1
|
Xu W, Ran B, Aizawa T, Liu W, Zhao J, Niu R, Liu Z, Gu R. The Hedgehog-GLI1 Pathway Regulates Osteogenic Differentiation of Human Cervical Posterior Longitudinal Ligament Cells by BMP Signalling Pathway. J Cell Mol Med 2025; 29:e70393. [PMID: 39910703 PMCID: PMC11798735 DOI: 10.1111/jcmm.70393] [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: 02/13/2024] [Revised: 12/17/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Cervical ossification of the posterior longitudinal ligament (OPLL) is an ectopic ossification disorder characterised by endochondral ossification. Its aetiology remains to be fully elucidated. This study aimed to clarify its pathogenesis through RNA sequencing of primary cells cultured from patients without cervical OPLL (control, PLL) and patients with cervical OPLL (disease, OPLL). We revealed for the first time the role of GLI1 within OPLL cells. Functional experiments indicated that GLI1, acting as a pivotal mediator between the upstream Hedgehog pathway and downstream BMP pathway, influences the pathogenesis of OPLL. The positive/negative effects on osteogenic differentiation following activation/inhibition of the Hedgehog pathway can be rescued by manipulating GLI1 expression. Overexpression of GLI1 activates BMP signalling, enhancing osteogenic capacity in PLL cells, while GLI1 knockdown suppresses BMP signal transduction, attenuating osteogenic differentiation in OPLL cells. Our findings highlight the significant role of the canonical Hedgehog signalling pathway and its interaction with the BMP pathway in the pathogenesis of OPLL.
Collapse
Affiliation(s)
- Wenbo Xu
- Department of Orthopaedic SurgeryChina‐Japan Union Hospital of Jilin UniversityJilinPeople's Republic of China
| | - Bingbing Ran
- Department of UltrasoundThe First Hospital of Jilin UniversityJilinPeople's Republic of China
| | - Toshimi Aizawa
- Department of Orthopaedic SurgeryTohoku University School of MedicineSendaiJapan
| | - Wanguo Liu
- Department of Orthopaedic SurgeryChina‐Japan Union Hospital of Jilin UniversityJilinPeople's Republic of China
| | - Jianhui Zhao
- Department of Orthopaedic SurgeryChina‐Japan Union Hospital of Jilin UniversityJilinPeople's Republic of China
| | - Renrui Niu
- Department of Orthopaedic SurgeryChina‐Japan Union Hospital of Jilin UniversityJilinPeople's Republic of China
| | - Zeping Liu
- Department of Orthopaedic SurgeryChina‐Japan Union Hospital of Jilin UniversityJilinPeople's Republic of China
| | - Rui Gu
- Department of Orthopaedic SurgeryChina‐Japan Union Hospital of Jilin UniversityJilinPeople's Republic of China
| |
Collapse
|
2
|
Chen R, Ai L, Zhang J, Jiang D. Dendritic Cell-Derived Exosomes Promote Tendon Healing and Regulate Macrophage Polarization in Preventing Tendinopathy. Int J Nanomedicine 2024; 19:11701-11718. [PMID: 39558915 PMCID: PMC11571930 DOI: 10.2147/ijn.s466363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Tendon injuries present a significant challenge for independent repair, and can progress into tendinopathy over time, highlighting the importance of early intervention. Dendritic cell-derived exosomes (DEXs) has been shown to shift the polarization of M1 macrophages, the predominant inflammatory cells in the early stages of tendon injury. This study introduces a therapeutic approach that effectively manages inflammation while promoting regeneration in the treatment of tendinopathy. Methods The purification and characterization of DEXs were meticulously conducted. Experiments were carried out using an Achilles tendon rupture mouse model, with weekly DEXs treatment starting on postoperative day (POD) 4. In vitro, the function of DEXs was assessed by coculturing them with tendon stem/progenitor cells (TSPCs) in culture medium containing IL-1β. Tendon healing progress was evaluated using Sirius Red staining, Masson's trichrome staining, biomechanical testing, and immunofluorescence microscopy. The inflammatory microenvironment of injured tendons was evaluated using the Luminex procedure and flow cytometry analysis. Results DEXs treatment significantly enhanced tendon cell differentiation, promoted collagen type I synthesis, and inhibited collagen type III synthesis, thereby expediting tendon healing. Furthermore, DEXs treatment improved the inflammatory microenvironment by reducing multiple cytokines (IL-1β, IL-4, IL-6, TNF-α, and IFN-γ) and induced the conversion of M1 macrophages to M2 macrophages by activating the PI3K/AKT pathway. Conclusion DEXs demonstrated a potent ability to promote tendon healing while ameliorating the inflammatory microenvironment, suggesting their potential as a therapeutic approach to prevent the development of tendinopathy.
