6.281] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/26/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Successful fracture healing requires the simultaneous regeneration of both the bone and vasculature; mesenchymal stem cells (MSCs) are directed to replace the bone tissue, while endothelial progenitor cells (EPCs) form the new vasculature that supplies blood to the fracture site. In the elderly, the healing process is slowed, partly due to decreased regenerative function of these stem and progenitor cells. MSCs from older individuals are impaired with regard to cell number, proliferative capacity, ability to migrate, and osteochondrogenic differentiation potential. The proliferation, migration and function of EPCs are also compromised with advanced age. Although the reasons for cellular dysfunction with age are complex and multidimensional, reduced expression of growth factors, accumulation of oxidative damage from reactive oxygen species, and altered signaling of the Sirtuin-1 pathway are contributing factors to aging at the cellular level of both MSCs and EPCs. Because of these geriatric-specific issues, effective treatment for fracture repair may require new therapeutic techniques to restore cellular function. Some suggested directions for potential treatments include cellular therapies, pharmacological agents, treatments targeting age-related molecular mechanisms, and physical therapeutics. Advanced age is the primary risk factor for a fracture, due to the low bone mass and inferior bone quality associated with aging; a better understanding of the dysfunctional behavior of the aging cell will provide a foundation for new treatments to decrease healing time and reduce the development of complications during the extended recovery from fracture healing in the elderly.
Collapse
Affiliation(s)
- Diane R Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Sonali Karnik
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Zachary J Gunderson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jeffery J Nielsen
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, United States
| | - Alanna Fennimore
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Hunter J Promer
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, United States
| | - M Terry Loghmani
- Department of Physical Therapy, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN 47907 United States
| | - Todd O McKinley
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, United States
| | - Matthias Clauss
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Jiliang Li
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, United States
| |
Collapse
|
41
|
Sheehy E, Kelly D, O'Brien F. Biomaterial-based endochondral bone regeneration: a shift from traditional tissue engineering paradigms to developmentally inspired strategies. Mater Today Bio 2019; 3:100009. [PMID: 32159148 PMCID: PMC7061547 DOI: 10.1016/j.mtbio.2019.100009] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 02/06/2023] Open
Abstract
There is an urgent, clinical need for an alternative to the use of autologous grafts for the ever increasing number of bone grafting procedures performed annually. Herein, we describe a developmentally inspired approach to bone tissue engineering, which focuses on leveraging biomaterials as platforms for recapitulating the process of endochondral ossification. To begin, we describe the traditional biomaterial-based approaches to tissue engineering that have been investigated as methods to promote in vivo bone regeneration, including the use of three-dimensional biomimetic scaffolds, the delivery of growth factors and recombinant proteins, and the in vitro engineering of mineralized bone-like tissue. Thereafter, we suggest that some of the hurdles encountered by these traditional tissue engineering approaches may be circumvented by modulating the endochondral route to bone repair and, to that end, we assess various biomaterials that can be used in combination with cells and signaling factors to engineer hypertrophic cartilaginous grafts capable of promoting endochondral bone formation. Finally, we examine the emerging trends in biomaterial-based approaches to endochondral bone regeneration, such as the engineering of anatomically shaped templates for bone and osteochondral tissue engineering, the fabrication of mechanically reinforced constructs using emerging three-dimensional bioprinting techniques, and the generation of gene-activated scaffolds, which may accelerate the field towards its ultimate goal of clinically successful bone organ regeneration.
