1
|
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
|
2
|
Zhang W, Liu M, Wu D, Hao Y, Cong B, Wang L, Wang Y, Gao M, Xu Y, Wu Y. PSO/SDF-1 composite hydrogel promotes osteogenic differentiation of PDLSCs and bone regeneration in periodontitis rats. Heliyon 2024; 10:e32686. [PMID: 38961957 PMCID: PMC11220005 DOI: 10.1016/j.heliyon.2024.e32686] [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: 02/20/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024] Open
Abstract
Periodontitis is an inflammatory disease characterized by the destruction of periodontal tissues, and the promotion of bone tissue regeneration is the key to curing periodontitis. Psoralen is the main component of Psoralea corylifolia Linn, and has multiple biological effects, including anti-osteoporosis and osteogenesis. We constructed a novel hydrogel loaded with psoralen (PSO) and stromal cell-derived factor-1 (SDF-1) for direct endogenous cell homing. This study aimed to evaluate the synergistic effects of PSO/SDF-1 on periodontal bone regeneration in patients with periodontitis. The results of CCK8, alkaline phosphatase (ALP) activity assay, and Alizarin Red staining showed that PSO/SDF-1 combination treatment promoted cell proliferation, chemotaxis ability, and ALP activity of PDLSCs. qRT-PCR and western blotting showed that the expression levels of alkaline phosphatase (ALP), dwarf-associated transcription factor 2 (RUNX2), and osteocalcin (OCN) gene were upregulated. Rat periodontal models were established to observe the effect of local application of the composite hydrogel on bone regeneration. These results proved that the PSO/SDF-1 combination treatment significantly promoted new bone formation. The immunohistochemical (IHC) results confirmed the elevated expression of ALP, RUNX2, and OCN osteogenic genes. PSO/SDF-1 composite hydrogel can synergistically regulate the biological function and promote periodontal bone formation. Thus, this study provides a novel strategy for periodontal bone regeneration.
Collapse
Affiliation(s)
- Wei Zhang
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, China
| | - Minghong Liu
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| | - Di Wu
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| | - Yuanping Hao
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| | - Beibei Cong
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| | - Lihui Wang
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, China
| | - Yujia Wang
- School of Stomatology, Shandong Second Medical University, Weifang, 261053, China
| | - Meihua Gao
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| | - Yingjie Xu
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| | - Yingtao Wu
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| |
Collapse
|
3
|
Yao M, Liang S, Zeng Y, Peng F, Zhao X, Du C, Ma X, Huang H, Wang D, Zhang Y. Dual Factor-Loaded Artificial Periosteum Accelerates Bone Regeneration. ACS Biomater Sci Eng 2024; 10:2200-2211. [PMID: 38447138 DOI: 10.1021/acsbiomaterials.3c01587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
In the clinic, inactivation of osteosarcoma using microwave ablation would damage the periosteum, resulting in frequent postoperative complications. Therefore, the development of an artificial periosteum is crucial for postoperative healing. In this study, we prepared an artificial periosteum using silk fibroin (SF) loaded with stromal cell-derived factor-1α (SDF-1α) and calcitonin gene-related peptide (CGRP) to accelerate bone remodeling after the microwave ablation of osteosarcoma. The prepared artificial periosteum showed a sustained release of SDF-1α and CGRP after 14 days of immersion. In vitro culture of rat periosteal stem cells (rPDSCs) demonstrated that the artificial periosteum is favorable for cell recruitment, the activity of alkaline phosphatase, and bone-related gene expression. Furthermore, the artificial periosteum improved the tube formation and angiogenesis-related gene expression of human umbilical vein endothelial cells (HUVECs). In an animal study, the periosteum in the femur of a rabbit was inactivated through microwave ablation and then removed. The damaged periosteum was replaced with the as-prepared artificial periosteum and favored bone regeneration. In all, the designed dual-factor-loaded artificial periosteum is a promising strategy to replace the damaged periosteum in the therapy of osteosarcoma for a better bone-rebuilding process.
Collapse
Affiliation(s)
- Mengyu Yao
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangzhou 510080, China
| | - Shengjie Liang
- Henan Key Laboratory of Energy Storage Materials and Processes, Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450003, China
| | - Yanyan Zeng
- Department of Hyperbaric Oxygen Rehabilitation (Intensive Rehabilitation Center), Southern Theater Command General Hospital of PLA, Guangzhou 510010, Guangdong, China
| | - Feng Peng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangzhou 510080, China
| | - Xiujuan Zhao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Chang Du
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Xiaohan Ma
- Division of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, U.K
| | - Huai Huang
- Department of Hyperbaric Oxygen Rehabilitation (Intensive Rehabilitation Center), Southern Theater Command General Hospital of PLA, Guangzhou 510010, Guangdong, China
| | - Donghui Wang
- Hebei Key Laboratory of Biomaterials and Smart Theranostics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin 300130, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
- Guangdong Engineering Technology Research Center of Functional Repair of Bone Defects and Biomaterials, Guangzhou 510080, China
| |
Collapse
|
4
|
Cai P, Li C, Ding Y, Lu H, Yu X, Cui J, Yu F, Wang H, Wu J, El-Newehy M, Abdulhameed MM, Song L, Mo X, Sun B. Elastic 3D-Printed Nanofibers Composite Scaffold for Bone Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2023; 15:54280-54293. [PMID: 37973614 DOI: 10.1021/acsami.3c12426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Loading nanoparticles into hydrogels has been a conventional approach to augment the printability of ink and the physicochemical characteristics of scaffolds in three-dimensional (3D) printing. However, the efficacy of this enhancement has often proven to be limited. We amalgamate electrospun nanofibers with 3D printing techniques to fabricate a composite scaffold reminiscent of a "reinforced concrete" structure, aimed at addressing bone defects. These supple silica nanofibers are synthesized through a dual-step process involving high-speed homogenization and low-temperature ball milling technology. The nanofibers are homogeneously blended with sodium alginate to create the printing ink. The resultant ink was extruded seamlessly, displaying commendable molding properties, thereby yielding scaffolds with favorable macroscopic morphology. In contrast to nanoparticle-reinforced scaffolds, composite scaffolds containing nanofibers exhibit superior mechanical attributes and bioactivity. These nanofiber composite scaffolds demonstrate enhanced osteoinductive properties in both in vitro and in vivo evaluations. To conclude, this research introduces a novel 3D printing approach where the fabricated nanofiber-infused 3D-printed scaffolds hold the potential to revolutionize the realm of 3D printing in the domain of bone tissue engineering.
Collapse
Affiliation(s)
- Pengfei Cai
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Chunchun Li
- Department of Stomatology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Yangfan Ding
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Hanting Lu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Xiao Yu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Jie Cui
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Fan Yu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Hongsheng Wang
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Jinglei Wu
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Mohamed El-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Meera Moydeen Abdulhameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Liang Song
- Department of Stomatology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xiumei Mo
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Binbin Sun
- Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine & College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, P. R. China
| |
Collapse
|
5
|
Huan Y, Zhou D, Wu X, He X, Chen H, Li S, Jia B, Dou Y, Fei X, Wu S, Wei J, Fei Z, Xu T, Fei F. 3D bioprinted autologous bone particle scaffolds for cranioplasty promote bone regeneration with both implanted and native BMSCs. Biofabrication 2023; 15. [PMID: 36812580 DOI: 10.1088/1758-5090/acbe21] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/22/2023] [Indexed: 02/24/2023]
Abstract
Although autologous bone (AB) grafting is considered to be the gold standard for cranioplasty, unresolved problems remain, such as surgical-site infections and bone flap absorption. In this study, an AB scaffold was constructed via three-dimensional (3D) bedside-bioprinting technology and used for cranioplasty. To simulate the skull structure, a polycaprolactone shell was designed as an external lamina, and 3D-printed AB and a bone marrow-derived mesenchymal stem cell (BMSC) hydrogel was used to mimic cancellous bone for bone regeneration. Ourin vitroresults showed that the scaffold exhibited excellent cellular affinity and promoted osteogenic differentiation of BMSCs in both two-dimensional and 3D culture systems. The scaffold was implanted in beagle dog cranial defects for up to 9 months, and the scaffold promoted new bone and osteoid formation. Furtherin vivostudies indicated that transplanted BMSCs differentiated into vascular endothelium, cartilage, and bone tissues, whereas native BMSCs were recruited into the defect. The results of this study provide a method for bedside bioprinting of a cranioplasty scaffold for bone regeneration, which opens up another window for clinical applications of 3D printing in the future.
Collapse
Affiliation(s)
- Yu Huan
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Xin He
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Hongqing Chen
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Sanzhong Li
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Bo Jia
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Yanan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Xiaowei Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Shuang Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Jialiang Wei
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Tao Xu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Department of Precision Medicine and Healthcare, Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, People's Republic of China
- Center for Bio-intelligent Manufacturing and Living Matter Bioprinting, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, People's Republic of China
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, Air Force Medical University, Xi'an 710032, People's Republic of China
| |
Collapse
|
6
|
Tang X, Yang F, Chu G, Li X, Fu Q, Zou M, Zhao P, Lu G. Characterizing the inherent activity of urinary bladder matrix for adhesion, migration, and activation of fibroblasts as compared with collagen-based synthetic scaffold. J Biomater Appl 2023; 37:1446-1457. [PMID: 36177498 DOI: 10.1177/08853282221130883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The mechanism of action underlying the intriguing prominent bioactivity of urinary bladder matrix (UBM) for in situ tissue regeneration of soft tissue defects remains to be elucidated. It is speculated that the activity of UBM for cell adhesion, migration, and activation is inherent. The bioactivity of UBM for in situ tissue regeneration and its relation with the structure and intact soluble components of UBM were investigated in comparison to a collagen-based scaffold, PELNAC (PEL). We isolated the soluble component of the two materials with urea buffer, and evaluated the respective effect of these soluble components on the in vitro adhesion and migration of L929 fibroblasts. The spatiotemporal pattern of endogenous-cell ingrowth into the scaffolds and cell activation were investigated using a model of murine subcutaneous implantation. UBM is more capable of promoting the adhesion, migration, and proliferation of fibroblasts than PEL in a serum-independent manner. In vivo, as compared with PEL, UBM exhibits significantly enhanced activity for fast endogenous cell ingrowth and produces a more prominent pro-regenerative and pro-remodeling microenvironment by inducing the expression of TGF-β1, VEGF, MMP-9, and murine type I collagen. Overall, our results suggest the prominent bioactivity of UBM for in situ tissue regeneration is inherent.