Collapse
Affiliation(s)
- Rao Chen
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, People’s Republic of China
- Department of Laboratory Animal Science, Peking University Health Science Center, Beijing, 100191, People’s Republic of China
| | - Liya Ai
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, People’s Republic of China
- Department of Laboratory Animal Science, Peking University Health Science Center, Beijing, 100191, People’s Republic of China
| | - Jiying Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, People’s Republic of China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, People’s Republic of China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, People’s Republic of China
| | - Dong Jiang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, People’s Republic of China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, People’s Republic of China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, People’s Republic of China
| |
Collapse
|
3
|
Deng L, Liu Y, Wu Q, Lai S, Yang Q, Mu Y, Dong M. Exosomes to exosome-functionalized scaffolds: a novel approach to stimulate bone regeneration. Stem Cell Res Ther 2024; 15:407. [PMID: 39521993 PMCID: PMC11550564 DOI: 10.1186/s13287-024-04024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Bone regeneration is a complex biological process that relies on the orchestrated interplay of various cellular and molecular events. Bone tissue engineering is currently the most promising method for treating bone regeneration. However, the immunogenicity, stable and cell quantity of seed cells limited their application. Recently, exosomes, which are small extracellular vesicles released by cells, have been found to effectively address these problems and better induce bone regeneration. Meanwhile, a growing line of research has shown the cargos of exosomes may provide effective therapeutic and biomarker tools for bone repair, including miRNA, lncRNA, and proteins. Moreover, engineered scaffolds loaded with exosomes can offer a cell-free bone repair strategy, addressing immunogenicity concerns and providing a more stable functional performance. Herein, we provide a comprehensive summary of the role played by scaffolds loaded with exosomes in bone regeneration, drawing on a systematic analysis of relevant literature available on PubMed, Scopus, and Google Scholar database.
Collapse
Affiliation(s)
- Li Deng
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yang Liu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Qian Wu
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Shuang Lai
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Qiu Yang
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China
| | - Yandong Mu
- Stomatology Department, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611135, Sichuan, China.
| |
Collapse
|
4
|
Qian Y, Chen B, Sun E, Lu X, Li Z, Wang R, Fang D. Mesenchymal Stem Cell-Derived Extracellular Vesicles Alleviate Brain Damage Following Subarachnoid Hemorrhage via the Interaction of miR-140-5p and HDAC7. Mol Neurobiol 2024; 61:9136-9154. [PMID: 38592585 DOI: 10.1007/s12035-024-04118-3] [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: 10/27/2023] [Accepted: 03/08/2024] [Indexed: 04/10/2024]
Abstract
Subarachnoid hemorrhage (SAH) triggers severe neuroinflammation and cognitive impairment, where microglial M1 polarization exacerbates the injury and M2 polarization mitigates damage. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs), carrying microRNA (miR)-140-5p, offer therapeutic promise by targeting the cAMP/PKA/CREB pathway and modulating microglial responses, demonstrating a novel approach for addressing SAH-induced brain injury. This research explored the role of miR-140-5p delivered by MSC-EVs in mitigating brain damage following SAH. Serum from SAH patients and healthy individuals was analyzed for miR-140-5p and cAMP levels. The association between miR-140-5p levels, brain injury severity, and patient survival was examined, along with the target relationship between miR-140-5p and histone deacetylases 7 (HDAC7). MSC-EVs were characterized for their ability to cross the blood-brain barrier and modulate the HDAC7/AKAP12/cAMP/PKA/CREB axis, reducing M1 polarization and inflammation. The therapeutic effect of MSC-EV-miR-140-5p was demonstrated in an SAH mouse model, showing reduced neuronal apoptosis and improved neurological function. This study highlights the potential of MSC-EV-miR-140-5p in mitigating SAH-induced neuroinflammation and brain injury, providing a foundation for developing MSC-EV-based treatments for SAH.