Collapse
Affiliation(s)
- E.J. Sheehy
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- 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
| | - D.J. Kelly
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- 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
| | - F.J. O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- 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
| |
Collapse
|
42
|
Dual functional approaches for osteogenesis coupled angiogenesis in bone tissue engineering. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109761. [PMID: 31349418 DOI: 10.1016/j.msec.2019.109761] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/11/2019] [Accepted: 05/15/2019] [Indexed: 12/31/2022]
Abstract
Bone fracture healing is a multistep and overlapping process of inflammation, angiogenesis and osteogenesis. It is initiated by inflammation, causing the release of various cytokines and growth factors. It leads to the recruitment of stem cells and formation of vasculature resulting in the functional bone formation. This combined phenomenon is used by bone tissue engineers from past few years to address the problem of vasculature and osteogenic differentiation during bone regeneration. In this review, we have discussed all major studies reporting the dual functioning approach to promote osteogenesis coupled angiogenesis using various scaffolds. These scaffolds are broadly classified into four types based on the nature of their structural and functional components. The functionality of the scaffold is either due to the structural components or the loaded cargo which conducts or induces the coupled functionality. Dual delivery system for osteoinductive and angioinductive factors ensures the co-delivery of two different types of molecules to induce osteogenesis and angiogenesis. Single delivery scaffold for angioinductive and osteoinductive molecule releases single type of molecules which could induce both angiogenesis and osteogenesis. Osteoconductive scaffold consisted of bone constituents releases angioinductive factors. Osteoconductive and angioconductive scaffold composed of components which provide the native substrate features for osteogenesis and angiogenesis. This review article also discusses the studies highlighting the synergism of physico-chemical stimuli as dual functioning feature to enhance angiogenesis and osteogenesis simultaneously. In addition, this article covers one of the least discussed area of the bone regeneration i.e. 'cartilage formation as a median between angiogenesis and osteogenesis'.
Collapse
|
43
|
Shih YV, Varghese S. Tissue engineered bone mimetics to study bone disorders ex vivo: Role of bioinspired materials. Biomaterials 2019; 198:107-121. [PMID: 29903640 PMCID: PMC6281816 DOI: 10.1016/j.biomaterials.2018.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/25/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022]
Abstract
Recent advances in materials development and tissue engineering has resulted in a substantial number of bioinspired materials that recapitulate cardinal features of bone extracellular matrix (ECM) such as dynamic inorganic and organic environment(s), hierarchical organization, and topographical features. Bone mimicking materials, as defined by its self-explanatory term, are developed based on the current understandings of the natural bone ECM during development, remodeling, and fracture repair. Compared to conventional plastic cultures, biomaterials that resemble some aspects of the native environment could elicit a more natural molecular and cellular response relevant to the bone tissue. Although current bioinspired materials are mainly developed to assist tissue repair or engineer bone tissues, such materials could nevertheless be applied to model various skeletal diseases in vitro. This review summarizes the use of bioinspired materials for bone tissue engineering, and their potential to model diseases of bone development and remodeling ex vivo. We largely focus on biomaterials, designed to re-create different aspects of the chemical and physical cues of native bone ECM. Employing these bone-inspired materials and tissue engineered bone surrogates to study bone diseases has tremendous potential and will provide a closer portrayal of disease progression and maintenance, both at the cellular and tissue level. We also briefly touch upon the application of patient-derived stem cells and introduce emerging technologies such as organ-on-chip in disease modeling. Faithful recapitulation of disease pathologies will not only offer novel insights into diseases, but also lead to enabling technologies for drug discovery and new approaches for cell-based therapies.
Collapse
Affiliation(s)
- Yuru Vernon Shih
- Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA.
