1
|
Liu J, Huang X, Su H, Yu J, Nie X, Liu K, Qin W, Zhao Y, Su Y, Kuang X, Chen D, Lu WW, Chen Y, Hua Q. Tibial Cortex Transverse Transport Facilitates Severe Diabetic Foot Wound Healing via HIF-1α-Induced Angiogenesis. J Inflamm Res 2024; 17:2681-2696. [PMID: 38707956 PMCID: PMC11070162 DOI: 10.2147/jir.s456590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Purpose Management of severe diabetic foot ulcers (DFUs) remains challenging. Tibial cortex transverse transport (TTT) facilitates healing and limb salvage in patients with recalcitrant DFUs. However, the underlying mechanism is largely unknown, necessitating the establishment of an animal model and mechanism exploration. Methods Severe DFUs were induced in rats, then assigned to TTT, sham, or control groups (n=16/group). The TTT group underwent a tibial corticotomy, with 6 days each of medial and lateral transport; the sham group had a corticotomy without transport. Ulcer healing was assessed through Laser Doppler, CT angiography, histology, and immunohistochemistry. Serum HIF-1α, PDGF-BB, SDF-1, and VEGF levels were measured by ELISA. Results The TTT group showed lower percentages of wound area, higher dermis thickness (all p < 0.001 expect for p = 0.001 for TTT vs Sham at day 6) and percentage of collagen content (all p < 0.001) than the other two groups. The TTT group had higher perfusion and vessel volume in the hindlimb (all p < 0.001). The number of CD31+ cells (all p < 0.001) and VEGFR2+ cells (at day 6, TTT vs Control, p = 0.001, TTT vs Sham, p = 0.006; at day 12, TTT vs Control, p = 0.003, TTT vs Sham, p = 0.01) were higher in the TTT group. The activity of HIF-1α, PDGF-BB, and SDF-1 was increased in the TTT group (all p < 0.001 except for SDF-1 at day 12, TTT vs Sham, p = 0.005). The TTT group had higher levels of HIF-1α, PDGF-BB, SDF-1, and VEGF in serum than the other groups (all p < 0.001). Conclusion TTT enhanced neovascularization and perfusion at the hindlimb and accelerated healing of the severe DFUs. The underlying mechanism is related to HIF-1α-induced angiogenesis.
Collapse
Affiliation(s)
- Jie Liu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Xiajie Huang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Hongjie Su
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Jie Yu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Xinyu Nie
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Kaibing Liu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Wencong Qin
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Yongxin Zhao
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Yongfeng Su
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Xiaocong Kuang
- Yulin Campus of Guangxi Medical University, Yulin, Guangxi, People’s Republic of China
| | - Di Chen
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China
| | - William W Lu
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yan Chen
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Qikai Hua
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| |
Collapse
|
2
|
Fu F, Li M, Yang S, Du G, Xu Y, Jiang J, Jia L, Zhang K, Li P. The effects of SDF-1 combined application with VEGF on femoral distraction osteogenesis in rats. Open Life Sci 2024; 19:20220851. [PMID: 38645752 PMCID: PMC11032098 DOI: 10.1515/biol-2022-0851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/03/2024] [Accepted: 03/08/2024] [Indexed: 04/23/2024] Open
Abstract
Bone regeneration and mineralization can be achieved by means of distraction osteogenesis (DO). In the present study, we investigated the effect of stromal cell-derived factor 1 (SDF-1) and vascular endothelial growth factor (VEGF) on the new bone formation during DO in rats. Forty-eight Sprague-Dawley rats were randomized into four groups of 12 rats each. We established the left femoral DO model in rats and performed a mid-femoral osteotomy, which was fixed with an external fixator. DO was performed at 0.25 mm/12 h after an incubation period of 5 days. Distraction was continued for 10 days, resulting in a total of 5 mm of lengthening. After distraction, the solution was locally injected into the osteotomy site, once a day 1 ml for 1 week. One group received the solvent alone and served as the control, and the other three groups were treated with SDF-1, VEGF, and SDF-1with VEGF in an aqueous. Sequential X-ray radiographs were taken two weekly. The regeneration was monitored with the use of micro-CT analysis, mechanical testing, and histology. Radiographs showed accelerated regenerate ossification in the SDF-1, VEGF, and SDF-1 with the VEGF group, with a larger amount of new bone compared with the control group, especially SDF-1 with the VEGF group. Micro-CT analysis and biomechanical tests showed Continuous injection of the SDF-1, VEGF, and SDF-1 with VEGF during the consolidation period significantly increased bone mineral density bone volume, mechanical maximum loading, and bone mineralization of the regenerate. Similarly, the expression of osteogenic-specific genes, as determined by real-time polymerase chain reaction , was significantly higher in SDF-1 with the VEGF group than in the other groups. Histological examination revealed more new trabeculae in the distraction gap and more mature bone tissue for the SDF-1 with the VEGF group. SDF-1 and VEGF promote bone regeneration and mineralization during DO, and there is a synergistic effect between the SDF-1 and VEGF. It is possible to provide a new and feasible method to shorten the period of treatment of DO.