Collapse
Affiliation(s)
- Yu Qian
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, P.R. China
| | - Bo Chen
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, P.R. China
| | - Eryi Sun
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, P.R. China
| | - Xinyu Lu
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, P.R. China
| | - Zheng Li
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, P.R. China
| | - Runpei Wang
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212000, P.R. China
| | - Dazhao Fang
- Department of Neurosurgery, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, West Huanghe Road, Huaiyin District, Huai'an, Jiangsu Province, 223300, P.R. China.
| |
Collapse
|
5
|
何 思, 文 楠, 陈 勋, 王 跃, 张 艇, 牟 雁. [ Lycium barbarum glycopeptide reduces bone loss caused by exosomes derived from human gingival fibroblasts with radiation exposure]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1752-1759. [PMID: 39505343 PMCID: PMC11744091 DOI: 10.12122/j.issn.1673-4254.2024.09.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE To explore the protective effect of Lycium barbanun glycopeptide (LbGP) against osteogenic inhibition induced by exosomes derived from human gingival fibroblasts (HGFs) exposed to radiation. METHODS Cultured HGFs with or without LbGP pretreatment were exposed to 8 Gy X-ray radiation, and the changes in cell apoptosis, senescence and α-SMA level were detected using RT-qPCR, Western blotting and β-galactosidase staining. The exosomes secreted by the treated cells were extracted, and after identification by electron microscopy, particle size analysis and Western blotting, the exosomes were added into primary cultured bone mesenchymal stem cells (BMSCs), and osteoclast activity and osteogenesis in the cell cultures were detected by Trap staining, Alizarin red staining, ALP staining, RT-qPCR and Western blotting. RESULTS In cultured HGFs, X-ray radiation significantly increased the percentage of senescent cells, which was obviously lowered by LbGP treatment. X-ray radiation significantly reduced Bcl-2/Bax ratio and increased α -SMA expression in HGFs, and these changes were significantly suppressed by LbGP pretreatment. In rat BMSCs, incubation with the exosomes derived from HGFs with radiation exposure caused a significant increase of osteoclasts, reduced calcium nodules and lowered alkaline phosphatase expression in the cells; The opposite changes were observed in the cells treated with exosomes from LbGPpretreated HGFs, which also significantly increased the cellular expressions of the osteogenic genes (BMP2, ALP, and RUNX2) and proteins (ALP and RUNX2) as compared with the exosomes from irradiated HGFs. CONCLUSION LbGP can effectively inhibit osteoclast activity and promote osteogenesis by acting on exosomes secreted by irradiated HGFs, suggesting its potential value for treatment of osteoradionecrosis of the jaw.