| | - Shyni Varghese
- Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA; Department of Materials Science and Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
44
|
Collignon AM, Castillo-Dali G, Gomez E, Guilbert T, Lesieur J, Nicoletti A, Acuna-Mendoza S, Letourneur D, Chaussain C, Rochefort GY, Poliard A. Mouse Wnt1-CRE
-Rosa
Tomato
Dental Pulp Stem Cells Directly Contribute to the Calvarial Bone Regeneration Process. Stem Cells 2019; 37:701-711. [DOI: 10.1002/stem.2973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/14/2018] [Accepted: 12/19/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Anne-Margaux Collignon
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
- University Hospitals, AP-HP; Paris France
| | - Gabriel Castillo-Dali
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Eduardo Gomez
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Thomas Guilbert
- Plateforme IMAG'IC, Institut Cochin, Inserm U1016-CNRS UMR8104; University Paris Descartes; Paris France
| | - Julie Lesieur
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Antonino Nicoletti
- INSERM U1148, Laboratory of Vascular Translational Science; University Paris Diderot, University Paris 13, Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE; Paris France
| | - Soledad Acuna-Mendoza
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Didier Letourneur
- INSERM U1148, Laboratory of Vascular Translational Science; University Paris Diderot, University Paris 13, Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE; Paris France
| | - Catherine Chaussain
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
- University Hospitals, AP-HP; Paris France
| | - Gael Y. Rochefort
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| | - Anne Poliard
- EA 2496 Orofacial Pathologies, Imagery, and Biotherapies, Dental School Faculty; University Paris Descartes, and Life Imaging Platform (PIV); Montrouge France
| |
Collapse
|
45
|
Li J, Kang F, Gong X, Bai Y, Dai J, Zhao C, Dou C, Cao Z, Liang M, Dong R, Jiang H, Yang X, Dong S. Ceria nanoparticles enhance endochondral ossification-based critical-sized bone defect regeneration by promoting the hypertrophic differentiation of BMSCs via DHX15 activation. FASEB J 2019; 33:6378-6389. [PMID: 30776318 DOI: 10.1096/fj.201802187r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Central ischemic necrosis is one of the biggest obstacles in the clinical application of traditional tissue-engineered bone (TEB) in critical-sized bone defect regeneration. Because of its ability to promote vascular invasion, endochondral ossification-based TEB has been applied for bone defect regeneration. However, inadequate chondrocyte hypertrophy can hinder vascular invasion and matrix mineralization during endochondral ossification. In light of recent studies suggesting that ceria nanoparticles (CNPs) improve the blood vessel distribution within TEB, we modified TEB scaffold surfaces with CNPs and investigated the effect and mechanism of CNPs on endochondral ossification-based bone regeneration. The CNPs used in this study were synthesized by the microemulsion method and modified with alendronate-anchored polyethylene glycol 600. We showed that CNPs accelerated new bone formation and enhanced endochondral ossification-based bone regeneration in both a subcutaneous ectopic osteogenesis model and a mouse model of critical-sized bone defects. Mechanistically, CNPs significantly promoted endochondral ossification-based bone regeneration by ensuring sufficient hypertrophic differentiation via the activation of the RNA helicase, DEAH (Asp-Glu-Ala-His) box helicase 15, and its downstream target, p38 MAPK. These results suggested that CNPs could be applied as a biomaterial to improve the efficacy of endochondral ossification-based bone regeneration in critical-sized bone defects.-Li, J., Kang, F., Gong, X., Bai, Y., Dai, J., Zhao, C., Dou, C., Cao, Z., Liang, M., Dong, R., Jiang, H., Yang, X., Dong, S. Ceria nanoparticles enhance endochondral ossification-based critical-sized bone defect regeneration by promoting the hypertrophic differentiation of BMSCs via DHX15 activation.
Collapse
Affiliation(s)
- Jianmei Li
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Fei Kang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Xiaoshan Gong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Yun Bai
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Jingjin Dai
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Chunrong Zhao
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Ce Dou
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Zhen Cao
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Mengmeng Liang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Rui Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Hong Jiang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Xiaochao Yang
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns, and Combined Injury, Third Military Medical University, Chongqing, China
| |
Collapse
|
46
|
Petersen A, Princ A, Korus G, Ellinghaus A, Leemhuis H, Herrera A, Klaumünzer A, Schreivogel S, Woloszyk A, Schmidt-Bleek K, Geissler S, Heschel I, Duda GN. A biomaterial with a channel-like pore architecture induces endochondral healing of bone defects. Nat Commun 2018; 9:4430. [PMID: 30361486 PMCID: PMC6202397 DOI: 10.1038/s41467-018-06504-7] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/30/2018] [Indexed: 12/22/2022] Open
Abstract
Biomaterials developed to treat bone defects have classically focused on bone healing via direct, intramembranous ossification. In contrast, most bones in our body develop from a cartilage template via a second pathway called endochondral ossification. The unsolved clinical challenge to regenerate large bone defects has brought endochondral ossification into discussion as an alternative approach for bone healing. However, a biomaterial strategy for the regeneration of large bone defects via endochondral ossification is missing. Here we report on a biomaterial with a channel-like pore architecture to control cell recruitment and tissue patterning in the early phase of healing. In consequence of extracellular matrix alignment, CD146+ progenitor cell accumulation and restrained vascularization, a highly organized endochondral ossification process is induced in rats. Our findings demonstrate that a pure biomaterial approach has the potential to recapitulate a developmental bone growth process for bone healing. This might motivate future strategies for biomaterial-based tissue regeneration.