Collapse
Affiliation(s)
- Fangang Fu
- Department of Orthopaedics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Mengqi Li
- Department of Orthopedics, Binzhou Medical University Hospital, Binzhou, 256603China
| | - Shuye Yang
- Department of Orthopedics, Binzhou Medical University Hospital, Binzhou, 256603China
| | - Gangqiang Du
- Department of Orthopedics, Binzhou Medical University Hospital, Binzhou, 256603China
| | - Yingjiang Xu
- Binzhou Medical University Hospital, Binzhou, China
| | - Jianhao Jiang
- Department of Orthopedics, Binzhou Medical University Hospital, Binzhou, 256603China
| | - Long Jia
- Department of Orthopedics, Binzhou Medical University Hospital, Binzhou, 256603China
| | - Kai Zhang
- Department of Orthopedics, Binzhou Medical University Hospital, Binzhou, 256603China
| | - Peng Li
- Department of Orthopedics, Binzhou Medical University Hospital, Binzhou, 256603China
| |
Collapse
|
3
|
Qin W, Nie X, Su H, Ding Y, He L, Liu K, Hou J, Pan K, He L, Yang S, Li L, Yang S, Peng X, Zhao J, Guan J, Kuang X, Hua Q. Efficacy and safety of unilateral tibial cortex transverse transport on bilateral diabetic foot ulcers: A propensity score matching study. J Orthop Translat 2023; 42:137-146. [PMID: 37736148 PMCID: PMC10509564 DOI: 10.1016/j.jot.2023.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/12/2023] [Accepted: 08/02/2023] [Indexed: 09/23/2023] Open
Abstract
Background Tibial Cortex Transverse Transport (TTT) has been demonstrated to be an effective treatment for unilateral diabetic foot ulcers (UDFUs). However, this retrospective study was designed to compare the efficacy and safety of unilateral TTT on bilateral diabetic foot ulcers (BDFUs). Methods This retrospective study included a review of patients with TTT treated from January 2017 to August 2019, Propensity Score Matching (PSM) was performed to compare patients with BDFUs to those with UDFUs. Ulcer healing, recurrence, and major amputation rates were evaluated at 1-year follow-up. Changes in foot vessels were assessed in the BDFUs group using computed tomography angiography (CTA). Results A total of 140 patients with DFUs (106 UDFUs and 34 BDFUs) were included in the study. UDFUs and BDFUs were matched in a 1:1 ratio (34 in each group) using PSM. No significant difference was observed at 1-year-follow-up [91.2% (31/34) vs. 76.5% (26/34), OR 0.315 (95% CI 0.08 to 1.31), P = 0.10] and 6-month-follow-up [70.6% (24/34) vs. 50.0% (17/34), OR 0.85 (95% CI 0.15 to 1.13), P = 0.08] in two groups. Significant differences in rates of major amputation and recurrence between the groups (P > 0.05) were not observed. The BDFUs group appeared more angiogenesis of the foot by CTA after 8 weeks of operation. Conclusion Results of this study suggest that severe BDFUs can be effectively treated by unilateral TTT. TTT is easy to operate and effective, which may be a good alternative for treating severe BDFUs. The translational potential of this article In previous retrospective clinical studies, TTT has demonstrated promising clinical outcomes in the management of diabetic foot ulcers. In this current study, we aim to investigate the potential use of TTT in treating distant tissue defects by evaluating the limited availability and safety of TTT for the management of bilateral diabetic foot. While additional basic and clinical research is necessary to fully elucidate the underlying mechanisms, our study offers insight into the potential therapeutic use of TTT for this condition.
Collapse
Affiliation(s)
- Wencong Qin
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xinyu Nie
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
- Department of Orthopedics, The Second Hospital, Jilin University, Changchun, Jilin, 130042, China
| | - Hongjie Su
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bio-Resource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
| | - Yi Ding
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Lihuan He
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Department of Orthopedics, Sinopharm Dongfeng General Hospital of Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Kaibing Liu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jun Hou
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bio-Resource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
| | - Kaixiang Pan
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Yulin Campus of Guangxi Medical University, Yulin, Guangxi, 537406, China
| | - Liexun He
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Sijie Yang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bio-Resource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
| | - Lisha Li
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Shenghui Yang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bio-Resource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
| | - Xiao Peng
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jinming Zhao
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bio-Resource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
| | - Jack Guan
- Bay Area Foot and Ankle Medical Clinic, San Jose, 3150, California, USA
| | - Xiaocong Kuang
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Yulin Campus of Guangxi Medical University, Yulin, Guangxi, 537406, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
| | - Qikai Hua
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
- Guangxi Diabetic Foot Salvage Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bio-Resource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Research Centre for Regenerative Medicine, Guangxi Medical University, China
| |
Collapse
|
4
|
Jin C, Xu G. Study on the Promotion of hADSCs Migration and Chemotaxis by SDF-1. Asia Pac J Ophthalmol (Phila) 2023; 12:303-309. [PMID: 37171133 DOI: 10.1097/apo.0000000000000613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/28/2023] [Indexed: 05/13/2023] Open
Abstract
PURPOSE The purpose of this study was to investigate the chemotaxis effect of stromal cell-derived factor-1 (SDF-1) on human adipose-derived stem cells (hADSCs). METHODS A lentivirus vector with the enhanced green fluorescent protein gene was constructed and transfected to hADSCs. A control group and an SDF-1 induction group were set to estimate the efficacy of SDF-1 in promoting hADSCs chemotaxis and migration. RESULTS After 7 days of infection with hADSCs by enhanced green fluorescent protein lentivirus, the positive rate of fluorescence expression detected by flow cytometry was 100%. After the addition of SDF-1 induction, the invasion ability of hADSCs was enhanced. CONCLUSIONS SDF-1 can promote hADSCs migration and chemotaxis, which may play a role in stem cell transplantation.
Collapse
Affiliation(s)
- Chen Jin
- Department of Ophthalmology, The First Affiliated Hospital, Fujian Medical University, Fujian Institute of Ophthalmology, Fuzhou, China
| | | |
Collapse
|
5
|
A Scoping Review of the Skeletal Effects of Naringenin. Nutrients 2022; 14:nu14224851. [PMID: 36432535 PMCID: PMC9699132 DOI: 10.3390/nu14224851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Osteoporosis is caused by the deterioration of bone density and microstructure, resulting in increased fracture risk. It transpires due to an imbalanced skeletal remodelling process favouring bone resorption. Various natural compounds can positively influence the skeletal remodelling process, of which naringenin is a candidate. Naringenin is an anti-inflammatory and antioxidant compound found in citrus fruits and grapefruit. This systematic review aims to present an overview of the available evidence on the skeletal protective effects of naringenin. METHOD A systematic literature search was conducted using the PubMed and Scopus databases in August 2022. Original research articles using cells, animals, or humans to investigate the bone protective effects of naringenin were included. RESULTS Sixteen eligible articles were included in this review. The existing evidence suggested that naringenin enhanced osteoblastogenesis and bone formation through BMP-2/p38MAPK/Runx2/Osx, SDF-1/CXCR4, and PI3K/Akt/c-Fos/c-Jun/AP-1 signalling pathways. Naringenin also inhibited osteoclastogenesis and bone resorption by inhibiting inflammation and the RANKL pathway. CONCLUSIONS Naringenin enhances bone formation while suppressing bone resorption, thus achieving its skeletal protective effects. It could be incorporated into the diet through fruit intake or supplements to prevent bone loss.
Collapse
|
6
|
Liu X, Sun Y, Shen J, Min HS, Xu J, Chai Y. Strontium doped mesoporous silica nanoparticles accelerate osteogenesis and angiogenesis in distraction osteogenesis by activation of Wnt pathway. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 41:102496. [PMID: 34838995 DOI: 10.1016/j.nano.2021.102496] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 12/27/2022]
Abstract
Distraction osteogenesis (DO) is a powerful method to reconstruct segmented bone defects in the extremities. However, the main shortcoming of DO is the time-consuming consolidation period. To shorten the consolidation process, two biocompatible inorganic ions, strontium and silicone, were applied to design a biocompatible material to enhance bone mineralization ability during DO. In the present study, we integrated strontium into a one-pot synthesis of mesoporous silica nanoparticles to obtain strontium-doped mesoporous silica nanoparticles characterized by a homogeneous spherical morphology and uniform ion-releasing dynamics. This dual-ion releasing osteogenic and angiogenic drug delivery system was investigated to accelerate mineralization in DO. Osteogenesis was promoted by activation of the Wnt/β-catenin pathway, while bone resorption was inhibited by reduction of the osteoclastogenic factor RANKL/OPG. In addition, angiogenesis may have been enhanced indirectly by secretion of vascular endothelial growth factor (VEGF) from bone marrow stem cells. Therefore, strontium-doped mesoporous silica nanoparticles could be a potential biomaterial candidate for accelerating consolidation during DO.