Collapse
Affiliation(s)
- 思齐 何
- 成飞医院,四川 成都 610073Cheng Fei Hospital, Chengdu 610073, China
| | - 楠 文
- 宜宾市第一人民医院,四川 宜宾 644000The First People's Hospital of Yibin, Yibin 644000, China
| | - 勋 陈
- 四川省人民医院口腔科,四川 成都 610072Stomatology department, Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - 跃 王
- 卷烟减害降焦四川省重点实验室,四川 成都 610100Harmful Components and Tar Reduction in Cigarette Key Laboratory of Sichuan Province, Chengdu 610100, China
| | - 艇 张
- 卷烟减害降焦四川省重点实验室,四川 成都 610100Harmful Components and Tar Reduction in Cigarette Key Laboratory of Sichuan Province, Chengdu 610100, China
| | - 雁东 牟
- 四川省人民医院口腔科,四川 成都 610072Stomatology department, Sichuan Provincial People's Hospital, Chengdu 610072, China
| |
Collapse
|
6
|
Li Y, Fan Y, Zhao S, Cheng B. Ultrasound-triggered piezoelectric polyetheretherketone with boosted osteogenesis via regulating Akt/GSK3β/β-catenin pathway. J Nanobiotechnology 2024; 22:539. [PMID: 39237993 PMCID: PMC11375992 DOI: 10.1186/s12951-024-02814-9] [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: 07/07/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024] Open
Abstract
Maxillofacial bone defects can severely impact quality of life by impairing physiological functions such as chewing, breathing, swallowing, and pronunciation. Polyether ether ketone (PEEK) is commonly used for the repair of maxillofacial defects due to its mechanical adaptability, while its osteogenic properties still need refinement. Herein, we have utilized the piezoelectric effect exhibited by barium titanate (BTO) under low-intensity pulsed ultrasound (LIPUS) to develop an ultrasound responsive PEEK (PDA@BTO-SPEEK, PBSP) through the mediating effect of polydopamine (PDA), for repairing maxillofacial bone defects. After modification by PDA@BTO, PBSP possesses better hydrophilicity, which is conducive to cell growth and adhesion. Simultaneously, by virtue of the piezoelectric characteristics of BTO, PBSP obtains a piezoelectric coefficient that matches the bone cortex. Notably, when PBSP is stimulated by LIPUS, it can generate stable electricity and effectively accelerate the osteogenic differentiation of osteoblasts through the regulation of the Piezo1-induced calcium (Ca2+) influx and Akt/GSK3β/β-catenin pathway. In addition, PBSP presents satisfactory therapeutic effects in rat skull defect models, and its osteogenic efficiency can be further improved under LIPUS stimulation with high tissue penetration. Collectively, PBSP + LIPUS exhibits great potential as a promising alternative strategy for the repair of maxillofacial bone defects.
Collapse
Affiliation(s)
- Yue Li
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Yingying Fan
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China
| | - Siyu Zhao
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| | - Bo Cheng
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
7
|
Xu J, Bao G, Jia B, Wang M, Wen P, Kan T, Zhang S, Liu A, Tang H, Yang H, Yue B, Dai K, Zheng Y, Qu X. An adaptive biodegradable zinc alloy with bidirectional regulation of bone homeostasis for treating fractures and aged bone defects. Bioact Mater 2024; 38:207-224. [PMID: 38756201 PMCID: PMC11096722 DOI: 10.1016/j.bioactmat.2024.04.027] [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: 03/01/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
Healing of fractures or bone defects is significantly hindered by overactivated osteoclasts and inhibited osteogenesis in patients with abnormal bone metabolism. Current clinical approaches using titanium alloys or stainless steel provide mechanical support but have no biological effects on bone regeneration. Therefore, designing and fabricating degradable metal materials with sufficient mechanical strength and bidirectional regulation of both osteoblasts and osteoclasts is a substantial challenge. Here, this study first reported an adaptive biodegradable Zn-0.8 Mg alloy with bidirectional regulation of bone homeostasis, which promotes osteogenic differentiation by activating the Pi3k/Akt pathway and inhibits osteoclast differentiation by inhibiting the GRB2/ERK pathway. The anti-osteolytic ability of the Zn-0.8 Mg alloy was verified in a mouse calvarial osteolysis model and its suitability for internal fracture fixation with high-strength screws was confirmed in the rabbit femoral condyle fracture model. Furthermore, in an aged postmenopausal rat femoral condyle defect model, 3D printed Zn-0.8 Mg scaffolds promoted excellent bone regeneration through adaptive structures with good mechanical properties and bidirectionally regulated bone metabolism, enabling personalized bone defect repair. These findings demonstrate the substantial potential of the Zn-0.8 Mg alloy for treating fractures or bone defects in patients with aberrant bone metabolism.