Collapse
Affiliation(s)
- A Petersen
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - A Princ
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - G Korus
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - A Ellinghaus
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - H Leemhuis
- Matricel GmbH, Kaiserstrasse 100, 52134, Herzogenrath, Germany
| | - A Herrera
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - A Klaumünzer
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - S Schreivogel
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - A Woloszyk
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Department of Orthopaedic Surgery, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Dr, 78229, San Antonio, TX, USA
| | - K Schmidt-Bleek
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - S Geissler
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - I Heschel
- Matricel GmbH, Kaiserstrasse 100, 52134, Herzogenrath, Germany
| | - G N Duda
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
47
|
3D Bone Biomimetic Scaffolds for Basic and Translational Studies with Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19103150. [PMID: 30322134 PMCID: PMC6213614 DOI: 10.3390/ijms19103150] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/04/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are recognized as an attractive tool owing to their self-renewal and differentiation capacity, and their ability to secrete bioactive molecules and to regulate the behavior of neighboring cells within different tissues. Accumulating evidence demonstrates that cells prefer three-dimensional (3D) to 2D culture conditions, at least because the former are closer to their natural environment. Thus, for in vitro studies and in vivo utilization, great effort is being dedicated to the optimization of MSC 3D culture systems in view of achieving the intended performance. This implies understanding cell–biomaterial interactions and manipulating the physicochemical characteristics of biomimetic scaffolds to elicit a specific cell behavior. In the bone field, biomimetic scaffolds can be used as 3D structures, where MSCs can be seeded, expanded, and then implanted in vivo for bone repair or bioactive molecules release. Actually, the union of MSCs and biomaterial has been greatly improving the field of tissue regeneration. Here, we will provide some examples of recent advances in basic as well as translational research about MSC-seeded scaffold systems. Overall, the proliferation of tools for a range of applications witnesses a fruitful collaboration among different branches of the scientific community.
Collapse
|
48
|
Raftery RM, Mencía-Castaño I, Sperger S, Chen G, Cavanagh B, Feichtinger GA, Redl H, Hacobian A, O'Brien FJ. Delivery of the improved BMP-2-Advanced plasmid DNA within a gene-activated scaffold accelerates mesenchymal stem cell osteogenesis and critical size defect repair. J Control Release 2018; 283:20-31. [PMID: 29782946 DOI: 10.1016/j.jconrel.2018.05.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/24/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022]
Abstract
Gene-activated scaffolds have been shown to induce controlled, sustained release of functional transgene both in vitro and in vivo. Bone morphogenetic proteins (BMPs) are potent mediators of osteogenesis however we found that the delivery of plasmid BMP-2 (pBMP-2) alone was not sufficient to enhance bone formation. Therefore, the aim of this study was to assess if the use of a series of modified BMP-2 plasmids could enhance the functionality of a pBMP-2 gene-activated scaffold and ultimately improve bone regeneration when implanted into a critical sized bone defect in vivo. A multi-cistronic plasmid encoding both BMP-2 and BMP-7 (BMP-2/7) was employed as was a BMP-2-Advanced plasmid containing a highly truncated intron sequence. With both plasmids, the highly efficient cytomegalovirus (CMV) promoter sequence was used. However, as there have been reports that the elongated factor 1-α promoter is more efficient, particularly in stem cells, a BMP-2-Advanced plasmid containing the EF1α promoter was also tested. Chitosan nanoparticles (CS) were used to deliver each plasmid to MSCs and induced transient up-regulation of BMP-2 protein expression, in turn significantly enhancing MSC-mediated osteogenesis when compared to untreated controls (p < 0.001). When incorporated into a bone mimicking collagen-hydroxyapatite scaffold, the BMP-2-Advanced plasmid, under