Collapse
Affiliation(s)
- Xiaoyu Tang
- 66478Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Guoping Chu
- 199193Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaoxiao Li
- 66478Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiuyan Fu
- 66374Jiangnan University, Wuxi, China
| | - Mingli Zou
- 66478Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Zhao
- 199193Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Guozhong Lu
- 199193Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
7
|
Sun H, Zhou X, Zhang Y, Zhang L, Yu X, Ye Z, Laurencin CT. Bone Implants (Bone Regeneration and Bone Cancer Treatments). BIOFABRICATION FOR ORTHOPEDICS 2022:265-321. [DOI: 10.1002/9783527831371.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
8
|
Peng J, Zhao J, Tang Q, Wang J, Song W, Lu X, Huang X, Chen G, Zheng W, Zhang L, Han Y, Yan C, Wan Q, Chen L. Low intensity near-infrared light promotes bone regeneration via circadian clock protein cryptochrome 1. Int J Oral Sci 2022; 14:53. [PMID: 36376275 PMCID: PMC9663728 DOI: 10.1038/s41368-022-00207-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/04/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Bone regeneration remains a great clinical challenge. Low intensity near-infrared (NIR) light showed strong potential to promote tissue regeneration, offering a promising strategy for bone defect regeneration. However, the effect and underlying mechanism of NIR on bone regeneration remain unclear. We demonstrated that bone regeneration in the rat skull defect model was significantly accelerated with low-intensity NIR stimulation. In vitro studies showed that NIR stimulation could promote the osteoblast differentiation in bone mesenchymal stem cells (BMSCs) and MC3T3-E1 cells, which was associated with increased ubiquitination of the core circadian clock protein Cryptochrome 1 (CRY1) in the nucleus. We found that the reduction of CRY1 induced by NIR light activated the bone morphogenetic protein (BMP) signaling pathways, promoting SMAD1/5/9 phosphorylation and increasing the expression levels of Runx2 and Osterix. NIR light treatment may act through sodium voltage-gated channel Scn4a, which may be a potential responder of NIR light to accelerate bone regeneration. Together, these findings suggest that low-intensity NIR light may promote in situ bone regeneration in a CRY1-dependent manner, providing a novel, efficient and non-invasive strategy to promote bone regeneration for clinical bone defects.
Collapse
|
9
|
Wang YH, Zhao CZ, Wang RY, Du QX, Liu JY, Pan J. The crosstalk between macrophages and bone marrow mesenchymal stem cells in bone healing. Stem Cell Res Ther 2022; 13:511. [PMID: 36333820 PMCID: PMC9636722 DOI: 10.1186/s13287-022-03199-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Bone injury plagues millions of patients worldwide every year, and it demands a heavy portion of expense from the public medical insurance system. At present, orthopedists think that autologous bone transplantation is the gold standard for treating large-scale bone defects. However, this method has significant limitations, which means that parts of patients cannot obtain a satisfactory prognosis. Therefore, a basic study on new therapeutic methods is urgently needed. The in-depth research on crosstalk between macrophages (Mϕs) and bone marrow mesenchymal stem cells (BMSCs) suggests that there is a close relationship between inflammation and regeneration. The in-depth understanding of the crosstalk between Mϕs and BMSCs is helpful to amplify the efficacy of stem cell-based treatment for bone injury. Only in the suitable inflammatory microenvironment can the damaged tissues containing stem cells obtain satisfactory healing outcomes. The excessive tissue inflammation and lack of stem cells make the transplantation of biomaterials necessary. We can expect that the crosstalk between Mϕs and BMSCs and biomaterials will become the mainstream to explore new methods for bone injury in the future. This review mainly summarizes the research on the crosstalk between Mϕs and BMSCs and also briefly describes the effects of biomaterials and aging on cell transplantation therapy.
Collapse
Affiliation(s)
- Yu-Hao Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Cheng-Zhi Zhao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Ren-Yi Wang
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Qian-Xin Du
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| | - Ji-Yuan Liu
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China
| | - Jian Pan
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, #14 Third Section, Renmin Road South, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 People’s Republic of China ,grid.13291.380000 0001 0807 1581Chengdu Advanced Medical Science Center, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan Province People’s Republic of China
| |
Collapse
|
10
|
Calcium Phosphate-Based Biomaterials for Bone Repair. J Funct Biomater 2022; 13:jfb13040187. [PMID: 36278657 PMCID: PMC9589993 DOI: 10.3390/jfb13040187] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022] Open
Abstract
Traumatic, tumoral, and infectious bone defects are common in clinics, and create a big burden on patient's families and society. Calcium phosphate (CaP)-based biomaterials have superior properties and have been widely used for bone defect repair, due to their similarities to the inorganic components of human bones. The biological performance of CaPs, as a determining factor for their applications, are dependent on their physicochemical properties. Hydroxyapatite (HAP) as the most thermally stable crystalline phase of CaP is mostly used in the form of ceramics or composites scaffolds with polymers. Nanostructured CaPs with large surface areas are suitable for drug/gene delivery systems. Additionally, CaP scaffolds with hierarchical nano-/microstructures have demonstrated excellent ability in promoting bone regeneration. This review focuses on the relationships and interactions between the physicochemical/biological properties of CaP biomaterials and their species, sizes, and morphologies in bone regeneration, including synthesis strategies, structure control, biological behavior, and the mechanisms of CaP in promoting osteogenesis. This review will be helpful for scientists and engineers to further understand CaP-based biomaterials (CaPs), and be useful in developing new high-performance biomaterials for bone repair.
Collapse
|
11
|
Meng L, Wei Y, Liang Y, Hu Q, Xie H. Stem cell homing in periodontal tissue regeneration. Front Bioeng Biotechnol 2022; 10:1017613. [PMID: 36312531 PMCID: PMC9607953 DOI: 10.3389/fbioe.2022.1017613] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022] Open
Abstract
The destruction of periodontal tissue is a crucial problem faced by oral diseases, such as periodontitis and tooth avulsion. However, regenerating periodontal tissue is a huge clinical challenge because of the structural complexity and the poor self-healing capability of periodontal tissue. Tissue engineering has led to advances in periodontal regeneration, however, the source of exogenous seed cells is still a major obstacle. With the improvement of in situ tissue engineering and the exploration of stem cell niches, the homing of endogenous stem cells may bring promising treatment strategies in the future. In recent years, the applications of endogenous cell homing have been widely reported in clinical tissue repair, periodontal regeneration, and cell therapy prospects. Stimulating strategies have also been widely studied, such as the combination of cytokines and chemokines, and the implantation of tissue-engineered scaffolds. In the future, more research needs to be done to improve the efficiency of endogenous cell homing and expand the range of clinical applications.
Collapse
Affiliation(s)
- Lingxi Meng
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yige Wei
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaxian Liang
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qin Hu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Huixu Xie
- State Key Laboratory of Oral Diseases, Department of Head and Neck Oncology Surgery, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Huixu Xie,
| |
Collapse
|
12
|
Zeng Y, Huang C, Duan D, Lou A, Guo Y, Xiao T, Wei J, Liu S, Wang Z, Yang Q, Zhou L, Wu Z, Wang L. Injectable temperature-sensitive hydrogel system incorporating deferoxamine-loaded microspheres promotes H-type blood vessel-related bone repair of a critical size femoral defect. Acta Biomater 2022; 153:108-123. [PMID: 36115651 DOI: 10.1016/j.actbio.2022.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/01/2022]
Abstract
Insufficient vascularization is a major challenge in the repair of critical-sized bone defects. Deferoxamine (DFO) has been reported to play a potential role in promoting the formation of H-type blood vessels, a specialized vascular subtype with coupled angiogenesis and osteogenesis. However, whether DFO promotes the expression of H-type vessels in critical femoral defects with complete periosteal damage remains unknown. Moreover, stable drug loading systems need to be designed owing to the short half-life and high-dose toxic effects of DFO. In this study, we developed an injectable DFO-gelatin microspheres (GMs) hydrogel complex as a stable drug loading system for the treatment of critical femoral defects in rats. Our results showed that sustained release of DFO in critical femoral defects stimulated the generation of functional H-type vessels. The DFO-GMs hydrogel complex effectively promoted proliferation, formation, and migration of human umbilical vein endothelial cells in vitro. In vivo, the application of the DFO-GMs hydrogel complex expanded the distribution range and prolonged the expression time of H-type vessels in the defect area and was positively correlated with the number of osterix+ cells and new bone tissue. Topical application of the HIF-1α inhibitor PX-478 partially blocked the stimulation of H-type vessels by DFO, whereas the osteogenic potential of the latter was also weakened. Our results extended the local application of DFO and provided a theoretical basis for targeting H-type vessels to treat large femoral defects. STATEMENT OF SIGNIFICANCE: Abundant functional blood vessels are essential for bone repair. The H-type blood vessel is a functional subtype with angiogenesis and osteogenesis coupling potential. A drug loading system with long-term controlled release was first used to investigate the formation of H-type blood vessels in critical femoral defects and promotion of bone repair. Our results showed that the application of DFO-GMs hydrogel complex expanded the distribution range and expression time of H-type vessels, and was positively correlated with the number of osteoblasts and volume of new bone tissue. These results expanded the local application approach of DFO and provide a theoretical basis for targeting H-type vessels to treat large femoral defects.
Collapse
Affiliation(s)
- Yuwei Zeng
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Chuang Huang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Dongming Duan
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Aiju Lou
- Department of Rheumatology, Liwan Central Hospital of Guangzhou, 35 Liwan Road, Guangzhou 510030, China
| | - Yuan Guo
- Department of Stomatology, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Tianhua Xiao
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Jianguo Wei
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Song Liu
- Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Zhao Wang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Qihao Yang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| | - Zenghui Wu
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| | - Le Wang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| |
Collapse
|
13
|
Wang Z, Zhao H, Tang X, Meng T, Khutsishvili D, Xu B, Ma S. CNS Organoid Surpasses Cell-Laden Microgel Assembly to Promote Spinal Cord Injury Repair. Research (Wash D C) 2022; 2022:9832128. [PMID: 36061824 PMCID: PMC9394056 DOI: 10.34133/2022/9832128] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/24/2022] [Indexed: 11/06/2022] Open
Abstract
The choice of therapeutic agents remains an unsolved issue in the repair of spinal cord injury. In this work, various agents and configurations were investigated and compared for their performance in promoting nerve regeneration, including bead assembly and bulk gel of collagen and Matrigel, under acellular and cell-laden conditions, and cerebral organoid (CO) as the in vitro preorganized agent. First, in Matrigel-based agents and the CO transplantations, the recipient animal gained more axon regeneration and the higher Basso, Beattie, and Bresnahan (BBB) scoring than the grafted collagen gels. Second, new nerves more uniformly infiltrated into the transplants in bead form assembly than the molded chunks. Third, the materials loaded the neural progenitor cells (NPCs) or the CO implantation groups received more regenerated nerve fibers than their acellular counterparts, suggesting the necessity to transplant exogenous cells for large trauma (e.g., a 5 mm long spinal cord transect). In addition, the activated microglial cells might benefit from neural regeneration after receiving CO transplantation in the recipient animals. The organoid augmentation may suggest that in vitro maturation of a microtissue complex is necessary before transplantation and proposes organoids as the premium therapeutic agents for nerve regeneration.