Collapse
Affiliation(s)
- Jialian Xu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Guo Bao
- Laboratory Animal centre, National Research Institute for Family Planning, Beijing, 100081, China
| | - Bo Jia
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Peng Wen
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Tianyou Kan
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Aobo Liu
- Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Haozheng Tang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Hongtao Yang
- School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| | - Kerong Dai
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200001, China
| |
Collapse
|
8
|
Xu C, Xu Z, Li G, Li J, Ye L, Ning Y, Du Y. CircFgfr2 promotes osteogenic differentiation of rat dental follicle cells by targeting the miR-133a-3p/DLX3 signaling pathway. Heliyon 2024; 10:e32498. [PMID: 38912473 PMCID: PMC11193016 DOI: 10.1016/j.heliyon.2024.e32498] [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: 02/12/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/25/2024] Open
Abstract
Dental follicle cells (DFCs) promote bone regeneration in vivo and in vitro. Circular RNAs (circRNAs) play crucial roles in bone development and regeneration. Our previous study demonstrated the upregulation of circFgfr2 expression during the osteogenic differentiation of DFCs. However, the molecular mechanisms and functional roles of circFgfr2 in DFCs osteogenesis remain unclear. In this study, we aimed to investigate the subcellular localization of circFgfr2 in DFCs using fluorescence in situ hybridization. In vitro investigations demonstrated that circFgfr2 overexpression promoted osteogenic differentiation, as evidenced by real-time quantitative polymerase chain reaction. By integrating the outcomes of bioinformatics analyses, dual luciferase reporter experiments, and chromatin isolation by RNA purification, we identified circFgfr2 as a sponge for miR-133a-3p, a key regulator of osteogenic differentiation. Moreover, miR-133a-3p suppressed osteogenic differentiation by targeting DLX3 and RUNX2 in DFCs. We validated that circFgfr2 promoted the osteogenic differentiation of DFCs through the miR-133a-3p/DLX3 axis. To further investigate the therapeutic potential of circFgfr2 in bone regeneration, we conducted in vivo experiments and histological analyses. Overall, these results confirmed the crucial role of circFgfr2 in promoting osteogenesis. In summary, our findings demonstrated that the circFgfr2/miR-133a-3p/DLX3 pathway acts as a cascade, thereby identifying circFgfr2 as a promising molecular target for bone tissue engineering.
Collapse
Affiliation(s)
- Cheng Xu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen University, Guangzhou, Guangdong, China
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Reasearch Institute of Stomatology, Nanjing University,Nanjing, Jiangsu, China
| | - Zhiqing Xu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen University, Guangzhou, Guangdong, China
| | - Guixian Li
- Operative Dentistry and Endodontics, Jiangmen Municipal Stomatological Hospital, Jiangmen, Guangdong, China
| | - Jing Li
- Department of Stomatology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Li Ye
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen University, Guangzhou, Guangdong, China
| | - Yang Ning
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen University, Guangzhou, Guangdong, China
| | - Yu Du
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen University, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
He C, Hu C, He WZ, Sun YC, Jiang Y, Liu L, Hou J, Chen KX, Jiao YR, Huang M, Huang M, Yang M, Lu Q, Wei J, Zeng C, Lei GH, Li CJ. Macrophage-derived extracellular vesicles regulate skeletal stem/progenitor Cell lineage fate and bone deterioration in obesity. Bioact Mater 2024; 36:508-523. [PMID: 39072285 PMCID: PMC11282946 DOI: 10.1016/j.bioactmat.2024.06.035] [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: 03/19/2024] [Revised: 06/07/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Obesity-induced chronic inflammation exacerbates multiple types of tissue/organ deterioration and stem cell dysfunction; however, the effects on skeletal tissue and the underlying mechanisms are still unclear. Here, we show that obesity triggers changes in the microRNA profile of macrophage-secreted extracellular vesicles, leading to a switch in skeletal stem/progenitor cell (SSPC) differentiation between osteoblasts and adipocytes and bone deterioration. Bone marrow macrophage (BMM)-secreted extracellular vesicles (BMM-EVs) from obese mice induced bone deterioration (decreased bone volume, bone