Collapse
Affiliation(s)
- Xuanzhe Liu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yi Sun
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Junjie Shen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hong Sung Min
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
7
|
Effects of local vs systemic administration of CXCR4 inhibitor AMD3100 on orthodontic tooth movement in rats. Am J Orthod Dentofacial Orthop 2022; 162:182-192. [DOI: 10.1016/j.ajodo.2021.03.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 03/01/2021] [Accepted: 03/01/2021] [Indexed: 12/16/2022]
|
8
|
Yang S, Wang N, Ma Y, Guo S, Guo S, Sun H. Immunomodulatory effects and mechanisms of distraction osteogenesis. Int J Oral Sci 2022; 14:4. [PMID: 35067679 PMCID: PMC8784536 DOI: 10.1038/s41368-021-00156-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 11/11/2022] Open
Abstract
Distraction osteogenesis (DO) is widely used for bone tissue engineering technology. Immune regulations play important roles in the process of DO like other bone regeneration mechanisms. Compared with others, the immune regulation processes of DO have their distinct features. In this review, we summarized the immune-related events including changes in and effects of immune cells, immune-related cytokines, and signaling pathways at different periods in the process of DO. We aim to elucidated our understanding and unknowns about the immunomodulatory role of DO. The goal of this is to use the known knowledge to further modify existing methods of DO, and to develop novel DO strategies in our unknown areas through more detailed studies of the work we have done.
Collapse
|
9
|
The Distraction Osteogenesis Callus: a Review of the Literature. Clin Rev Bone Miner Metab 2022. [DOI: 10.1007/s12018-021-09282-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
10
|
Mi J, Xu J, Yao Z, Yao H, Li Y, He X, Dai B, Zou L, Tong W, Zhang X, Hu P, Ruan YC, Tang N, Guo X, Zhao J, He J, Qin L. Implantable Electrical Stimulation at Dorsal Root Ganglions Accelerates Osteoporotic Fracture Healing via Calcitonin Gene-Related Peptide. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103005. [PMID: 34708571 PMCID: PMC8728818 DOI: 10.1002/advs.202103005] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/11/2021] [Indexed: 05/18/2023]
Abstract
The neuronal engagement of the peripheral nerve system plays a crucial role in regulating fracture healing, but how to modulate the neuronal activity to enhance fracture healing remains unexploited. Here it is shown that electrical stimulation (ES) directly promotes the biosynthesis and release of calcitonin gene-related peptide (CGRP) by activating Ca2+ /CaMKII/CREB signaling pathway and action potential, respectively. To accelerate rat femoral osteoporotic fracture healing which presents with decline of CGRP, soft electrodes are engineered and they are implanted at L3 and L4 dorsal root ganglions (DRGs). ES delivered at DRGs for the first two weeks after fracture increases CGRP expression in both DRGs and fracture callus. It is also identified that CGRP is indispensable for type-H vessel formation, a biological event coupling angiogenesis and osteogenesis, contributing to ES-enhanced osteoporotic fracture healing. This proof-of-concept study shows for the first time that ES at lumbar DRGs can effectively promote femoral fracture healing, offering an innovative strategy using bioelectronic device to enhance bone regeneration.
Collapse
Affiliation(s)
- Jie Mi
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of OrthopaedicsShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011People's Republic of China
| | - Jian‐Kun Xu
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Zhi Yao
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Hao Yao
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Ye Li
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xuan He
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Bing‐Yang Dai
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Li Zou
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Wen‐Xue Tong
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xiao‐Tian Zhang
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Pei‐Jie Hu
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Ye Chun Ruan
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Ning Tang
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| | - Xia Guo
- Department of Biomedical EngineeringThe Hong Kong Polytechnic UniversityHung Hom999077Hong Kong
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopaedic ImplantsDepartment of OrthopaedicsShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine639 Zhizaoju RoadShanghai200011People's Republic of China
| | - Ju‐Fang He
- Departments of Neuroscience and Biomedical SciencesCity University of Hong KongKowloon Tong999077Hong Kong
| | - Ling Qin
- Musculoskeletal Research LaboratoryDepartment of Orthopedics & TraumatologyInnovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong KongHong Kong999077China
| |
Collapse
|
11
|
He Q, Li R, Hu B, Li X, Wu Y, Sun P, Jia Y, Guo Y. Stromal cell-derived factor-1 promotes osteoblastic differentiation of human bone marrow mesenchymal stem cells via the lncRNA-H19/miR-214-5p/BMP2 axis. J Gene Med 2021; 23:e3366. [PMID: 34032330 DOI: 10.1002/jgm.3366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Stromal cell-derived factor-1 (SDF-1) plays an important role in the osteoblastic differentiation of human bone marrow mesenchymal stem cells (hBMMSCs), but the specific mechanism remains unclear. Our study aimed to clarify the role of the lncRNA-H19/miR-214-5p/BMP2 axis in the osteoblastic differentiation of hBMMSCs induced by SDF-1. METHODS We used reverse-transcriptase polymerase chain reaction, western blotting, alkaline phosphatase activity test, and Alizarin red staining to evaluate the osteoblastic differentiation of primary hBMMSCs and the luciferase reporter assay to determine if lncRNA-H19 binds with miR-214-5p. RESULTS Our results indicated that SDF-1 (50 ng/mL) promotes the osteoblastic differentiation of hBMMSCs, significantly upregulates osteoblastogenic genes (OCN, OSX, RUNX2, and ALP), and increases Alizarin red staining, alkaline phosphatase activity, and lncRNA-H19 expression. Luciferase reporter assay verified that lncRNA-H19 binds with and represses miR-214-5p, thereby upregulating BMP2 expression. Use of miR-214-5p inhibitor or overexpression of lncRNA-H19 can promote the osteoblastic differentiation of hBMMSCs, but miR-214-5p or shH19 inhibits the osteoblastic differentiation of hBMMSCs. Treatment with an miR-214-5p inhibitor could rescue the inhibitory effect of shH19 on the osteoblastic differentiation of hBMMSCs. CONCLUSIONS Taken together, SDF-1 promotes the osteoblastic differentiation of hBMMSCs through the lncRNA-H19/miR-214-5p/BMP2 axis. Increased osteoblastic differentiation by an miR-214-5p inhibitor reveals a new possible strategy for the treatment of bone defect and osteoporosis.