Collapse
Affiliation(s)
- Zitian Wang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Haoran Zhao
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
| | - Xiaowei Tang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
| | - Tianyu Meng
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
| | - Davit Khutsishvili
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
| | - Bing Xu
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen, China
- Institute for Brain and Cognitive Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
14
|
Yao D, Lv Y. A cell-free difunctional demineralized bone matrix scaffold enhances the recruitment and osteogenesis of mesenchymal stem cells by promoting inflammation resolution. BIOMATERIALS ADVANCES 2022; 139:213036. [PMID: 35905556 DOI: 10.1016/j.bioadv.2022.213036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
The dialogue between host macrophages (Mφs) and endogenous mesenchymal stem cells (MSCs) promotes M2 Mφs polarization to resolve early-stage inflammation, thereby effectively guiding in situ bone regeneration. Once inflammation is unresolved/incontrollable, it will induce the impediment of MSCs homing at bone defect site, implying the seasonable resolution of inflammation in balancing bone homeostasis. Repeatedly, evidence elucidated that specialized pro-resolving mediators (SPMs) could conduce to proper resolve inflammation and promote the repairing of bone defect. A difunctional demineralized bone matrix (DBM) scaffold co-modified by maresin 1 (MaR1) and aptamer 19S (Apt19S) was fabricated to facilitate the osteogenesis of MSCs. To confirm the osteogenesis and immunomodulatory role of the difunctional DBM scaffold, the proliferation, recruitment, and osteogenic differentiation of MSCs and the Mφs M2 subtype polarization were evaluated in vitro. Then, the DBM scaffolds were implanted into mice model with critical size calvarial defect to evaluate bone repair efficiency. Finally, the specific resolution mechanism in Mφs cultured on the difunctional DBM scaffold was further in-depth investigated. This difunctional DBM scaffold exhibited an enhanced function on the recruitment, proliferation, migration, osteogenesis of MSCs and the resolution of inflammation, finally improved bone-scaffold integration. At the same time, MaR1 modified on the difunctional DBM scaffold increased the biosynthesis of 12-lipoxygenase (12-LOX) and 12S-hydroxy-eicosatetraenoic acid (12S-HETE), and also directly stimulated lipid droplets (LDs) biogenesis in Mφs, which suggested that MaR1 regulated Mφ lipid metabolism at bone repair site. Findings based on this synergy strategy demonstrated that Mφ lipid metabolism was essential in bone homeostasis, which might provide a theoretical direction for the treatment-associated application of MaR1 in inflammatory bone disease.
Collapse
Affiliation(s)
- Dongdong Yao
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, PR China.
| |
Collapse
|
15
|
Guimarães CF, Marques AP, Reis RL. Pushing the Natural Frontier: Progress on the Integration of Biomaterial Cues toward Combinatorial Biofabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105645. [PMID: 35419887 DOI: 10.1002/adma.202105645] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The engineering of fully functional, biological-like tissues requires biomaterials to direct cellular events to a near-native, 3D niche extent. Natural biomaterials are generally seen as a safe option for cell support, but their biocompatibility and biodegradability can be just as limited as their bioactive/biomimetic performance. Furthermore, integrating different biomaterial cues and their final impact on cellular behavior is a complex equation where the outcome might be very different from the sum of individual parts. This review critically analyses recent progress on biomaterial-induced cellular responses, from simple adhesion to more complex stem cell differentiation, looking at the ever-growing possibilities of natural materials modification. Starting with a discussion on native material formulation and the inclusion of cell-instructive cues, the roles of shape and mechanical stimuli, the susceptibility to cellular remodeling, and the often-overlooked impact of cellular density and cell-cell interactions within constructs, are delved into. Along the way, synergistic and antagonistic combinations reported in vitro and in vivo are singled out, identifying needs and current lessons on the development of natural biomaterial libraries to solve the cell-material puzzle efficiently. This review brings together knowledge from different fields envisioning next-generation, combinatorial biomaterial development toward complex tissue engineering.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
16
|
Young SAE, Rummler M, Taïeb HM, Garske DS, Ellinghaus A, Duda GN, Willie BM, Cipitria A. In vivo microCT-based time-lapse morphometry reveals anatomical site-specific differences in bone (re)modeling serving as baseline parameters to detect early pathological events. Bone 2022; 161:116432. [PMID: 35569733 DOI: 10.1016/j.bone.2022.116432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022]
Abstract
The bone structure is very dynamic and continuously adapts its geometry to external stimuli by modeling and remodeling the mineralized tissue. In vivo microCT-based time-lapse morphometry is a powerful tool to study the temporal and spatial dynamics of bone (re)modeling. Here an advancement in the methodology to detect and quantify site-specific differences in bone (re)modeling of 12-week-old BALB/c nude mice is presented. We describe our method of quantifying new bone surface interface readouts and how these are influenced by bone curvature. This method is then used to compare bone surface (re)modeling in mice across different anatomical regions to demonstrate variations in the rate of change and spatial gradients thereof. Significant differences in bone (re)modeling baseline parameters between the metaphyseal and epiphyseal, as well as cortical and trabecular bone of the distal femur and proximal tibia are shown. These results are validated using conventional static in vivo microCT analysis. Finally, the insights from these new baseline values of physiological bone (re)modeling were used to evaluate pathological bone (re)modeling in a pilot breast cancer bone metastasis model. The method shows the potential to be suitable to detect early pathological events and track their spatio-temporal development in both cortical and trabecular bone. This advancement in (re)modeling surface analysis and defined baseline parameters according to distinct anatomical regions will be valuable to others investigating various disease models with site-distinct local alterations in bone (re)modeling.
Collapse
Affiliation(s)
- Sarah A E Young
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Maximilian Rummler
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany; Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Department of Pediatric Surgery, McGill University, Montreal, Canada
| | - Hubert M Taïeb
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Daniela S Garske
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute & Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute & Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bettina M Willie
- Research Centre, Shriners Hospital for Children-Canada, Montreal, Canada; Department of Pediatric Surgery, McGill University, Montreal, Canada
| | - Amaia Cipitria
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany; Biodonostia Health Research Institute, Group of Bioengineering in Regeneration and Cancer, San Sebastian, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
17
|
Hao Z, Li H, Wang Y, Hu Y, Chen T, Zhang S, Guo X, Cai L, Li J. Supramolecular Peptide Nanofiber Hydrogels for Bone Tissue Engineering: From Multihierarchical Fabrications to Comprehensive Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103820. [PMID: 35128831 PMCID: PMC9008438 DOI: 10.1002/advs.202103820] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/02/2022] [Indexed: 05/03/2023]
Abstract
Bone tissue engineering is becoming an ideal strategy to replace autologous bone grafts for surgical bone repair, but the multihierarchical complexity of natural bone is still difficult to emulate due to the lack of suitable biomaterials. Supramolecular peptide nanofiber hydrogels (SPNHs) are emerging biomaterials because of their inherent biocompatibility, satisfied biodegradability, high purity, facile functionalization, and tunable mechanical properties. This review initially focuses on the multihierarchical fabrications by SPNHs to emulate natural bony extracellular matrix. Structurally, supramolecular peptides based on distinctive building blocks can assemble into nanofiber hydrogels, which can be used as nanomorphology-mimetic scaffolds for tissue engineering. Biochemically, bioactive motifs and bioactive factors can be covalently tethered or physically absorbed to SPNHs to endow various functions depending on physiological and pharmacological requirements. Mechanically, four strategies are summarized to optimize the biophysical microenvironment of SPNHs for bone regeneration. Furthermore, comprehensive applications about SPNHs for bone tissue engineering are reviewed. The biomaterials can be directly used in the form of injectable hydrogels or composite nanoscaffolds, or they can be used to construct engineered bone grafts by bioprinting or bioreactors. Finally, continuing challenges and outlook are discussed.
Collapse
Affiliation(s)
- Zhuowen Hao
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Hanke Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yi Wang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Yingkun Hu
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Tianhong Chen
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Shuwei Zhang
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Xiaodong Guo
- Department of OrthopedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyJiefang Road 1277Wuhan430022China
| | - Lin Cai
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| | - Jingfeng Li
- Department of OrthopedicsZhongnan Hospital of Wuhan UniversityDonghu Road 169Wuhan430071China
| |
Collapse
|
18
|
Microenvironmental stiffness mediates cytoskeleton re-organization in chondrocytes through laminin-FAK mechanotransduction. Int J Oral Sci 2022; 14:15. [PMID: 35277477 PMCID: PMC8917190 DOI: 10.1038/s41368-022-00165-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/07/2022] [Accepted: 01/24/2022] [Indexed: 12/11/2022] Open
Abstract
AbstractMicroenvironmental biophysical factors play a fundamental role in controlling cell behaviors including cell morphology, proliferation, adhesion and differentiation, and even determining the cell fate. Cells are able to actively sense the surrounding mechanical microenvironment and change their cellular morphology to adapt to it. Although cell morphological changes have been considered to be the first and most important step in the interaction between cells and their mechanical microenvironment, their regulatory network is not completely clear. In the current study, we generated silicon-based elastomer polydimethylsiloxane (PDMS) substrates with stiff (15:1, PDMS elastomer vs. curing agent) and soft (45:1) stiffnesses, which showed the Young’s moduli of ~450 kPa and 46 kPa, respectively, and elucidated a new path in cytoskeleton re-organization in chondrocytes in response to changed substrate stiffnesses by characterizing the axis shift from the secreted extracellular protein laminin β1, focal adhesion complex protein FAK to microfilament bundling. We first showed the cellular cytoskeleton changes in chondrocytes by characterizing the cell spreading area and cellular synapses. We then found the changes of secreted extracellular linkage protein, laminin β1, and focal adhesion complex protein, FAK, in chondrocytes in response to different substrate stiffnesses. These two proteins were shown to be directly interacted by Co-IP and colocalization. We next showed that impact of FAK on the cytoskeleton organization by showing the changes of microfilament bundles and found the potential intermediate regulators. Taking together, this modulation axis of laminin β1-FAK-microfilament could enlarge our understanding about the interdependence among mechanosensing, mechanotransduction, and cytoskeleton re-organization.
Collapse
|
19
|
Power RN, Cavanagh BL, Dixon JE, Curtin CM, O’Brien FJ. Development of a Gene-Activated Scaffold Incorporating Multifunctional Cell-Penetrating Peptides for pSDF-1α Delivery for Enhanced Angiogenesis in Tissue Engineering Applications. Int J Mol Sci 2022; 23:1460. [PMID: 35163379 PMCID: PMC8835777 DOI: 10.3390/ijms23031460] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/18/2022] Open
Abstract
Non-viral gene delivery has become a popular approach in tissue engineering, as it permits the transient delivery of a therapeutic gene, in order to stimulate tissue repair. However, the efficacy of non-viral delivery vectors remains an issue. Our lab has created gene-activated scaffolds by incorporating various non-viral delivery vectors, including the glycosaminoglycan-binding enhanced transduction (GET) peptide into collagen-based scaffolds with proven osteogenic potential. A modification to the GET peptide (FLR) by substitution of arginine residues with histidine (FLH) has been designed to enhance plasmid DNA (pDNA) delivery. In this study, we complexed pDNA with combinations of FLR and FLH peptides, termed GET* nanoparticles. We sought to enhance our gene-activated scaffold platform by incorporating GET* nanoparticles into collagen-nanohydroxyapatite scaffolds with proven osteogenic capacity. GET* N/P 8 was shown to be the most effective formulation for delivery to MSCs in 2D. Furthermore, GET* N/P 8 nanoparticles incorporated into collagen-nanohydroxyapatite (coll-nHA) scaffolds at a 1:1 ratio of collagen:nanohydroxyapatite was shown to be the optimal gene-activated scaffold. pDNA encoding stromal-derived factor 1α (pSDF-1α), an angiogenic chemokine which plays a role in BMP mediated differentiation of MSCs, was then delivered to MSCs using our optimised gene-activated scaffold platform, with the aim of significantly increasing angiogenesis as an important precursor to bone repair. The GET* N/P 8 coll-nHA scaffolds successfully delivered pSDF-1α to MSCs, resulting in a significant, sustained increase in SDF-1α protein production and an enhanced angiogenic effect, a key precursor in the early stages of bone repair.