microstructural deterioration, and increased adipocyte numbers) when administered to lean mice. Conversely, BMM-EVs from lean mice rejuvenated bone deterioration in obese recipients. We further screened the differentially expressed microRNAs in obese BMM-EVs and found that among the candidates, miR-140 (with the function of promoting adipogenesis) and miR-378a (with the function of enhancing osteogenesis) coordinately determine SSPC fate of osteogenic and adipogenic differentiation by targeting the Pparα-Abca1 axis. BMM miR-140 conditional knockout mice showed resistance to obesity-induced bone deterioration, while miR-140 overexpression in SSPCs led to low bone mass and marrow adiposity in lean mice. BMM miR-378a conditional depletion in mice led to obesity-like bone deterioration. More importantly, we used an SSPC-specific targeting aptamer to precisely deliver miR-378a-3p-overloaded BMM-EVs to SSPCs via an aptamer-engineered extracellular vesicle delivery system, and this approach rescued bone deterioration in obese mice. Thus, our study reveals the critical role of BMMs in mediating obesity-induced bone deterioration by transporting selective extracellular-vesicle microRNAs into SSPCs and controlling SSPC fate.
Collapse
Affiliation(s)
- Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Chen Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Wen-Zhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu-Chen Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yangzi Jiang
- School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), The Chinese University of Hong Kong, Hong Kong SAR, 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
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Kai-Xuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yu-Rui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Min Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jie Wei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Guang-Hua Lei
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
10
|
Geng X, Tang Y, Gu C, Zeng J, Zhao Y, Zhou Q, Jia L, Zhou S, Chen X. Integrin αVβ3 antagonist-c(RGDyk) peptide attenuates the progression of ossification of the posterior longitudinal ligament by inhibiting osteogenesis and angiogenesis. Mol Med 2024; 30:57. [PMID: 38698308 PMCID: PMC11067224 DOI: 10.1186/s10020-024-00822-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 04/22/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Ossification of the posterior longitudinal ligament (OPLL), an emerging heterotopic ossification disease, causes spinal cord compression, resulting in motor and sensory dysfunction. The etiology of OPLL remains unclear but may involve integrin αVβ3 regulating the process of osteogenesis and angiogenesis. In this study, we focused on the role of integrin αVβ3 in OPLL and explored the underlying mechanism by which the c(RGDyk) peptide acts as a potent and selective integrin αVβ3 inhibitor to inhibit osteogenesis and angiogenesis in OPLL. METHODS OPLL or control ligament samples were collected in surgery. For OPLL samples, RNA-sequencing results revealed activation of the integrin family, particularly integrin αVβ3. Integrin αVβ3 expression was detected by qPCR, Western blotting, and immunohistochemical analysis. Fluorescence microscopy was used to observe the targeted inhibition of integrin αVβ3 by the c(RGDyk) peptide on ligaments fibroblasts (LFs) derived from patients with OPLL and endothelial cells (ECs). The effect of c(RGDyk) peptide on the ossification of pathogenic LFs was detected using qPCR, Western blotting. Alkaline phosphatase staining or alizarin red staining were used to test the osteogenic capability. The effect of the c(RGDyk) peptide on angiogenesis was determined by EC migration and tube formation assays. The effects of the c(RGDyk) peptide on heterotopic bone formation were evaluated by micro-CT, histological, immunohistochemical, and immunofluorescence analysis in vivo. RESULTS The results indicated that after being treated with c(RGDyk), the osteogenic differentiation of LFs was significantly decreased. Moreover, the c(RGDyk) peptide inhibited the migration of ECs and thus prevented the nutritional support required for osteogenesis. Furthermore, the c(RGDyk) peptide inhibited ectopic bone formation in mice. Mechanistic analysis revealed that c(RGDyk) peptide could inhibit osteogenesis and angiogenesis in OPLL by targeting integrin αVβ3 and regulating the FAK/ERK pathway. CONCLUSIONS Therefore, the integrin αVβ3 appears to be an emerging therapeutic target for OPLL, and the c(RGDyk) peptide has dual inhibitory effects that may be valuable for the new therapeutic strategy of OPLL.