Collapse
Affiliation(s)
- Qiting He
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ruibin Li
- Department of Orthopedic Surgery, Linyi central hospital, Linyi, Shandong, China
| | - Beibei Hu
- Department of Ultrasound, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xuezhou Li
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yunpeng Wu
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Pengfei Sun
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuhua Jia
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yongyuan Guo
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
12
|
Fang J, Xu J, Zhang Y, Chen H, Ma Z, Huang Z, Hu J. Stromal cell-derived factor-1 may play pivotal role in distraction-stimulated neovascularization of diabetic foot ulcer. Med Hypotheses 2021; 149:110548. [PMID: 33690002 DOI: 10.1016/j.mehy.2021.110548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/16/2021] [Accepted: 02/24/2021] [Indexed: 02/05/2023]
Abstract
Diabetic foot ulcer (DFU) has become a major medical, social and economic concern worldwide. It is highly desirable to develop promising new solutions to effectively and appropriately treat DFU. In recent years, investigators have used an innovative technology called proximal tibial cortex transverse distraction (PTCTD) to treat DFU and have achieved satisfactory results in terms of improved wound healing and circumvention of amputation as a consequence of enhanced neovascularization and perfusion of the ulcerated feet after the operation, but the underlying mechanism has not been explored. Previous studies have suggested that in addition to stimulating osteogenesis, bone distraction also facilitates neovascularization, which may be associated with the chemokine stromal cell-derived factor-1 (SDF-1). As an important member of the chemokine family, SDF-1 is primarily responsible for the homing and migration of endothelial progenitor cells (EPCs) or bone marrow-derived mesenchymal stem cells (BMSCs), and plays a central role in the process of neovascularization. In vivo or in vitro experiments show that bone distraction can induce the expression of SDF-1 and increase its plasma concentration. Moreover, some researchers have found that an insufficient level of SDF-1 in the circulation and wounds of patients with DFU can lead to impaired neovascularization. Therefore, we believe that SDF-1 plays an important role in promoting neovascularization of DFU as a result of bone distraction. We summarize the currently relevant literature to put forward an undisclosed but meaningful mechanism of bone distraction in the treatment of DFU.
Collapse
Affiliation(s)
- Jiezhuang Fang
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jiankun Xu
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuantao Zhang
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Hongjiang Chen
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zebin Ma
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhonglian Huang
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jun Hu
- Department of Orthopedics, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
13
|
Zhang L, He H, Zhang M, Wu Y, Xu X, Yang M, Mei L. Assessing the effect and related mechanism of naringenin on the proliferation, osteogenic differentiation and endothelial differentiation of human periodontal ligament stem cells. Biochem Biophys Res Commun 2020; 534:337-342. [PMID: 33250176 DOI: 10.1016/j.bbrc.2020.11.081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 11/29/2022]
Abstract
Naringenin (NAR) is a natural flavonoid which exerts extensive biological activity, including anti-oxidation, anti-inflammation, anti-cancer, immune regulation and so on. However, the effect and mechanism of NAR in the alveolar bone regeneration are still unclear, which limits its clinical use. Hence, we investigated the effects of NAR in the proliferation, osteogenic and endothelial differentiation of human periodontal ligament stem cells (hPDLSCs) and explore the possible mechanism. The results showed that the proper concentrations (100 nM-10 μM) of NAR can promote the proliferation rate, osteogenic and endothelial differentiation of hPDLSCs. And the 1 μM NAR had the best proliferation promoting effect, while the 10 μM NAR had the best ability of promoting osteogenic and endothelial differentiation. NAR also promoted the mRNA expression of SDF-1 in a concentration dependent manner in PDLSCs. After adding the selective CXCR4 antagonist AMD3100, the osteogenic effect of NAR on PDLSCs is slightly enhanced, while the endothelial differentiation effect of NAR on hPDLSCs is attenuated. In summary, these results indicated that NAR promoted the proliferation of hPDLSCs, and promoted endothelial differentiation of hPDLSCs via SDF-1 to activate SDF-1/CXCR4 signaling pathway. However, the mechanism of which SDF-1 related signaling pathway is activated by NAR to enhance the osteogenic differentiation of hPDLSCs still needs to be investigated.
Collapse
Affiliation(s)
- Li Zhang
- Department of Orthodontics, Hospital of Stomatology, Southwest Medical University, Luzhou, 646000, China; Oral&Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, 646000, China
| | - Haiyan He
- Department of Orthodontics, Hospital of Stomatology, Southwest Medical University, Luzhou, 646000, China; Oral&Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, 646000, China
| | - Min Zhang
- Department of Orthodontics, Hospital of Stomatology, Southwest Medical University, Luzhou, 646000, China; Oral&Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, 646000, China
| | - Yujie Wu
- Department of Orthodontics, Hospital of Stomatology, Southwest Medical University, Luzhou, 646000, China; Oral&Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, 646000, China
| | - Xiaomei Xu
- Department of Orthodontics, Hospital of Stomatology, Southwest Medical University, Luzhou, 646000, China; Oral&Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, 646000, China.
| | - Maohua Yang
- Oral&Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, 646000, China
| | - Li Mei
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, Dunedin, 9054, New Zealand
| |
Collapse
|
14
|
Mi J, Xu J, Yao H, Li X, Tong W, Li Y, Dai B, He X, Chow DHK, Li G, Lui KO, Zhao J, Qin L. Calcitonin Gene-Related Peptide Enhances Distraction Osteogenesis by Increasing Angiogenesis. Tissue Eng Part A 2020; 27:87-102. [PMID: 32375579 DOI: 10.1089/ten.tea.2020.0009] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Distraction osteogenesis (DO) is a well-established surgical technique for treating bone defect and limb lengthening. The major drawback of DO is the long treatment period as the external fixator has to be kept in place until consolidation is completed. Calcitonin gene-related peptide (CGRP) has been reported to promote angiogenesis by affecting endothelial progenitor cells (EPCs) in limb ischemia and wound healing. Thus, the goal of this study was to evaluate the angiogenic effect of exogenous CGRP on bone regeneration in a rat DO model. Exogenous CGRP was directly injected into the bone defect after each cycle of distraction in vivo. Microcomputed tomography, biomechanical test, and histological analysis were performed to assess the new bone formation. Angiography and immunofluorescence were performed to assess the formation of blood vessels. CD31+CD144+ EPCs in the bone defect were quantified with flow cytometry. In in vitro study, bone marrow stem cells (BMSCs) were used to investigate the effect of CGRP on EPCs production during endothelial differentiation. Our results showed that CGRP significantly promoted bone regeneration and vessel formation after consolidation. CGRP significantly increased the fraction of CD31+CD144+EPCs and the capillary density in the bone defect at the end of distraction phase. CGRP increased EPC population in the endothelial differentiation of BMSCs in vitro by activating PI3K/AKT signaling pathway. Furthermore, differentiated EPCs rapidly assembled into tube-like structures and promoted osteogenic differentiation of BMSCs. In conclusion, CGRP increased EPC population and promoted blood vessel formation and bone regeneration at the defect region in a DO model. Impact statement Distraction osteogenesis (DO) is a well-established surgical technique for limb lengthening and bone defect. The disadvantage of this technique is that external fixator is needed to be kept in place for about 12 months. This may result in increased risk of infection, financial burden, and negative psychological impacts. In this study, we have injected calcitonin gene-related peptide (CGRP) into the defect region after distraction and found that CGRP enhanced vessel formation and bone regeneration in a rat DO model. This suggests that a controlled delivery system for CGRP could be developed and applied clinically for accelerating bone regeneration in patients with DO.