the control of the CMV promotor, induced MSCs to produce approximately 2500 μg of calcium per scaffold, significantly higher (p < 0.001) than all other groups. Just 4 weeks post-implantation in vivo, this cell-free gene-activated scaffold induced significantly more bone tissue formation compared to a pBMP-2 gene-activated scaffold (p < 0.001) as indicated by microCT and histomorphometry. Immunohistochemistry revealed that the BMP-2-Advanced plasmid accelerated differentiation of osteoprogenitor cells to mature osteoblasts, thus causing rapid healing of the bone defects. This study confirms that optimising the plasmid construct can enhance the functionality of gene-activated scaffolds and translate to accelerated bone formation in a critical sized defect.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Irene Mencía-Castaño
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Simon Sperger
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, The Austrian Cluster for Tissue Regeneration, European Institute of Excellence on Tissue Engineering and Regenerative Medicine Research (Expertissues EEIG), Vienna, Austria
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Georg A Feichtinger
- Division of Oral Biology, School of Dentistry, Faculty of Medicine and Health, University of Leeds, United Kingdom
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, The Austrian Cluster for Tissue Regeneration, European Institute of Excellence on Tissue Engineering and Regenerative Medicine Research (Expertissues EEIG), Vienna, Austria
| | - Ara Hacobian
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, The Austrian Cluster for Tissue Regeneration, European Institute of Excellence on Tissue Engineering and Regenerative Medicine Research (Expertissues EEIG), Vienna, Austria
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Dept. of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.
| |
Collapse
|
49
|
Silva MDA, Leite YKDC, de Carvalho CES, Feitosa MLT, Alves MMDM, Carvalho FADA, Neto BCV, Miglino MA, Jozala AF, de Carvalho MAM. Behavior and biocompatibility of rabbit bone marrow mesenchymal stem cells with bacterial cellulose membrane. PeerJ 2018; 6:e4656. [PMID: 29736332 PMCID: PMC5933324 DOI: 10.7717/peerj.4656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/01/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Tissue engineering has been shown to exhibit great potential for the creation of biomaterials capable of developing into functional tissues. Cellular expansion and integration depends on the quality and surface-determinant factors of the scaffold, which are required for successful biological implants. The objective of this research was to characterize and evaluate the in vitro characteristics of rabbit bone marrow mesenchymal stem cells (BM-MSCs) associated with a bacterial cellulose membrane (BCM). We assessed the adhesion, expansion, and integration of the biomaterial as well as its ability to induce macrophage activation. Finally, we evaluated the cytotoxicity and toxicity of the BCM. METHODS Samples of rabbit bone marrow were collected. Mesenchymal stem cells were isolated from medullary aspirates to establish fibroblast colony-forming unit assay. Osteogenic, chondrogenic, and adipogenic differentiation was performed. Integration with the BCM was assessed by scanning electron microscopy at 1, 7, and 14 days. Cytotoxicity was assessed via the production of nitric oxide, and BCM toxicity was assessed with the MTT assay; phagocytic activity was also determined. RESULTS The fibroblastoid colony-forming unit (CFU-F) assay showed cells with a fibroblastoid morphology organized into colonies, and distributed across the culture area surface. In the growth curve, two distinct phases, lag and log phase, were observed at 15 days. Multipotentiality of the cells was evident after induction of osteogenic, chondrogenic, and adipogenic lineages. Regarding the BM-MSCs' bioelectrical integration with the BCM, BM-MSCs were anchored in the BCM in the first 24 h. On day 7 of culture, the cytoplasm was scattered, and on day 14, the cells were fully integrated with the biomaterial. We also observed significant macrophage activation; analysis of the MTT assay and the concentration of nitric oxide revealed no cytotoxicity of the biomaterial. CONCLUSION The BCM allowed the expansion and biointegration of bone marrow progenitor cells with a stable cytotoxic profile, thus presenting itself as a biomaterial with potential for tissue engineering.