Collapse
Affiliation(s)
- Xiangwu Geng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Yifan Tang
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Changjiang Gu
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Junkai Zeng
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Yin Zhao
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Quanwei Zhou
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Lianshun Jia
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China
| | - Shengyuan Zhou
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| | - Xiongsheng Chen
- Spine Center, Department of Orthopaedics, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, China.
| |
Collapse
|
11
|
Wang L, Jiang W, Zhao S, Xie D, Chen Q, Zhao Q, Wu H, Luo J, Yang L. Sorafenib inhibits ossification of the posterior longitudinal ligament by blocking LOXL2-mediated vascularization. Bone Res 2024; 12:24. [PMID: 38594260 PMCID: PMC11004159 DOI: 10.1038/s41413-024-00327-7] [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: 05/24/2023] [Revised: 03/10/2024] [Accepted: 03/16/2024] [Indexed: 04/11/2024] Open
Abstract
Ossification of the Posterior Longitudinal Ligament (OPLL) is a degenerative hyperostosis disease characterized by the transformation of the soft and elastic vertebral ligament into bone, resulting in limited spinal mobility and nerve compression. Employing both bulk and single-cell RNA sequencing, we elucidate the molecular characteristics, cellular components, and their evolution during the OPLL process at a single-cell resolution, and validate these findings in clinical samples. This study also uncovers the capability of ligament stem cells to exhibit endothelial cell-like phenotypes in vitro and in vivo. Notably, our study identifies LOXL2 as a key regulator in this process. Through gain-and loss-of-function studies, we elucidate the role of LOXL2 in the endothelial-like differentiation of ligament cells. It acts via the HIF1A pathway, promoting the secretion of downstream VEGFA and PDGF-BB. This function is not related to the enzymatic activity of LOXL2. Furthermore, we identify sorafenib, a broad-spectrum tyrosine kinase inhibitor, as an effective suppressor of LOXL2-mediated vascular morphogenesis. By disrupting the coupling between vascularization and osteogenesis, sorafenib demonstrates significant inhibition of OPLL progression in both BMP-induced and enpp1 deficiency-induced animal models while having no discernible effect on normal bone mass. These findings underscore the potential of sorafenib as a therapeutic intervention for OPLL.
Collapse
Affiliation(s)
- Longqing Wang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Wenhao Jiang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China
| | - Siyuan Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Dong Xie
- Department of Orthopaedics, No. 905 Hospital of PLA Navy, Shanghai, PR China
| | - Qing Chen
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Qi Zhao
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Hao Wu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, PR China.