Collapse
Affiliation(s)
- Jie Mi
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xisheng Li
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Li
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xuan He
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Li
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Kathy O Lui
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jie Zhao
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Department of Orthopedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
15
|
An update to the advances in understanding distraction histogenesis: From biological mechanisms to novel clinical applications. J Orthop Translat 2020. [DOI: 10.1016/j.jot.2020.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
16
|
Chang M, Lin H, Fu H, Wang J, Yang Y, Wan Z, Han G. CREB activation affects mesenchymal stem cell migration and differentiation in periodontal tissues due to orthodontic force. Int J Biochem Cell Biol 2020; 129:105862. [PMID: 33045372 DOI: 10.1016/j.biocel.2020.105862] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/31/2022]
Abstract
During the orthodontic tooth movement, cells in periodontal ligament could differentiate into osteoblasts to synthesize alveolar bone as well as affect the proliferation, migration and differentiation of mesenchymal stem cells, which also contribute to bone remodeling. However, the mechanism is still largely elusive. Here, we evaluated the expression of CREB at the tension site of mouse periodontal ligament under orthodontic mechanical strain and in the cyclic tension strain treated human periodontal ligament cells. Then, through gain and loss of function analysis, we revealed that CREB in PDLCs promotes SDF-1 and FGF2 secretion, which enhance the migration and osteoblastic differentiation of BMSCs. We further discovered that CREB transcriptionally activates FGF2 and SDF-1 expressions by binding to the promoter regions.In conclusion, this study confirms that CREB is an upregulated gene in periodontal ligament under orthodontic tension strain stimulation and plays an important role in regulating BMSCs' physiological activity in orthodontic tension strain-induced bone formation.
Collapse
Affiliation(s)
- Maolin Chang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Heng Lin
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Haidi Fu
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jie Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Yang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ziqiu Wan
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guangli Han
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
17
|
Jia Y, Qiu S, Xu J, Kang Q, Chai Y. Exosomes Secreted by Young Mesenchymal Stem Cells Promote New Bone Formation During Distraction Osteogenesis in Older Rats. Calcif Tissue Int 2020; 106:509-517. [PMID: 32103287 DOI: 10.1007/s00223-019-00656-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/31/2019] [Indexed: 02/07/2023]
Abstract
Distraction osteogenesis (DO) is a clinically effective procedure to regenerate large bone defects. However, the treatment duration is undesirably lengthy, especially in elderly patients. Exosomes derived from mesenchymal stem cells (MSC-Exos) could exert the beneficial effects while avoiding the possible complications of stem cell transplantation. This study aimed to evaluate the effects of MSC-Exos on bone regeneration during DO in older rats. Exosomes were isolated from the supernatants of young bone marrow mesenchymal stem cells (BMSCs) through ultra-centrifugation, and characterized using transmission electron microscopy, western blot, and tunable resistive pulse sensing analysis. The effects of MSC-Exos on the proliferation and differentiation of older BMSCs were evaluated using CCK-8 assay, ALP and ARS staining, and qRT-PCR. Unilateral tibial DO model was established on older Sprague-Dawley rats and MSC-Exos or phosphate buffer saline was locally injected into the distraction gaps after distraction weekly. Bone regeneration were evaluated using X-ray, Micro-CT, mechanical test, and histological staining. The MSC-Exos were round or cup-shaped vesicles ranging from 60 to 130 nm in diameter and expressed markers including CD9, CD63, and TSG101. The in vitro results indicated that MSC-Exos could enhance the proliferation and osteogenic differentiation of older BMSCs. Bone regeneration was markedly accelerated in rats treated with MSC-Exos according to the results of X-ray, micro-CT, and histological analysis. The distracted tibias from the MSC-Exos group also demonstrated better mechanical properties. These results suggest that MSC-Exos promote DO-mediated bone regeneration in older rats through enhancing the proliferation and osteogenic capacity of BMSCs.
Collapse
Affiliation(s)
- Yachao Jia
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China
| | - Shuo Qiu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China
| | - Qinglin Kang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China.
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Rd 600, Shanghai, 200233, China.
| |
Collapse
|
18
|
Wang J, Xu J, Hopkins C, Chow DH, Qin L. Biodegradable Magnesium-Based Implants in Orthopedics-A General Review and Perspectives. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902443. [PMID: 32328412 PMCID: PMC7175270 DOI: 10.1002/advs.201902443] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/06/2020] [Indexed: 05/10/2023]
Abstract
Biodegradable Mg-based metals may be promising orthopedic implants for treating challenging bone diseases, attributed to their desirable mechanical and osteopromotive properties. This Review summarizes the current status and future research trends for Mg-based orthopedic implants. First, the properties between Mg-based implants and traditional orthopedic implants are compared on the following aspects: in vitro and in vivo degradation mechanisms of Mg-based implants, peri-implant bone responses, the fate of the degradation products, and the cellular and molecular mechanisms underlying the beneficial effects of Mg ions on osteogenesis. Then, the preclinical studies conducted at the low weight bearing sites of animals are introduced. The innovative strategies (for example, via designing Mg-containing hybrid systems) are discussed to address the limitations of Mg-based metals prior to their clinical applications at weight-bearing sites. Finally, the available clinical studies are summarized and the challenges and perspectives of Mg-based orthopedic implants are discussed. Taken together, the progress made on the development of Mg-based implants in basic, translational, and clinical research has laid down a foundation for developing a new era in the treatment of challenging and prevalent bone diseases.