Collapse
Affiliation(s)
- Rachael N. Power
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), D02 YN77 Dublin, Ireland; (R.N.P.); (C.M.C.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI, D02 YN77 Dublin, Ireland
| | | | - James E. Dixon
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Caroline M. Curtin
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), D02 YN77 Dublin, Ireland; (R.N.P.); (C.M.C.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI, D02 YN77 Dublin, Ireland
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Royal College of Surgeons in Ireland (RCSI), D02 YN77 Dublin, Ireland; (R.N.P.); (C.M.C.)
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI, D02 YN77 Dublin, Ireland
| |
Collapse
|
20
|
Kim S, Choi Y, Lee W, Kim K. Fabrication Parameter-Dependent Physico-Chemical Properties of Thiolated Gelatin/PEGDA Interpenetrating Network Hydrogels. Tissue Eng Regen Med 2021; 19:309-319. [PMID: 34905183 PMCID: PMC8971263 DOI: 10.1007/s13770-021-00413-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/29/2021] [Accepted: 11/14/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The development of three-dimensional hydrogels using polymeric biomaterials is a key technology for tissue engineering and regenerative medicine. Successful tissue engineering requires the control and identification of the physicochemical properties of hydrogels. METHODS Interpenetrating network (IPN) hydrogel was developed using thiolated gelatin (GSH) and poly(ethylene glycol) diacrylate (PEGDA), with the aid of ammonium persulfate (APS) and N,N,N,N'-tetramethylethylenediamine (TEMED) as radical initiators. Each component was prepared in the following concentrations, respectively: 2.5 and 5% GSH (LG and HG), 12.5 and 25% PEGDA (LP and HP), 3% APS/1.5% TEMED (LI), and 4% APS/2% TEMED (HI). IPN hydrogel was fabricated by the mixing of GSH, PEGDA, and initiators in 5:4:1 volume ratios, and incubated at 37 °C for 30 min in the following 6 experimental formulations: (1) HG-LP-LI, (2) HG-LP-HI, (3) LG-HP-LI, (4) LG-HP-HI, (5) HG-HP-HI, and (6) HG-HP-LI. Herein, the physico-chemical characteristics of IPN hydrogels, including their morphological structures, hydrolytic degradation properties, mechanical properties, embedded protein release kinetics, and biocompatibility, were investigated. RESULTS The characteristics of the hydrogel were significantly manipulated by the concentration of the polymer, especially the conversion between HP and LP, rather than the concentration of the initiator, and no hydrogel formulation exhibited any toxicity to fibroblast and HaCaT cells. CONCLUSION We provide structural-physical relationships of the hydrogels by which means their physical properties could be conveniently controlled through component control, which could be versatilely utilized for various organizational engineering strategies.
Collapse
Affiliation(s)
- Sungjun Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, 04620 Korea
| | - Yunyoung Choi
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, 04620 Korea
| | - Wonjeong Lee
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, 04620 Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul, 04620 Korea
| |
Collapse
|
21
|
Ding X, Shi J, Wei J, Li Y, Wu X, Zhang Y, Jiang X, Zhang X, Lai H. A biopolymer hydrogel electrostatically reinforced by amino-functionalized bioactive glass for accelerated bone regeneration. SCIENCE ADVANCES 2021; 7:eabj7857. [PMID: 34890238 PMCID: PMC8664252 DOI: 10.1126/sciadv.abj7857] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Composite hydrogels incorporating natural polymers and bioactive glass (BG) are promising materials for bone regeneration. However, their applications are compromised by the poor interfacial compatibility between organic and inorganic phases. In this study, we developed an electrostatically reinforced hydrogel (CAG) with improved interfacial compatibility by introducing amino-functionalized 45S5 BG to the alginate/gellan gum (AG) matrix. BAG composed of AG and unmodified BG (10 to 100 μm in size) was prepared as a control. Compared with BAG, CAG had a more uniform porous structure with a pore size of 200 μm and optimal compressive strength of 66 kPa. Furthermore, CAG promoted the M2 phenotype transition of macrophages and up-regulated the osteogenic gene expression of stem cells. The new bone formation in vivo was also accelerated due to the enhanced biomineralization of CAG. Overall, this work suggests CAG with improved interfacial compatibility is an ideal material for bone regeneration application.
Collapse
|
22
|
Zhao C, Qiu P, Li M, Liang K, Tang Z, Chen P, Zhang J, Fan S, Lin X. The spatial form periosteal-bone complex promotes bone regeneration by coordinating macrophage polarization and osteogenic-angiogenic events. Mater Today Bio 2021; 12:100142. [PMID: 34647005 PMCID: PMC8495177 DOI: 10.1016/j.mtbio.2021.100142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022] Open
Abstract
Bone defects associated with soft tissue injuries are an important cause of deformity that threatens people’s health and quality of life. Although bone substitutes have been extensively explored, effective biomaterials that can coordinate early inflammation regulation and subsequent repair events are still lacking. We prepared a spatial form periosteal bone extracellular matrix (ECM) scaffold, which has advantages in terms of low immunogenicity, good retention of bioactive ingredients, and a natural spatial structure. The periosteal bone ECM scaffold with the relatively low-stiffness periosteum (41.6 ± 3.7 kPa) could inhibit iNOS and IL-1β expression, which might be related to actin-mediated YAP translocation. It also helped to promote CD206 expression with the potential influence of proteins related to immune regulation. Moreover, the scaffold combined the excellent properties of decalcified bone and periosteum, promoted the formation of blood vessels, and good osteogenic differentiation (RUNX2, Col 1α1, ALP, OPN, and OCN), and achieved good repair of a cranial defect in rats. This scaffold, with its natural structural and biological advantages, provides a new idea for bone healing treatment that is aligned with bone physiology. We provided a spatial form periosteal-bone complex. The scaffold preserved major biological components and spatial structure. The periosteum part of the scaffold acted as a physical barrier. The scaffold participated in the transformation of the macrophage phenotype. The scaffold promoted osteogenesis and angiogenesis.
Collapse
Affiliation(s)
- C. Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - P. Qiu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - M. Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - K. Liang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Z. Tang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - P. Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - J. Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - S. Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
- Corresponding author.
| | - X. Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
- Corresponding author.
| |
Collapse
|
23
|
Sakemi Y, Hayashi K, Tsuchiya A, Nakashima Y, Ishikawa K. Reconstruction of critical-size segmental defects in rat femurs using carbonate apatite honeycomb scaffolds. J Biomed Mater Res A 2021; 109:1613-1622. [PMID: 33644971 DOI: 10.1002/jbm.a.37157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/01/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022]
Abstract
Critical-size segmental defects are formidable challenges in orthopedic surgery. Various scaffolds have been developed to facilitate bone reconstruction within such defects. Many previously studied scaffolds achieved effective outcomes with a combination of high cost, high-risk growth factors or stem cells. Herein, we developed honeycomb scaffolds (HCSs) comprising carbonate apatite (CO3 Ap) containing 8% carbonate, identical to human bone composition. The CO3 Ap HCSs were white-columned blocks harboring regularly arranged macropore channels of a size and wall thickness of 156 ± 5 μm and 102 ± 10 μm, respectively. The compressive strengths of the HCSs parallel and perpendicular to the macropore channel direction were 51.0 ± 11.8 and 15.6 ± 2.2 MPa, respectively. The HCSs were grafted into critical-sized segmental defects in rat femurs. The HCSs bore high-load stresses without any observed breakage. Two-weeks post-implantation, calluses formed around the HCSs and immature bone formed in the HCS interior. The calluses and immature bone matured until 8 weeks via endochondral ossification. At 12 weeks post-implantation, large parts of the HCSs were gradually replaced by newly formed bone. The bone reconstruction efficacy of the CO3 Ap HCSs alone was comparable to that of protein and cell scaffolds, while achieving a lower cost and increased safety.
Collapse
Affiliation(s)
- Yuta Sakemi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichiro Hayashi
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Akira Tsuchiya
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kunio Ishikawa
- Department of Biomaterials, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
24
|
Kim H, Kumbar SG, Nukavarapu SP. Biomaterial-directed cell behavior for tissue engineering. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 17:100260. [PMID: 33521410 PMCID: PMC7839921 DOI: 10.1016/j.cobme.2020.100260] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful tissue regeneration strategies focus on the use of novel biomaterials, structures, and a variety of cues to control cell behavior and promote regeneration. Studies discovered how biomaterial/ structure cues in the form of biomaterial chemistry, material stiffness, surface topography, pore, and degradation properties play an important role in controlling cellular events in the contest of in vitro and in vivo tissue regeneration. Advanced biomaterials structures and strategies are developed to focus on the delivery of bioactive factors, such as proteins, peptides, and even small molecules to influence cell behavior and regeneration. The present article is an effort to summarize important findings and further discuss biomaterial strategies to influence and control cell behavior directly via physical and chemical cues. This article also touches on various modern methods in biomaterials processing to include bioactive factors as signaling cues to program cell behavior for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Hyun Kim
- Biomedical Engineering, University of Connecticut, Storrs-06269
| | - Sangamesh G. Kumbar
- Biomedical Engineering, University of Connecticut, Storrs-06269
- Materials Science & Engineering, University of Connecticut, Storrs-06269
- Orthopaedic Surgery, University of Connecticut Health, Farmington-06030
| | - Syam P. Nukavarapu
- Biomedical Engineering, University of Connecticut, Storrs-06269
- Materials Science & Engineering, University of Connecticut, Storrs-06269
- Orthopaedic Surgery, University of Connecticut Health, Farmington-06030
| |
Collapse
|
25
|
Liang Q, Du L, Zhang R, Kang W, Ge S. Stromal cell-derived factor-1/Exendin-4 cotherapy facilitates the proliferation, migration and osteogenic differentiation of human periodontal ligament stem cells in vitro and promotes periodontal bone regeneration in vivo. Cell Prolif 2021; 54:e12997. [PMID: 33511708 PMCID: PMC7941242 DOI: 10.1111/cpr.12997] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 12/12/2022] Open
Abstract
Objectives Stromal cell‐derived factor‐1 (SDF‐1) actively directs endogenous cell homing. Exendin‐4 (EX‐4) promotes stem cell osteogenic differentiation. Studies revealed that EX‐4 strengthened SDF‐1‐mediated stem cell migration. However, the effects of SDF‐1 and EX‐4 on periodontal ligament stem cells (PDLSCs) and bone regeneration have not been investigated. In this study, we aimed to evaluate the effects of SDF‐1/EX‐4 cotherapy on PDLSCs in vitro and periodontal bone regeneration in vivo. Methods Cell‐counting kit‐8 (CCK8), transwell assay, qRT‐PCR and western blot were used to determine the effects and mechanism of SDF‐1/EX‐4 cotherapy on PDLSCs in vitro. A rat periodontal bone defect model was developed to evaluate the effects of topical application of SDF‐1 and systemic injection of EX‐4 on endogenous cell recruitment, osteoclastogenesis and bone regeneration in vivo. Results SDF‐1/EX‐4 cotherapy had additive effects on PDLSC proliferation, migration, alkaline phosphatase (ALP) activity, mineral deposition and osteogenesis‐related gene expression compared to SDF‐1 or EX‐4 in vitro. Pretreatment with ERK inhibitor U0126 blocked SDF‐1/EX‐4 cotherapy induced ERK signal activation and PDLSC proliferation. SDF‐1/EX‐4 cotherapy significantly promoted new bone formation, recruited more CXCR4+ cells and CD90+/CD34‐ stromal cells to the defects, enhanced early‐stage osteoclastogenesis and osteogenesis‐related markers expression in regenerated bone compared to control, SDF‐1 or EX‐4 in vivo. Conclusions SDF‐1/EX‐4 cotherapy synergistically regulated PDLSC activities, promoted periodontal bone formation, thereby providing a new strategy for periodontal bone regeneration.