Collapse
Affiliation(s)
- Marcello de Alencar Silva
- Integrated Nucleus of Morphology and Stem Cell Research, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | | | - Matheus Levi Tajra Feitosa
- Integrated Nucleus of Morphology and Stem Cell Research, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | | | - Bartolomeu Cruz Viana Neto
- Department of Physics/Advanced Microscopy Multiuser Laboratory/Laboratory of Physics Material, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Maria Angélica Miglino
- Departament of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Angela Faustino Jozala
- Laboratory of Industrial Microbiology and Fermentation Process, University of Sorocaba, Sorocaba, São Paulo, Brazil
| | | |
Collapse
|
50
|
Markides H, McLaren JS, Telling ND, Alom N, Al-Mutheffer EA, Oreffo ROC, Zannettino A, Scammell BE, White LJ, El Haj AJ. Translation of remote control regenerative technologies for bone repair. NPJ Regen Med 2018; 3:9. [PMID: 29675269 PMCID: PMC5904134 DOI: 10.1038/s41536-018-0048-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 02/22/2018] [Accepted: 03/22/2018] [Indexed: 01/06/2023] Open
Abstract
The role of biomechanical stimuli, or mechanotransduction, in normal bone homeostasis and repair is understood to facilitate effective osteogenesis of mesenchymal stem cells (MSCs) in vitro. Mechanotransduction has been integrated into a multitude of in vitro bone tissue engineering strategies and provides an effective means of controlling cell behaviour towards therapeutic outcomes. However, the delivery of mechanical stimuli to exogenous MSC populations, post implantation, poses a significant translational hurdle. Here, we describe an innovative bio-magnetic strategy, MICA, where magnetic nanoparticles (MNPs) are used to remotely deliver mechanical stimuli to the mechano-receptor, TREK-1, resulting in activation and downstream signalling via an external magnetic array. In these studies, we have translated MICA to a pre-clinical ovine model of bone injury to evaluate functional bone repair. We describe the development of a magnetic array capable of in vivo MNP manipulation and subsequent osteogenesis at equivalent field strengths in vitro. We further demonstrate that the viability of MICA-activated MSCs in vivo is unaffected 48 h post implantation. We present evidence to support early accelerated repair and preliminary enhanced bone growth in MICA-activated defects within individuals compared to internal controls. The variability in donor responses to MICA-activation was evaluated in vitro revealing that donors with poor osteogenic potential were most improved by MICA-activation. Our results demonstrate a clear relationship between responders to MICA in vitro and in vivo. These unique experiments offer exciting clinical applications for cell-based therapies as a practical in vivo source of dynamic loading, in real-time, in the absence of pharmacological agents.
Collapse
Affiliation(s)
- Hareklea Markides
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB UK
| | - Jane S. McLaren
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD UK
| | - Neil D. Telling
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB UK
| | - Noura Alom
- Centre for Biomolecular Sciences, University of Nottingham, Nottingham, NG7 2RD UK
| | | | - Richard O. C. Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Andrew Zannettino
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, and South Australian Health and Medical Research Institute, Adelaide, SA 5000 Australia
| | - Brigitte E. Scammell
- Academic Orthopaedics, Trauma and Sports Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham, NG7 2UH UK
| | - Lisa J. White
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD UK
| | - Alicia J. El Haj
- Institute for Science and Technology in Medicine, Keele University, Stoke-on-Trent, ST4 7QB UK
| |
Collapse
|