Collapse
Affiliation(s)
- Jia‐Li Wang
- School of Biomedical EngineeringSun Yat‐sen UniversityGuangzhou510006P. R. China
- Musculoskeletal Research LaboratoryDepartment of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARP. R. China
| | - Jian‐Kun Xu
- Musculoskeletal Research LaboratoryDepartment of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARP. R. China
- Innovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARP. R. China
| | - Chelsea Hopkins
- Musculoskeletal Research LaboratoryDepartment of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARP. R. China
| | - Dick Ho‐Kiu Chow
- Musculoskeletal Research LaboratoryDepartment of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARP. R. China
- Innovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARP. R. China
| | - Ling Qin
- Musculoskeletal Research LaboratoryDepartment of Orthopaedics & TraumatologyThe Chinese University of Hong KongHong Kong SARP. R. China
- Innovative Orthopaedic Biomaterial and Drug Translational Research LaboratoryLi Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARP. R. China
| |
Collapse
|
19
|
Yang Y, Pan Q, Zou K, Wang H, Zhang X, Yang Z, Lee WYW, Wei B, Gu W, Yang YP, Lin S, Li G. Administration of allogeneic mesenchymal stem cells in lengthening phase accelerates early bone consolidation in rat distraction osteogenesis model. Stem Cell Res Ther 2020; 11:129. [PMID: 32197646 PMCID: PMC7083044 DOI: 10.1186/s13287-020-01635-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/20/2020] [Accepted: 03/04/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Distraction osteogenesis (DO) is a surgical technique to promote bone regeneration which may require long duration for bone consolidation. Bone marrow-derived mesenchymal stem cells (MSCs) have been applied to accelerate bone formation in DO. However, the optimal time point for cell therapy in DO remains unknown. This study sought to determine the optimal time point of cell administration to achieve early bone consolidation in DO. We hypothesized that the ratio of circulating MSCs to peripheral mononuclear cells and the level of cytokines in serum might be indicators for cell administration in DO. METHODS Unilateral tibial osteotomy with an external fixator was performed in adult Sprague Dawley rats. Three days after osteotomy, the tibia was lengthened at 0.5 mm/12 h for 5 days. At first, 5 rats were used to analyze the blood components at 6 different time points (3 days before lengthening, on the day lengthening began, or 3, 6, 10, or 14 days after lengthening began) by sorting circulating MSCs and measuring serum levels of stromal cell-derived factor 1 (SDF-1) and interleukin 1β. Then, 40 rats were used for cell therapy study. A single dose of 5 × 105 allogeneic MSCs was locally injected at the lengthening site on day 3, 6, or 10 after lengthening began, or 3 doses of MSCs were injected at the three time points. Sequential X-ray radiographs were taken weekly. Endpoint examinations included micro-computed tomography analysis, mechanical testing, histomorphometry, and histology. RESULTS The number of circulating MSCs and serum level of SDF-1 were significantly increased during lengthening, and then decreased afterwards. Single injection of MSCs during lengthening phase (on day 3, but not day 6 or 10) significantly increased bone volume fraction, mechanical maximum loading, and bone mineralization of the regenerate. Triple injections of MSCs at three time points also significantly increased bone volume and maximum loading of the regenerates. CONCLUSION This study demonstrated that bone consolidation could be accelerated by a single injection of MSCs during lengthening when the ratio of peripheral MSCs to mononuclear cells and the serum SDF-1 presented at peak levels concurrently, suggesting that day 3 after lengthening began may be the optimal time point for cell therapy to promote early bone consolidation.
Collapse
Affiliation(s)
- Yanhua Yang
- Department of Central Laboratory, Changzhou Seventh People's Hospital, Changzhou, China.,Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Qi Pan
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Kaijie Zou
- Department of Central Laboratory, Changzhou Seventh People's Hospital, Changzhou, China.,Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Haixing Wang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Xiaoting Zhang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Zhengmeng Yang
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Wayne Yuk Wai Lee
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Bo Wei
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China
| | - Weidong Gu
- Department of Central Laboratory, Changzhou Seventh People's Hospital, Changzhou, China
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, USA.,Department of Materials Science and Engineering, School of Engineering, Stanford University, Stanford, USA.,Department of Bioengineering, School of Medicine, Stanford University, Stanford, USA
| | - Sien Lin
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China. .,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China. .,Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, China. .,Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, USA.
| | - Gang Li
- Department of Orthopaedic and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China. .,Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China. .,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China. .,Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
20
|
miR-142-5p as a CXCR4-Targeted MicroRNA Attenuates SDF-1-Induced Chondrocyte Apoptosis and Cartilage Degradation via Inactivating MAPK Signaling Pathway. Biochem Res Int 2020; 2020:4508108. [PMID: 32047668 PMCID: PMC7003277 DOI: 10.1155/2020/4508108] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/06/2020] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a chronic joint function disorder with characteristics of chondrocytes reduction and extracellular matrix (ECM) components destruction. MicroRNAs (miRNAs) and the SDF-1/CXCR4 axis are essential factors of chondrocyte apoptosis and ECM degeneration. However, very few studies have investigated the correlation between miRNAs and the SDF-1/CXCR4 axis in osteoarthritis so far. Here, through miRNAs microarray and bioinformatics analyses, we identified miR-142-5p as a CXCR4-targeted and dramatically downregulated miRNA in cartilage from OA patients, as well as in SDF-1-induced OA chondrocytes in vitro. In SDF-1-treated primary human OA chondrocytes that were transfected with a miR-142-5p mimic or inhibitor, the expression of CXCR4 was found to be inversely correlated with the expression of miR-142-5p. The dual luciferase reporter assay further verified the target relationship between miR-142-5p and CXCR4. Overexpression of miR-142-5p alleviated OA pathology by suppressing chondrocyte apoptosis, even in CXCR4 overexpressed OA chondrocytes. This was associated with decreased cartilage matrix degradation, reduced cartilage inflammation, and inactivated MAPK signaling pathway. Our study suggests that upregulated expression of CXCR4-targeted miR-142-5p can inhibit apoptosis, inflammation, and matrix catabolism and inactivate the MAPK signaling pathway in OA chondrocytes. Our work provides important insight into targeting miR-142-5p and the SDF-1/CXCR4 axis in OA therapy.