Collapse
Affiliation(s)
- Qianyu Liang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan Shandong, China
| | - Lingqian Du
- Department of Stomatology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan Shandong, China
| | - Rui Zhang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan Shandong, China.,Department of Endodontics, Hospital of stomatology, Zunyi Medical University, Zunyi Guizhou, China
| | - Wenyan Kang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan Shandong, China
| | - Shaohua Ge
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan Shandong, China
| |
Collapse
|
26
|
iRoot SP Promotes Osteo/Odontogenesis of Bone Marrow Mesenchymal Stem Cells via Activation of NF- κB and MAPK Signaling Pathways. Stem Cells Int 2021; 2020:6673467. [PMID: 33424977 PMCID: PMC7775135 DOI: 10.1155/2020/6673467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
The regeneration of bone and tooth tissues, and related cellular therapies, has attracted widespread attention. Bone marrow mesenchymal stem cells (BMSCs) are potential candidates for such regeneration. iRoot SP is a premixed bioceramic root canal sealer widely used in clinical settings. However, the effect of iRoot SP on the biological features of BMSCs has not been elucidated. In the present study, we found that 0.2 mg/ml iRoot SP conditioned medium promoted osteo/odontogenic differentiation and enhanced mineralization of BMSCs without affecting the proliferative ability. Mechanistically, the NF-κB and MAPK signaling pathways were activated in SP-treated BMSCs, and differentiation was inhibited when cultured with the specific inhibitor. Taken together, these findings demonstrate that iRoot SP promotes osteo/odontogenic differentiation of BMSCs via the NF-κB and MAPK signaling pathways, which could provide a new theoretical basis for clinical applications of iRoot SP and a new therapeutic target for the regeneration of bone and tooth tissue in the future.
Collapse
|
27
|
Li Q, Zhang H, Pan J, Teng B, Zeng Z, Chen Y, Hei Y, Zhang S, Wei S, Sun Y. Tripeptide-based macroporous hydrogel improves the osteogenic microenvironment of stem cells. J Mater Chem B 2021; 9:6056-6067. [PMID: 34278393 DOI: 10.1039/d1tb01175h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Due to the ability to combine multiple osteogenic induction "cues" at the same time, hydrogels are widely used in the three-dimensional (3D) culture of human mesenchymal stem cells (hMSCs) and osteoinduction. However, the survival and proliferation of stem cells in a 3D culture system are limited, which reduces their osteogenic differentiation efficiency. In addition, the cells inside the hydrogel are prone to apoptosis due to hypoxia, which is a serious challenge for tissue engineering based on stem cells. In this study, a tripeptide-based macroporous alginate hydrogel was prepared to improve the osteogenic microenvironment of stem cells. The arginine-glycine-aspartate (RGD) peptide promoted the adhesion and proliferation of stem cells, and the degradation of gelatin microspheres (GMs) produced a macroporous structure to enhance further the migration and aggregation of stem cells. Mesoporous silica nanoparticles (MSNs) sustained-release bone-forming peptide-1 (BFP-1) induced osteogenic differentiation, and the sustained release of the QK peptide from the GMs promoted angiogenesis. In vitro experiments have shown that this functionalized hydrogel stimulates the proliferation of hMSCs, encourages larger cell cluster formation, and enhances the osteogenic differentiation efficiency. The released QK facilitates the proliferation and migration of endothelial cells. In vivo experiments have also verified that this system has a better osteogenic effect, and more blood vessels were observed inside the hydrogel, than in other systems. In general, this research has led to the development of a tripeptide macroporous hydrogel that can simultaneously promote osteogenesis and angiogenesis, showing great promise for applications of 3D cultures and stem cell-based tissue engineering.
Collapse
Affiliation(s)
- Qian Li
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China and Department of Oral and Maxillofacial Surgery, Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - He Zhang
- Department of Oral and Maxillofacial Surgery, Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Jijia Pan
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Binhong Teng
- Department of Oral and Maxillofacial Surgery, Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Ziqian Zeng
- Department of Oral and Maxillofacial Surgery, Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Yang Chen
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yu Hei
- College of Engineering, Peking University, Beijing 100871, China
| | - Siqi Zhang
- Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Shicheng Wei
- Laboratory of Biomaterials and Regenerative Medicine, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China and Department of Oral and Maxillofacial Surgery, Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| | - Yuhua Sun
- School of Stomatology, Xuzhou Medical University, Xuzhou 221004, China and Department of Stomatology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
28
|
Xia B, Deng Y, Lv Y, Chen G. Stem cell recruitment based on scaffold features for bone tissue engineering. Biomater Sci 2020; 9:1189-1203. [PMID: 33355545 DOI: 10.1039/d0bm01591a] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stem-cell based therapy strategies are promising approaches for the treatment of bone defects. However, extensive cell expansion steps, the low rate of cell survival and uncontrolled differentiation of stem cells transplanted into the body currently remain key challenges in advancing stem cell therapeutics. An alternative strategy is to use specifically designed bone scaffolds to recruit endogenous stem cells upon implantation and to stimulate new bone formation and remodeling. Stem cell recruitment based on scaffold features for bone tissue engineering relies on the development of scaffolds that can effectively mobilize and recruit endogenous stem cells to the implantation site. This article addresses the recent advances in the recruitment of endogenous stem cells in applications of bone scaffolds, particularly focusing on chemical modification and physical characteristic modification of the scaffold for endogenous stem cell homing and recruitment. Finally, the continuing challenges and future directions of scaffold-based stem cell recruitment are discussed.
Collapse
Affiliation(s)
- Bin Xia
- Chongqing Technology and Business University, Chongqing 400067, P. R. China
| | | | | | | |
Collapse
|
29
|
Arthur A, Gronthos S. Clinical Application of Bone Marrow Mesenchymal Stem/Stromal Cells to Repair Skeletal Tissue. Int J Mol Sci 2020; 21:E9759. [PMID: 33371306 PMCID: PMC7767389 DOI: 10.3390/ijms21249759] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
There has been an escalation in reports over the last decade examining the efficacy of bone marrow derived mesenchymal stem/stromal cells (BMSC) in bone tissue engineering and regenerative medicine-based applications. The multipotent differentiation potential, myelosupportive capacity, anti-inflammatory and immune-modulatory properties of BMSC underpins their versatile nature as therapeutic agents. This review addresses the current limitations and challenges of exogenous autologous and allogeneic BMSC based regenerative skeletal therapies in combination with bioactive molecules, cellular derivatives, genetic manipulation, biocompatible hydrogels, solid and composite scaffolds. The review highlights the current approaches and recent developments in utilizing endogenous BMSC activation or exogenous BMSC for the repair of long bone and vertebrae fractures due to osteoporosis or trauma. Current advances employing BMSC based therapies for bone regeneration of craniofacial defects is also discussed. Moreover, this review discusses the latest developments utilizing BMSC therapies in the preclinical and clinical settings, including the treatment of bone related diseases such as Osteogenesis Imperfecta.
Collapse
Affiliation(s)
- Agnieszka Arthur
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia;
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Stan Gronthos
- Mesenchymal Stem Cell Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5001, Australia;
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| |
Collapse
|
30
|
Shi Z, Xu Y, Mulatibieke R, Zhong Q, Pan X, Chen Y, Lian Q, Luo X, Shi Z, Zhu Q. Nano-Silicate-Reinforced and SDF-1α-Loaded Gelatin-Methacryloyl Hydrogel for Bone Tissue Engineering. Int J Nanomedicine 2020; 15:9337-9353. [PMID: 33262591 PMCID: PMC7699450 DOI: 10.2147/ijn.s270681] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Autologous bone grafts are the gold standard for treating bone defects. However, limited bone supply and morbidity at the donor site restrict its extensive use. Therefore, developing bone graft materials as an alternative to autologous grafts has gained considerable attention. Injectable hydrogels endowed with osteogenic potential have the ability to fill irregular bone defects using minimally invasive procedures and have thus been attracting researchers’ attention. However, from a clinical perspective, most fabrication methods employed for the current injectable osteogenic hydrogels are difficult and inconvenient. In the current study, we fabricated an injectable osteogenic hydrogel using a simple and convenient strategy. Materials and Methods Gelatin-methacryloyl (GelMA) pre-polymer was synthetized. Nano silicate (SN) and stromal cell-derived factor-1 alpha (SDF-1α) were introduced into the pre-polymer to achieve injectability, controlled release property, excellent osteogenic ability, and efficient stem cell homing. Results The GelMA-SN-SDF-1α demonstrated excellent injectability via a 17-G needle at room temperature. The loaded SDF-1α exhibited a long-term controlled release pattern and efficiently stimulated MSC migration and homing. The GelMA-SN-SDF-1α hydrogel amplified cell spreading, migration, osteogenic-related biomarker expression, and matrix mineralization. The GelMA-SN-SDF-1α hydrogel filled critical-sized calvaria defects in rats and demonstrated excellent bone regeneration ability, as assessed using micro-CT scanning and histomorphometric staining. Conclusion The GelMA-SN-SDF-1α hydrogel provides a simple and convenient strategy for the fabrication of injectable osteogenic graft materials. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/FhyefSKUa34
Collapse
Affiliation(s)
- Zhe Shi
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yichuan Xu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ruzha Mulatibieke
- Department of Plastic Surgery, The Third Affiliated Hospital of Sun Yet-Sen University, Guangzhou, People's Republic of China
| | - Qiang Zhong
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xin Pan
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yuhang Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Qiang Lian
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xin Luo
- Rehabilitation Medical School, Guangzhou International Economics College, Guangzhou, People's Republic of China
| | - Zhanjun Shi
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Qingan Zhu
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
31
|
Fadera S, Cheng NC, Young TH, Lee IC. In vitro study of SDF-1α-loaded injectable and thermally responsive hydrogels for adipose stem cell therapy by SDF-1/CXCR4 axis. J Mater Chem B 2020; 8:10360-10372. [PMID: 33108417 DOI: 10.1039/d0tb01961e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Stem cell-based approaches have become a promising therapeutic strategy for treating ischemic diseases. The aim of this study was to develop injectable hydrogel systems for the local release of stromal cell-derived factor-1α (SDF-1α) to recruit adipose stem cells (ASCs) that express CXCR4 to achieve stem cell therapy and therapeutic angiogenesis. Thermoresponsive and injectable chitosan (CS)/β-glycerophosphate disodium salt pentahydrate (βGP) hydrogels with different concentrations of hyaluronic acid (HA) were designed and fabricated to achieve local and sustained release of SDF-1α for ASC recruitment. Herein, the material structures, physical properties, gelation temperature, and gelation time of hydrogels with different compositions were determined. The incorporation of 0.9% HA in CS-based hydrogels not only enhanced the gelation time but also increased the strength of the hydrogels. In addition, the results revealed that the thermoresponsive and injectable CS/βGP/HA hydrogels showed good biocompatibility. In addition, the in vitro release profiles showed that the hydrogels achieved sustained release of SDF-1α over 7 days and enhanced ASC migration. The results revealed that the hydrogels with HA enhanced the sustained release effect compared with the hydrogel without HA, indicating that the HA content regulated the physical and release properties of the injectable hydrogels. Therefore, thermoresponsive and injectable CS/βGP/HA hydrogels may provide an alternative for treating ischemic diseases via SDF-1/CXCR4 axis for ASC recruitment and retention.
Collapse
Affiliation(s)
- Siaka Fadera
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Nai-Chen Cheng
- Department of Surgery, National Taiwan University Hospital and College of Medicine, 7 Chung-Shan S Rd, Taipei 100, Taiwan
| | - Tai-Horng Young
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, 1 Jen-Ai Rd, Taipei 100, Taiwan.
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
32
|
Lueckgen A, Garske DS, Ellinghaus A, Mooney DJ, Duda GN, Cipitria A. Dual alginate crosslinking for local patterning of biophysical and biochemical properties. Acta Biomater 2020; 115:185-196. [PMID: 32736118 DOI: 10.1016/j.actbio.2020.07.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 01/10/2023]
Abstract
Hydrogels with patterned biophysical and biochemical properties have found increasing attention in the biomaterials community. In this work, we explore alginate-based materials with two orthogonal crosslinking mechanisms: the spontaneous Diels-Alder reaction and the ultraviolet light-initiated thiol-ene reaction. Combining these mechanisms in one material and spatially restricting the location of the latter using photomasks, enables the formation of dual-crosslinked hydrogels with patterns in stiffness, biomolecule presentation and degradation, granting local control over cell behavior. Patterns in stiffness are characterized morphologically by confocal microscopy and mechanically by uniaxial compression and microindentation measurement. Mouse embryonic fibroblasts seeded on stiffness-patterned substrates attach preferably and attain a spread morphology on stiff compared to soft regions. Human mesenchymal stem cells demonstrate preferential adipogenic differentiation on soft surfaces and osteogenic differentiation on stiff surfaces. Patterns in biomolecule presentation reveal favored attachment of mouse pre-osteoblasts on stripe regions, where thiolated cell-adhesive biomolecules have been coupled. Patterns in degradation are visualized by microindentation measurement following collagenase exposure. Patterned tissue infiltration into degradable regions on the surface is discernible in n=5/12 samples, when these materials are implanted subcutaneously into the backs of mice. Taken together, these results demonstrate that our hydrogel system with patterns in biophysical and biochemical properties enables the study of how environmental cues affect multiple cell behaviors in vitro and could be applied to guide endogenous tissue growth in diverse healing scenarios in vivo. STATEMENT OF SIGNIFICANCE: Hydrogels with patterns in biophysical and biochemical properties have been explored in the biomaterials community in order to spatially control or guide cell behavior. In our alginate-based system, we demonstrate the effect of local substrate stiffness and biomolecule presentation on the in vitro cell attachment, morphology, migration and differentiation behavior of two different mouse cell lines and human primary cells. Additionally, the effect of degradation patterns on the in vivo tissue infiltration is analyzed following subcutaneous implantation into a mouse model. The achievement of patterned tissue infiltration following the hydrogel template represents an important step towards guiding endogenous healing responses, thus inviting application in various tissue engineering contexts.
Collapse
|
33
|
Brennan MÁ, Layrolle P, Mooney DJ. Biomaterials functionalized with MSC secreted extracellular vesicles and soluble factors for tissue regeneration. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909125. [PMID: 32952493 PMCID: PMC7494127 DOI: 10.1002/adfm.201909125] [Citation(s) in RCA: 198] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Indexed: 05/05/2023]
Abstract
The therapeutic benefits of mesenchymal stromal cell (MSC) transplantation have been attributed to their secreted factors, including extracellular vesicles (EVs) and soluble factors. The potential of employing the MSC secretome as an alternative acellular approach to cell therapy is being investigated in various tissue injury indications, but EVs administered via bolus injections are rapidly sequestered and cleared. However, biomaterials offer delivery platforms to enhance EV retention rates and healing efficacy. In this review, we highlight the mechanisms underpinning the therapeutic effects of MSC-EVs and soluble factors as effectors of immunomodulation and tissue regeneration, conferred primarily via their nucleic acid and protein contents. We discuss how manipulating the cell culture microenvironment or genetic modification of MSCs can further augment the potency of their secretions. The most recent advances in the development of EV-functionalized biomaterials that mediate enhanced angiogenesis and cell survival, while attenuating inflammation and fibrosis, are presented. Finally, some technical challenges to be considered for the clinical translation of biomaterials carrying MSC-secreted bioactive cargo are discussed.
Collapse
Affiliation(s)
- Meadhbh Á Brennan
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Pierre Layrolle
- INSERM, UMR 1238, PHY-OS, Bone sarcomas and remodeling of calcified tissues, Faculty of Medicine, University of Nantes, Nantes, France
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
34
|
Photocrosslinkable nanocomposite ink for printing strong, biodegradable and bioactive bone graft. Biomaterials 2020; 263:120378. [PMID: 32932140 DOI: 10.1016/j.biomaterials.2020.120378] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 09/07/2020] [Indexed: 01/29/2023]
Abstract
3D printing is known as a cost-effective technique that shows huge potential in fabrication of graft substitutes for bone tissue regeneration. However, the tradeoff between 3D printability, mechanical strength and bioactivity of the printed materials (i.e., inks) remains a challenge. In this work, we present a novel photocrosslinkable nanocomposite ink composed of tri-block poly (lactide-co-propylene glycol-co-lactide) dimethacrylate (PmLnDMA, m and n respectively represent the unit length of propylene glycol and lactide) and hydroxyethyl methacrylate (HEMA)-functionalized hydroxyapatite nanoparticles (nHAMA). The reactive HEMA-conjugated nHAMA, is designed to covalently crosslink with the surrounding polymer matrix to further increase the interfacial bonding between them. We find that the nHAMA can rapidly interact with PmLnDMA upon light exposure within 140 s and form an inorganic-organic co-crosslinked nanocomposite network, further enhancing the nanofiller-matrix interfacial compatibility. Notably, our nanocomposites possess significantly improved mechanical performances compared to the polymer, with compressive modulus increasing by nearly 10 times (from ⁓40 to ⁓400 MPa). Moreover, thanks to the low exothermic heat generation (<37 °C) during photocrosslinking, our nanocomposite ink enables facile encapsulation and long-term release of heat-labile biomolecules like bone morphogenic protein-2 (BMP-2). Furthermore, it demonstrates a readily tunable rheological property, wettability, degradation, and printability as a 3D bone scaffold. Together with its superior osteogenic ability both in vitro and in vivo, we envision that our nanocomposite ink holds great promise in 3D printing of bone grafts.
Collapse
|
35
|
Zheng X, Liu Y, Liu Y, Pan Y, Yao Q. Novel three-dimensional bioglass functionalized gelatin nanofibrous scaffolds for bone regeneration. J Biomed Mater Res B Appl Biomater 2020; 109:517-526. [PMID: 32864862 DOI: 10.1002/jbm.b.34720] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/17/2022]
Abstract
The clinical use of FDA-approved bone morphogenetic proteins (BMPs) are impeded by high costs, super-high dosage requirement, short half-life, and other undesirable side effects. Therefore, designing a biomaterial that can promote new bone formation without using exogenous BMPs is highly desirable in clinical applications. In the present work, a new kind of nanofibrous scaffold composed of gelatin and 45S5 bioglass (GF/45S5 BG) was prepared through thermally induced phase separation method together with the particle leach technique (TIPS&P). In addition to the significantly higher mechanical strength, the composite scaffolds (GF/45S5 BG) significantly increased osteogenic differentiation of human mesenchymal stem cells (hMSCs) in vitro compared with the neat scaffold (GF) without adding other biological agents, for example, BMPs or hormones. Most importantly, our in vivo studies also indicated that GF/45S5 BG scaffolds could directly promote ectopic bone regeneration in SD rats without exogenous BMP2. In summary, both in vitro and in vivo results indicated that the novel 45S5 bioglass functionalized GF nanofibrous scaffold is a promising alternative for bone tissue engineering.