Collapse
|
21
|
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: 0.8] [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
|
22
|
Meeson R, Sanghani-Keri A, Coathup M, Blunn G. CXCR4 Antagonism to Treat Delayed Fracture Healing. Tissue Eng Part A 2019; 25:1242-1250. [PMID: 30612520 PMCID: PMC6864747 DOI: 10.1089/ten.tea.2018.0265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A significant number of fractures develop nonunion. Stem cell homing is regulated through stromal cell-derived factor 1 (SDF1) and its receptor CXCR4. Stem/progenitor cell populations can be endogenously mobilized by administering growth factors with a pharmacological antagonist of CXCR4, AMD3100, which may be a means to improve fracture healing. A 1.5 mm femoral osteotomy in Wistar rats was stabilized with an external fixator. Rats were pretreated with phosphate buffered saline [PBS(P)], vascular endothelial growth factor [VEGF(V)], insulin-like growth factor-1 [IGF1(I)], or granulocyte colony stimulating factor [GCSF(G)] before AMD3100. A control group (C) did not receive growth factors or AMD3100. Bone formation after 5 weeks was analyzed. Group P had a significant increase in total bone volume (BV) (p = 0.01) and group I in percentage bone in the fracture gap (p = 0.035). Group G showed a decrease in BV. All treated groups had an increase in trabecular thickness. Histology showed decreased cartilage tissue associated with increased bone in groups with improved healing, and increased fibrous tissue in poorly performing groups. Antagonism of SDF1-CXCR4 axis can boost impaired fracture healing. AMD3100 given alone was the most effective means to boost healing, whereas pretreatment with GCSF reduced healing. AMD3100 is likely mobilizing stem cells into the blood stream that home to the fracture site enhancing healing.
Collapse
Affiliation(s)
- Richard Meeson
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,Department of Clinical Services and Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | - Anita Sanghani-Keri
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom
| | - Melanie Coathup
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,University of Central Florida, Orlando, Florida
| | - Gordon Blunn
- Division of Surgery, Institute of Orthopaedics and Musculoskeletal Science, University College London, London, United Kingdom.,University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
23
|
Meeson R, Sanghani‐Keri A, Coathup M, Blunn G. VEGF with AMD3100 endogenously mobilizes mesenchymal stem cells and improves fracture healing. J Orthop Res 2019; 37:1294-1302. [PMID: 30345545 PMCID: PMC6563072 DOI: 10.1002/jor.24164] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/11/2018] [Indexed: 02/04/2023]
Abstract
A significant number of fractures develop non-union. Mesenchymal stem cell (MSC) therapy may be beneficial, however, this requires cell acquisition, culture and delivery. Endogenous mobilization of stem cells offers a non-invasive alternative. The hypothesis was administration of VEGF and the CXCR4 antagonist AMD3100 would increase the circulating pool of available MSCs and improve fracture healing. Ex-breeder female wistar rats received VEGF followed by AMD3100, or sham PBS. Blood prepared for culture and colonies were counted. P3 cells were analyzed by flow cytometry, bi-differentiation. The effect of mobilization on fracture healing was evaluated with 1.5 mm femoral osteotomy stabilized with an external fixator in 12-14 week old female Wistars. The mobilized group had significantly greater number of cfus/ml compared to controls, p = 0.029. The isolated cells expressed 1.8% CD34, 35% CD45, 61% CD29, 78% CD90, and differentiated into osteoblasts but not into adipocytes. The fracture gap in animals treated with VEGF and AMD3100 showed increased bone volume; 5.22 ± 1.7 µm3 and trabecular thickness 0.05 ± 0.01 µm compared with control animals (4.3 ± 3.1 µm3 , 0.04 ± 0.01 µm, respectively). Radiographic scores quantifying fracture healing (RUST) showed that the animals in the mobilization group had a higher healing score compared to controls (9.6 vs. 7.7). Histologically, mobilization resulted in significantly lower group variability in bone formation (p = 0.032) and greater amounts of bone and less fibrous tissue than the control group. Clinical significance: This pre-clinical study demonstrates a beneficial effect of endogenous MSC mobilization on fracture healing, which may have translation potential to prevent or treat clinical fractures at risk of delayed or non-union fractures. © 2018 The Authors. Journal of Orthopaedic Research® Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 37:1294-1302, 2019.
Collapse
Affiliation(s)
- Richard Meeson
- Division of SurgeryUniversity College LondonStanmoreUnited Kingdom,Royal Veterinary CollegeHertfordshireUnited Kingdom
| | | | - Melanie Coathup
- Division of SurgeryUniversity College LondonStanmoreUnited Kingdom,University of Central FloridaFlorida
| | - Gordon Blunn
- Division of SurgeryUniversity College LondonStanmoreUnited Kingdom,University of PortsmouthPortsmouthUnited Kingdom
| |
Collapse
|
24
|
Howait M, Albassam A, Yamada C, Sasaki H, Bahammam L, Azuma MM, Cintra LTA, Satoskar AR, Yamada S, White R, Kawai T, Movila A. Elevated Expression of Macrophage Migration Inhibitory Factor Promotes Inflammatory Bone Resorption Induced in a Mouse Model of Periradicular Periodontitis. THE JOURNAL OF IMMUNOLOGY 2019; 202:2035-2043. [PMID: 30737274 DOI: 10.4049/jimmunol.1801161] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/15/2019] [Indexed: 12/23/2022]
Abstract
Locally produced osteoclastogenic factor RANKL plays a critical role in the development of bone resorption in periradicular periodontitis. However, because RANKL is also required for healthy bone remodeling, it is plausible that a costimulatory molecule that upregulates RANKL production in inflammatory periradicular periodontitis may be involved in the pathogenic bone loss processes. We hypothesized that macrophage migration inhibitory factor (MIF) would play a role in upregulating the RANKL-mediated osteoclastogenesis in the periradicular lesion. In response to pulp exposure, the bone loss and level of MIF mRNA increased in the periradicular periodontitis, which peaked at 14 d, in conjunction with the upregulated expressions of mRNAs for RANKL, proinflammatory cytokines (TNF-α, IL-6, and IL-1β), chemokines (MCP-1 and SDF-1), and MIF's cognate receptors CXCR4 and CD74. Furthermore, expressions of those mRNAs were found significantly higher in wild-type mice compared with that of MIF-/- mice. In contrast, bacterial LPS elicited the production of MIF from ligament fibroblasts in vitro, which, in turn, enhanced their productions of RANKL and TNF-α. rMIF significantly upregulated the number of TRAP+ osteoclasts in vitro. Finally, periapical bone loss induced in wild-type mice were significantly diminished in MIF-/- mice. Altogether, the current study demonstrated that MIF appeared to function as a key costimulatory molecule to upregulate RANKL-mediated osteoclastogenesis, leading to the pathogenically augmented bone resorption in periradicular lesions. These data also suggest that the approach to neutralize MIF activity may lead to the development of a therapeutic regimen for the prevention of pathogenic bone loss in periradicular periodontitis.