Collapse
Affiliation(s)
- Xiao Zheng
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Liu
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yangxi Liu
- Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Evanston, Illinois, USA.,Department of Surgery, Feinberg School of Medicine, Northwestern University, Evanston, Illinois, USA
| | - Yining Pan
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingqing Yao
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Advanced Materials for Nano-Bio Applications, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
36
|
Garske DS, Schmidt-Bleek K, Ellinghaus A, Dienelt A, Gu L, Mooney DJ, Duda GN, Cipitria A. Alginate Hydrogels for In Vivo Bone Regeneration: The Immune Competence of the Animal Model Matters. Tissue Eng Part A 2020; 26:852-862. [DOI: 10.1089/ten.tea.2019.0310] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Daniela S. Garske
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Anke Dienelt
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Luo Gu
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- Department of Materials Science and Engineering, Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - David J. Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Georg N. Duda
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Amaia Cipitria
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
37
|
Ponte F, Kim HN, Iyer S, Han L, Almeida M, Manolagas SC. Cxcl12 Deletion in Mesenchymal Cells Increases Bone Turnover and Attenuates the Loss of Cortical Bone Caused by Estrogen Deficiency in Mice. J Bone Miner Res 2020; 35:1441-1451. [PMID: 32154948 PMCID: PMC7725417 DOI: 10.1002/jbmr.4002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 12/16/2022]
Abstract
CXCL12 is abundantly expressed in reticular cells associated with the perivascular niches of the bone marrow (BM) and is indispensable for B lymphopoiesis. Cxcl12 promotes osteoclastogenesis and has been implicated in pathologic bone resorption. We had shown earlier that estrogen receptor α deletion in osteoprogenitors and estrogen deficiency in mice increase Cxcl12 mRNA and protein levels in the BM plasma, respectively. We have now generated female and male mice with conditional deletion of a Cxcl12 allele in Prrx1 targeted cells (Cxcl12∆Prrx1 ) and show herein that they have a 90% decrease in B lymphocytes but increased erythrocytes and adipocytes in the marrow. Ovariectomy increased the expression of Cxcl12 and B-cell number in the Cxcl12f/f control mice, but these effects were abrogated in the Cxcl12∆Prrx1 mice. Cortical bone mass was not affected in Cxcl12∆Prrx1 mice. Albeit, the cortical bone loss caused by ovariectomy was greatly attenuated. Most unexpectedly, the rate of bone turnover in sex steroid-sufficient female or male Cxcl12∆Prrx1 mice was dramatically increased, as evidenced by a more than twofold increase in several osteoblast- and osteoclast-specific mRNAs, as well as increased mineral apposition and bone formation rate and increased osteoclast number in the endosteal surface. The magnitude of the Cxcl12∆Prrx1 -induced changes were much greater than those caused by ovariectomy or orchidectomy in the Cxcl12f/f mice. These results strengthen the evidence that CXCL12 contributes to the loss of cortical bone mass caused by estrogen deficiency. Moreover, they reveal for the first time that in addition to its effects on hematopoiesis, CXCL12 restrains bone turnover-without changing the balance between resorption and formation-by suppressing osteoblastogenesis and the osteoclastogenesis support provided by cells of the osteoblast lineage. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Filipa Ponte
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ha-Neui Kim
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Srividhya Iyer
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Li Han
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,The Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Stavros C Manolagas
- Division of Endocrinology and Metabolism, Center for Osteoporosis and Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,The Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| |
Collapse
|
38
|
Liu Q, Wen Y, Qiu J, Zhang Z, Jin Z, Cao M, Jiao Y, Yang H. Local SDF-1α application enhances the therapeutic efficacy of BMSCs transplantation in osteoporotic bone healing. Heliyon 2020; 6:e04347. [PMID: 32637715 PMCID: PMC7330617 DOI: 10.1016/j.heliyon.2020.e04347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/22/2020] [Accepted: 06/25/2020] [Indexed: 02/08/2023] Open
Abstract
Bone defect healing is markedly impaired in osteoporotic patient due to poor bone regeneration ability. Stromal cell derived factor-1α (SDF-1α) plays a pivotal role in the repair of various injured tissues including bone. Here, we definite that SDF-1α hydrogels potentiates in vivo osteogenesis of bone marrow-derived stromal stem cells (BMSCs) in osteoporosis. The characteristics of rat primary BMSCs including superficial markers by flow cytometry and multi-lineage differentiation by induction were determined. At different time intervals, the release media from the SDF-1α-releasing hydrogels were collected to identificate SDF-1α exhibited a sustained release profile and maintained its bioactivity after release from the hydrogels to stimulate chemotaxis of BMSCs in a time dependent manner. Bilateral alveolar defects were operated in ovariectomized (OVX) rats and repaired with systemic BMSCs transplantation with or without the hydrogels. Local administration of SDF-1α significantly enhanced BMSCs recruitment and promoted more bone regeneration as well as the expression of OCN and Runx2 compared with the effect of BMSCs transplantation alone. Moreover, after BMSCs transplantation with SDF-1α delivery, macrophage polarization was promoted toward the M2 phenotype, that is identified as an important symbol in tissue regeneration process. Taken together, local SDF-1α application enhances the efficacy of BMSCs transplantation therapy in osteoporotic bone healing, suggesting clinical potential of SDF-1α to serve as a therapeutic drug target for osteoporosis treatment.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease & Shaanxi Key Laboratory of Oral Disease, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yi Wen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease & Shaanxi Key Laboratory of Oral Disease, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jun Qiu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Zhaoyichun Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Zuolin Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease & Shaanxi Key Laboratory of Oral Disease, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Meng Cao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease & Shaanxi Key Laboratory of Oral Disease, Department of Orthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yang Jiao
- Department of Stomatology, The 7th Medical Center of PLA General Hospital, Beijing, China
| | - Hongxu Yang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
39
|
Zhang Y, Yu T, Peng L, Sun Q, Wei Y, Han B. Advancements in Hydrogel-Based Drug Sustained Release Systems for Bone Tissue Engineering. Front Pharmacol 2020; 11:622. [PMID: 32435200 PMCID: PMC7218105 DOI: 10.3389/fphar.2020.00622] [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: 03/12/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Bone defects caused by injury, disease, or congenital deformity remain a major health concern, and efficiently regenerating bone is a prominent clinical demand worldwide. However, bone regeneration is an intricate process that requires concerted participation of both cells and bioactive factors. Mimicking physiological bone healing procedures, the sustained release of bioactive molecules plays a vital role in creating an optimal osteogenic microenvironment and achieving promising bone repair outcomes. The utilization of biomaterial scaffolds can positively affect the osteogenesis process by integrating cells with bioactive factors in a proper way. A high water content, tunable physio-mechanical properties, and diverse synthetic strategies make hydrogels ideal cell carriers and controlled drug release reservoirs. Herein, we reviewed the current advancements in hydrogel-based drug sustained release systems that have delivered osteogenesis-inducing peptides, nucleic acids, and other bioactive molecules in bone tissue engineering (BTE).
Collapse
Affiliation(s)
- Yunfan Zhang
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Tingting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Liying Peng
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Qiannan Sun
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, China
| | - Bing Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| |
Collapse
|
40
|
Zhong J, Yang Y, Liao L, Zhang C. Matrix stiffness-regulated cellular functions under different dimensionalities. Biomater Sci 2020; 8:2734-2755. [DOI: 10.1039/c9bm01809c] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The microenvironments that cells encounter with in vitro.
Collapse
Affiliation(s)
- Jiajun Zhong
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Yuexiong Yang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| | - Liqiong Liao
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering
- Biomaterials Research Center
- School of Biomedical Engineering
- Southern Medical University
- Guangzhou
| | - Chao Zhang
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instruments (Sun Yat-sen University)
- School of Biomedical Engineering
- Sun Yat-Sen University
- Guangzhou
- P. R. China
| |
Collapse
|
41
|
Yu X, Sun H, Yang J, Liu Y, Zhang Z, Wang J, Deng F. Evaluation of bone-regeneration effects and ectopic osteogenesis of collagen membrane chemically conjugated with stromal cell-derived factor-1 in vivo. ACTA ACUST UNITED AC 2019; 15:015009. [PMID: 31665702 DOI: 10.1088/1748-605x/ab52da] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Because the collagen membrane lacks osteoinductivity, it must be modified with bioactive components to trigger rapid bone regeneration. In this study, we aimed to evaluate the bone regeneration effects of a collagen membrane chemically conjugated with stromal cell-derived factor-1 alpha (SDF-1α) in rat models. To this end, different collagen membranes from four groups including a control group with a Bio-Oss bone substitute + collagen membrane; physical adsorption group with Bio-Oss + SDF-1α physically adsorbed on the collagen membrane; chemical cross-linking group with Bio-Oss + SDF-1α chemically cross-linked to the collagen membrane; and cell-seeding group with Bio-Oss + bone marrow mesenchymal stem cells (BMSCs) seeded onto the collagen membrane were placed in critical-sized defect models using a guided bone regeneration technique. At 4 and 8 weeks, the specimens were analyzed by scanning electron microscopy, energy-dispersive x-ray spectroscopy, micro-computed tomography, and histomorphology analyzes. Furthermore, ectopic osteogenesis was examined by histological analysis with Von Kossa staining, with the samples counterstained by hematoxylin and eosin and immunohistochemical staining. The results showed that in the chemical cross-linking group and cell-seeding group, the bone volume fraction, bone surface area fraction, and trabecular number were significantly increased and showed more new bone formation compared to the control and physical adsorption groups. Von Kossa-stained samples counterstained with hematoxylin and eosin and subjected to immunohistochemical staining of 4-week implanted membranes revealed that the chemical cross-linking group had the largest number of microvessels. The collagen membrane chemically conjugated with SDF-1α to significantly promote new bone and microvessel formation compared to SDF-1α physical adsorption and showed similar effects on new bone formation as a BMSC seeding method. This study provided a cell-free approach for shortening the bone healing time and improving the success rate of guided bone regeneration.
Collapse
Affiliation(s)
- Xiaolin Yu
- Department of Oral Implantology, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
42
|
Abdulghani S, Mitchell GR. Biomaterials for In Situ Tissue Regeneration: A Review. Biomolecules 2019; 9:E750. [PMID: 31752393 PMCID: PMC6920773 DOI: 10.3390/biom9110750] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/15/2019] [Accepted: 11/17/2019] [Indexed: 12/11/2022] Open
Abstract
This review focuses on a somewhat unexplored strand of regenerative medicine, that is in situ tissue engineering. In this approach manufactured scaffolds are implanted in the injured region for regeneration within the patient. The scaffold is designed to attract cells to the required volume of regeneration to subsequently proliferate, differentiate, and as a consequence develop tissue within the scaffold which in time will degrade leaving just the regenerated tissue. This review highlights the wealth of information available from studies of ex-situ tissue engineering about the selection of materials for scaffolds. It is clear that there are great opportunities for the use of additive manufacturing to prepare complex personalized scaffolds and we speculate that by building on this knowledge and technology, the development of in situ tissue engineering could rapidly increase. Ex-situ tissue engineering is handicapped by the need to develop the tissue in a bioreactor where the conditions, however optimized, may not be optimum for accelerated growth and maintenance of the cell function. We identify that in both methodologies the prospect of tissue regeneration has created much promise but delivered little outside the scope of laboratory-based experiments. We propose that the design of the scaffolds and the materials selected remain at the heart of developments in this field and there is a clear need for predictive modelling which can be used in the design and optimization of materials and scaffolds.
Collapse
Affiliation(s)
- Saba Abdulghani
- Centre for Rapid and Sustainable Product Development, Polytechnic of Leiria, 2430-080 Marinha Grande, Portugal;
| | | |
Collapse
|
43
|
Abstract
This review focuses on a somewhat unexplored strand of regenerative medicine, that is in situ tissue engineering. In this approach manufactured scaffolds are implanted in the injured region for regeneration within the patient. The scaffold is designed to attract cells to the required volume of regeneration to subsequently proliferate, differentiate, and as a consequence develop tissue within the scaffold which in time will degrade leaving just the regenerated tissue. This review highlights the wealth of information available from studies of ex-situ tissue engineering about the selection of materials for scaffolds. It is clear that there are great opportunities for the use of additive manufacturing to prepare complex personalized scaffolds and we speculate that by building on this knowledge and technology, the development of in situ tissue engineering could rapidly increase. Ex-situ tissue engineering is handicapped by the need to develop the tissue in a bioreactor where the conditions, however optimized, may not be optimum for accelerated growth and maintenance of the cell function. We identify that in both methodologies the prospect of tissue regeneration has created much promise but delivered little outside the scope of laboratory-based experiments. We propose that the design of the scaffolds and the materials selected remain at the heart of developments in this field and there is a clear need for predictive modelling which can be used in the design and optimization of materials and scaffolds.
Collapse
|
44
|
Xiang G, Lippens E, Hafeez S, Duda GN, Geissler S, Qazi TH. Oxidized alginate beads for tunable release of osteogenically potent mesenchymal stromal cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109911. [DOI: 10.1016/j.msec.2019.109911] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/26/2019] [Accepted: 06/21/2019] [Indexed: 12/31/2022]
|
45
|
Abstract
Nanoparticulate materials displaying enzyme-like properties, so-called nanozymes, are explored as substitutes for natural enzymes in several industrial, energy-related, and biomedical applications. Outstanding high stability, enhanced catalytic activities, low cost, and availability at industrial scale are some of the fascinating features of nanozymes. Furthermore, nanozymes can also be equipped with the unique attributes of nanomaterials such as magnetic or optical properties. Due to the impressive development of nanozymes during the last decade, their potential in the context of tissue engineering and regenerative medicine also started to be explored. To highlight the progress, in this review, we discuss the two most representative nanozymes, namely, cerium- and iron-oxide nanomaterials, since they are the most widely studied. Special focus is placed on their applications ranging from cardioprotection to therapeutic angiogenesis, bone tissue engineering, and wound healing. Finally, current challenges and future directions are discussed.