Collapse
Affiliation(s)
- Mohammed Howait
- School of Dental Medicine, Harvard University, Boston, MA 02115.,Faculty of Dentistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia.,The Forsyth Institute, Cambridge, MA 02142
| | - Abdullah Albassam
- School of Dental Medicine, Harvard University, Boston, MA 02115.,Faculty of Dentistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia.,The Forsyth Institute, Cambridge, MA 02142
| | - Chiaki Yamada
- College of Dental Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33324
| | - Hajime Sasaki
- School of Dental Medicine, Harvard University, Boston, MA 02115.,The Forsyth Institute, Cambridge, MA 02142.,School of Dentistry, University of Michigan, Ann Arbor, MI 48109
| | - Laila Bahammam
- Faculty of Dentistry, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mariane Maffei Azuma
- The Forsyth Institute, Cambridge, MA 02142.,School of Dentistry, University of Michigan, Ann Arbor, MI 48109
| | | | - Abhay R Satoskar
- Department of Microbiology, The Ohio State University, Columbus, OH 43210; and
| | - Satoru Yamada
- Graduate School of Dentistry, Tohoku University, Tohoku, Sendai 980-8575, Japan
| | - Robert White
- School of Dental Medicine, Harvard University, Boston, MA 02115
| | - Toshihisa Kawai
- College of Dental Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33324
| | - Alexandru Movila
- School of Dental Medicine, Harvard University, Boston, MA 02115; .,The Forsyth Institute, Cambridge, MA 02142.,College of Dental Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33324
| |
Collapse
|
25
|
Shen J, Ye X. [Effect of "accordion" technique on bone consolidation during distraction osteogenesis]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2018; 32:558-567. [PMID: 29806343 DOI: 10.7507/1002-1892.201712094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective To investigate the effect, right timing, and mechanism of "accordion" technique on bone regeneration in rat distraction osteogenesis model. Methods Fifty-four 12-week-old male Sprague Dawley rats underwent right tibial distraction osteogenesis procedure. After a 5-day latency, the distraction was performed for 7 days followed by 6-week consolidation. All animals were randomly divided into 4 groups based on different periods of "accordion" maneuvers in consolidation phase: control group ( n=18) with no manipulation, and three experimental groups including early-phase group ( n=18), mid-phase group ( n=12), and late-phase group ( n=6) with "accordion" maneuvers applied at 1, 3, and 5 weeks, respectively. The duration of the "accordion" maneuver was 7 days consisting of a 3.5-day compression and 3.5-day distraction. Rats in control group and early-phase group were sacrificed at 2, 4, and 6 weeks of the consolidation phase; rats in mid-phase group were sacrificed at 4 and 6 weeks of the consolidation phase; and rats in late-phase group were sacrificed at 6 weeks of the consolidation phase. Bilateral tibias from 6 rats in each group at each time point were obtained. Callus formation was monitored by X-ray radiography every week; new bone was reconstructed by Micro-CT three-dimensional reconstruction. The change of bone structure was evaluated, and parameters containing bone volume (BV)/tissue volume (TV) ratio (BV/TV) and bone mineral density (BMD) in three thresholds (158-211, 211-1 000, 158-1 000) were recorded and calculated at 6 weeks. Mechanical test consisting of ultimate load, modulus of elasticity, and energy to failure was performed. Histological analysis, such as Von Kossa staining, Safranin O staining, and HE staining, was done. Immunohistochemical staining using markers of osterix (OSX), osteocalcin (OCN), and vascular endothelial growth factor (VEGF) was analyzed. Results Images of X-ray showed that callus formation increased significantly in the mid-phase group. Micro-CT three-dimensional reconstruction demonstrated the mid-phase group owned fastest reconstructed speed among 4 groups, the cortical bone was continual at 6 weeks. At 6 weeks, the BMD and BV/TV in thresholds 158-1 000 and 211-1 000 in mid-phase group were higher than those in other groups. The results of mechanical test showed that ultimate load, modulus of elasticity, and energy to failure in mid-phase group were significantly higher than those in other groups ( P<0.05). Histological testing showed that the continuity of bone marrow cavity in mid-phase group was evident at 6 weeks after distraction. Immunohistochemical analyses confirmed the expression levels of osteogenesis (OCN, OSX) and angiogenesis (VEGF) elevated remarkably and then returned to normal in mid-phase group. Conclusion The "accordion" technique is beneficial for new callus formation in distraction area. Applying the maneuver during the middle phase of the consolidation period was effective to accelerate new bone formation in rat distraction osteogenesis model.
Collapse
Affiliation(s)
- Jie Shen
- Department of Orthopedics, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003, P.R.China
| | - Xiaojian Ye
- Department of Orthopedics, Changzheng Hospital, the Second Military Medical University, Shanghai, 200003,
| |
Collapse
|
26
|
Wang R, Liu W, Du M, Yang C, Li X, Yang P. The differential effect of basic fibroblast growth factor and stromal cell‑derived factor‑1 pretreatment on bone morrow mesenchymal stem cells osteogenic differentiation potency. Mol Med Rep 2017; 17:3715-3721. [PMID: 29359787 PMCID: PMC5802181 DOI: 10.3892/mmr.2017.8316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/03/2017] [Indexed: 12/17/2022] Open
Abstract
In situ tissue engineering has become a novel strategy to repair periodontal/bone tissue defects. The choice of cytokines that promote the recruitment and proliferation, and potentiate and maintain the osteogenic differentiation ability of mesenchymal stem cells (MSCs) is the key point in this technique. Stromal cell‑derived factor‑1 (SDF‑1) and basic fibroblast growth factor (bFGF) have the ability to promote the recruitment, and proliferation of MSCs; however, the differential effect of SDF‑1 and bFGF pretreatment on MSC osteogenic differentiation potency remains to be explored. The present study comparatively observed osteogenic differentiation of bone morrow MSCs (BMMSCs) pretreated by bFGF or SDF‑1 in vitro. The gene and protein expression levels of alkaline phosphatase (ALP), runt related transcription factor 2 (Runx‑2) and bone sialoprotein (BSP) were detected using reverse transcription‑quantitative polymerase chain reaction and western blotting. The results showed that the expression of ALP mRNA on day 3, and BSP and Runx‑2 mRNA on day 7 in the bFGF pretreatment group was significantly higher than those in SDF‑1 pretreatment group. Expression levels of Runx‑2 mRNA, and ALP and Runx‑2 protein on day 3 in the SDF‑1 pretreatment group were higher than those in the bFGF pretreatment group. However, there was no significant difference in osteogenic differentiation ability on day 14 and 28 between the bFGF‑ or SDF‑1‑pretreatment groups and the control. In conclusion, bFGF and SDF‑1 pretreatment inhibits osteogenic differentiation of BMMSCs at the early stage, promotes it in the medium phase, and maintains it in the later stage during osteogenic induction, particularly at the mRNA level. Out of the two cytokines, bFGF appeared to have a greater effect on osteogenic differentiation.
Collapse
Affiliation(s)
- Ruolin Wang
- Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wenhua Liu
- Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Mi Du
- Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Chengzhe Yang
- Department of Oral and Maxillofacial Surgery, Qilu Hospital and Institute of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xuefen Li
- Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Pishan Yang
- Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|