Collapse
|
46
|
Huebsch N. Translational mechanobiology: Designing synthetic hydrogel matrices for improved in vitro models and cell-based therapies. Acta Biomater 2019; 94:97-111. [PMID: 31129361 DOI: 10.1016/j.actbio.2019.05.055] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 12/27/2022]
Abstract
Synthetic hydrogels have ideal physiochemical properties to serve as reductionist mimics of the extracellular matrix (ECM) for studies on cellular mechanosensing. These studies range from basic observation of correlations between ECM mechanics and cell fate changes to molecular dissection of the underlying mechanisms. Despite intensive work on hydrogels to study mechanobiology, many fundamental questions regarding mechanosensing remain unanswered. In this review, I first discuss historical motivation for studying cellular mechanobiology, and challenges impeding this effort. I next overview recent efforts to engineer hydrogel properties to study cellular mechanosensing. Finally, I focus on in vitro modeling and cell-based therapies as applications of hydrogels that will exploit our ability to create micro-environments with physiologically relevant elasticity and viscoelasticity to control cell biology. These translational applications will not only use our current understanding of mechanobiology but will also bring new tools to study the fundamental problem of how cells sense their mechanical environment. STATEMENT OF SIGNIFICANCE: Hydrogels are an important tool for understanding how our cells can sense their mechanical environment, and to exploit that understanding in regenerative medicine. In the current review, I discuss historical work linking mechanics to cell behavior in vitro, and highlight the role hydrogels played in allowing us to understand how cells monitor mechanical cues. I then highlight potential translational applications of hydrogels with mechanical properties similar to those of the tissues where cells normally reside in our bodies, and discuss how these types of studies can provide clues to help us enhance our understanding of mechanosensing.
Collapse
Affiliation(s)
- Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, United States.
| |
Collapse
|
47
|
Lueckgen A, Garske DS, Ellinghaus A, Mooney DJ, Duda GN, Cipitria A. Enzymatically-degradable alginate hydrogels promote cell spreading and in vivo tissue infiltration. Biomaterials 2019; 217:119294. [PMID: 31276949 DOI: 10.1016/j.biomaterials.2019.119294] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023]
Abstract
Enzymatically-degradable materials recapitulate the dynamic and reciprocal interactions between cells and their native microenvironment by allowing cells to actively shape the degradation process. In order to engineer a synthetic 3D environment enabling cells to orchestrate the degradation of the surrounding material, norbornene-modified alginate was crosslinked with two different peptide crosslinkers susceptible to cleavage by matrix metalloproteinases using UV-initiated thiol-ene chemistry. Resulting hydrogels were characterized for their initial mechanical and rheological properties, and their degradation behavior was measured by tracking changes in wet weight upon enzyme incubation. This process was found to be a function of the crosslinker type and enzyme concentration, indicating that degradation kinetics could be controlled and tuned. When mouse embryonic fibroblasts were encapsulated in 3D, cell number remained constant and viability was high in all materials, while cell spreading and extensive filopodia formation was observed only in the degradable gels, not in non-degradable controls. After implanting hydrogels into the backs of C57/Bl6 mice for 8 weeks, histological stainings of recovered gel remnants and surrounding tissue revealed higher tissue and cell infiltration into degradable materials compared to non-degradable controls. This alginate-based material platform with cell-empowered enzymatic degradation could prove useful in diverse tissue engineering contexts, such as regeneration and drug delivery.
Collapse
Affiliation(s)
- Aline Lueckgen
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Daniela S Garske
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, 14476 Potsdam, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - David J Mooney
- School of Engineering and Applied Sciences - Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Georg N Duda
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Amaia Cipitria
- Julius Wolff Institute & Center for Musculoskeletal Surgery, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany; Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, 14476 Potsdam, Germany; Berlin-Brandenburg Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, 13353 Berlin, Germany.
| |
Collapse
|
48
|
He XT, Li X, Xia Y, Yin Y, Wu RX, Sun HH, Chen FM. Building capacity for macrophage modulation and stem cell recruitment in high-stiffness hydrogels for complex periodontal regeneration: Experimental studies in vitro and in rats. Acta Biomater 2019; 88:162-180. [PMID: 30735811 DOI: 10.1016/j.actbio.2019.02.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
Recently, we found that although high-stiffness matrices stimulated osteogenic differentiation of bone marrow-derived stromal cells (BMSCs), the macrophages (Mφs) in high-stiffness transglutaminase crosslinked gelatins (TG-gels) tended to undergo M1 polarization and hence compromised cell osteogenesis. In this study, we hypothesized that the copresentation of interleukin (IL)-4 and stromal cell-derived factor (SDF)-1α in high-stiffness TG-gels may enhance periodontal regeneration by modulating Mφ polarization and promoting endogenous stem cell recruitment. We found that Mφs were more likely to polarize toward an immunomodulatory M2 state in the presence of IL-4 and hence positively influence the osteogenic differentiation of BMSCs when these cells coexisted in either indirect or direct co-culture systems. In cell migration assays, BMSCs exhibited an enhanced capability to move toward gels containing SDF-1α, and more cells could be recruited into the three-dimensional matrix of TG-gels. When TG-gels containing IL-4 and/or SDF-1α were used to repair periodontal defects, more new bone (MicroCT) was formed in animals that received the dual cytokine-loaded transplants at 4 weeks postsurgery. Mφs were recruited to all the transplanted gels, and after one week, more M1-phenotype cells were found in the groups without IL-4, while the presence of IL-4 was more likely to result in M2 polarization (immunofluorescence staining). When the tissue biopsies were histologically examined, the TG-gels containing both IL-4 and SDF-1α led to a generally satisfactory regeneration with respect to attachment recovery (epithelial and connective tissue) and hybrid tissue regeneration (bone, periodontal ligament and cementum). Our data suggest that the incorporation of IL-4 into high-stiffness TG-gels may promote the M2 polarization of Mφs and that SDF-1α can be applied to guide endogenous cell homing. Overall, building capacity for Mφ modulation and cell recruitment in high-stiffness hydrogels represents a simple and effective strategy that can support high levels of periodontal tissue regeneration. STATEMENT OF SIGNIFICANCE: The development of hydrogel-based regenerative therapies centered on the mobilization and stimulation of native cells for therapeutics opens a window toward realizing periodontal endogenous regeneration. In the present study, the parallel use of immunomodulatory and homing factors in high-stiffness hydrogel materials is shown to induce stem cell homing, modulate cell differentiation and indeed induce regrowth of the periodontium. We found that incorporation of interleukin (IL)-4 in high-stiffness TG-gels coaxed macrophages to polarize into M2 phenotypes, and stromal cell-derived factor (SDF)-1α could be applied to direct endogenous cell homing. Hence, we present for the first time a clinically relevant strategy based on macrophage modulation and host cell recruitment that can support high levels of periodontal tissue regeneration.
Collapse
Affiliation(s)
- Xiao-Tao He
- State Key Laboratory of Military Stomatology Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Xuan Li
- State Key Laboratory of Military Stomatology Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Yu Xia
- State Key Laboratory of Military Stomatology Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Rui-Xin Wu
- State Key Laboratory of Military Stomatology Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Hai-Hua Sun
- State Key Laboratory of Military Stomatology Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China; Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China.
| |
Collapse
|
49
|
Mettl3 Regulates Osteogenic Differentiation and Alternative Splicing of Vegfa in Bone Marrow Mesenchymal Stem Cells. Int J Mol Sci 2019; 20:ijms20030551. [PMID: 30696066 PMCID: PMC6387109 DOI: 10.3390/ijms20030551] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/20/2019] [Accepted: 01/25/2019] [Indexed: 12/16/2022] Open
Abstract
Bone mesenchymal stem cells (BMSCs) can be a useful cell resource for developing biological treatment strategies for bone repair and regeneration, and their therapeutic applications hinge on an understanding of their physiological characteristics. N6-methyl-adenosine (m6A) is the most prevalent internal chemical modification of mRNAs and has recently been reported to play important roles in cell lineage differentiation and development. However, little is known about the role of m6A modification in the cell differentiation of BMSCs. To address this issue, we investigated the expression of N6-adenosine methyltransferases (Mettl3 and Mettl14) and demethylases (Fto and Alkbh5) and found that Mettl3 was upregulated in BMSCs undergoing osteogenic induction. Furthermore, we knocked down Mettl3 and demonstrated that Mettl3 knockdown decreased the expression of bone formation-related genes, such as Runx2 and Osterix. The alkaline phosphatase (ALP) activity and the formation of mineralized nodules also decreased after Mettl3 knockdown. RNA sequencing analysis revealed that a vast number of genes affected by Mettl3 knockdown were associated with osteogenic differentiation and bone mineralization. Kyoto encyclopedia of genes and genomes (KEGG) pathway analysis revealed that the phosphatidylinositol 3-kinase/AKT (PI3K-Akt) signaling pathway appeared to be one of the most enriched pathways, and Western blotting results showed that Akt phosphorylation was significantly reduced after Mettl3 knockdown. Mettl3 has been reported to play an important role in regulating alternative splicing of mRNA in previous research. In this study, we found that Mettl3 knockdown not only reduced the expression of Vegfa but also decreased the level of its splice variants, vegfa-164 and vegfa-188, in Mettl3-deficient BMSCs. These findings might contribute to novel progress in understanding the role of epitranscriptomic regulation in the osteogenic differentiation of BMSCs and provide a promising perspective for new therapeutic strategies for bone regeneration.
Collapse
|
50
|
Ingavle GC, Gionet-Gonzales M, Vorwald CE, Bohannon LK, Clark K, Galuppo LD, Leach JK. Injectable mineralized microsphere-loaded composite hydrogels for bone repair in a sheep bone defect model. Biomaterials 2019; 197:119-128. [PMID: 30641263 DOI: 10.1016/j.biomaterials.2019.01.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/03/2019] [Accepted: 01/04/2019] [Indexed: 12/20/2022]
Abstract
The efficacy of cell-based therapies as an alternative to autologous bone grafts requires biomaterials to localize cells at the defect and drive osteogenic differentiation. Hydrogels are ideal cell delivery vehicles that can provide instructional cues via their composition or mechanical properties but commonly lack osteoconductive components that nucleate mineral. To address this challenge, we entrapped mesenchymal stromal cells (MSCs) in a composite hydrogel based on two naturally-derived polymers (alginate and hyaluronate) containing biomineralized polymeric microspheres. Mechanical properties of the hydrogels were dependent upon composition. The presentation of the adhesive tripeptide Arginine-Glycine-Aspartic Acid (RGD) from both polymers induced greater osteogenic differentiation of ovine MSCs in vitro compared to gels formed of RGD-alginate or RGD-alginate/hyaluronate alone. We then evaluated the capacity of this construct to stimulate bone healing when transplanting autologous, culture-expanded MSCs into a surgical induced, critical-sized ovine iliac crest bone defect. At 12 weeks post-implantation, defects treated with MSCs transplanted in composite gels exhibited significant increases in blood vessel density, osteoid formation, and bone formation compared to acellular gels or untreated defects. These findings demonstrate the capacity of osteoconductive hydrogels to promote bone formation with autologous MSCs in a large animal bone defect model and provide a promising vehicle for cell-based therapies of bone healing.
Collapse
Affiliation(s)
- Ganesh C Ingavle
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA; Symbiosis Center for Stem Cell Research, Symbiosis International University, Pune 412115, India
| | | | - Charlotte E Vorwald
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Laurie K Bohannon
- Department of Surgical & Radiological Sciences, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | - Kaitlin Clark
- Department of Surgical & Radiological Sciences, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | - Larry D Galuppo
- Department of Surgical & Radiological Sciences, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | - J Kent Leach
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA; Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA 95817, USA.
| |
Collapse
|