| | - Lili Yang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
12
|
Wang P, Liu B, Song C, Jia J, Wang Y, Pang K, Wang Y, Chen C. Exosome MiR-21-5p Upregulated by HIF-1α Induces Adipose Stem Cell Differentiation to Promote Ectopic Bone Formation. Chem Biodivers 2024; 21:e202301972. [PMID: 38342761 DOI: 10.1002/cbdv.202301972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/29/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Heterotopic bone occurs after burns, trauma and major orthopedic surgery, which cannot be completely cured by current treatments. The development of new treatments requires more in-depth research into the mechanism of HO. Available evidence suggests that miR-21-5p plays an important role in bone formation. However, its mechanism in traumatic HO is still unclear. First, we identified exosomes extracted from L6 cells using TEM observation of the structure and western blotting detection of the surface marker CD63. Regulation effect of HIF-1α to miR-21-5p was confirmed by q-PCR assay. Then we co-cultured L6 cells with ASCs and performed alizarin red staining and ALP detection. Overexpression of miR-21-5p upregulated BMP4, p-smad1/5/8, OCN and OPN, which suggests the BMP4-smad signaling pathway may be involved in miR-21-5p regulation of osteogenic differentiation of ASCs. Finally in vivo experiments showed that miR-21-5p exosomes promoted ectopic formation in traumatized mice. This study confirms that HIF-1α could modulate miR-21-5p exosomes to promote post-traumatic ectopic bone formation by inducing ASCs cell differentiation. Our study reveals the mechanisms of miR-21-5p in ectopic ossification formation after trauma.
Collapse
Affiliation(s)
- Peng Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Bo Liu
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Chunhao Song
- Department of Medical Imaging, Weihai Wendeng District People Hospital, Weihai, 264200, China
| | - Jun Jia
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Yuanhao Wang
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Kai Pang
- Department of Operations Management, Wehai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Yitao Wang
- Department of Laboratory, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| | - Cong Chen
- Department of Spine Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, 264200, China
| |
Collapse
|
13
|
Liu F, Zhao Y, Pei Y, Lian F, Lin H. Role of the NF-kB signalling pathway in heterotopic ossification: biological and therapeutic significance. Cell Commun Signal 2024; 22:159. [PMID: 38439078 PMCID: PMC10910758 DOI: 10.1186/s12964-024-01533-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
Heterotopic ossification (HO) is a pathological process in which ectopic bone develops in soft tissues within the skeletal system. Endochondral ossification can be divided into the following types of acquired and inherited ossification: traumatic HO (tHO) and fibrodysplasia ossificans progressiva (FOP). Nuclear transcription factor kappa B (NF-κB) signalling is essential during HO. NF-κB signalling can drive initial inflammation through interactions with the NOD-like receptor protein 3 (NLRP3) inflammasome, Sirtuin 1 (SIRT1) and AMP-activated protein kinase (AMPK). In the chondrogenesis stage, NF-κB signalling can promote chondrogenesis through interactions with mechanistic target of rapamycin (mTOR), phosphatidylinositol-3-kinase (PI3K)/AKT (protein kinase B, PKB) and other molecules, including R-spondin 2 (Rspo2) and SRY-box 9 (Sox9). NF-κB expression can modulate osteoblast differentiation by upregulating secreted protein acidic and rich in cysteine (SPARC) and interacting with mTOR signalling, bone morphogenetic protein (BMP) signalling or integrin-mediated signalling under stretch stimulation in the final osteogenic stage. In FOP, mutated ACVR1-induced NF-κB signalling exacerbates inflammation in macrophages and can promote chondrogenesis and osteogenesis in mesenchymal stem cells (MSCs) through interactions with smad signalling and mTOR signalling. This review summarizes the molecular mechanism of NF-κB signalling during HO and highlights potential therapeutics for treating HO.
Collapse
Affiliation(s)
- Fangzhou Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yike Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yiran Pei
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Fengyu Lian
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
14
|
Jia L, Tian H, Sun S, Hao X, Wen Y. EID3 inhibits the osteogenic differentiation of periodontal ligament stem cells and mediates the signal transduction of TAZ-EID3-AKT/MTOR/ERK. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119662. [PMID: 38216090 DOI: 10.1016/j.bbamcr.2024.119662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/16/2023] [Accepted: 01/03/2024] [Indexed: 01/14/2024]
Abstract
Exploring the molecular mechanisms of cell behaviors is beneficial for promoting periodontal ligament stem cell (PDLSC)-mediated tissue regeneration. This study intends to explore the regulatory effects of EID3 on cell proliferation, apoptosis, and osteogenic differentiation and to preliminarily explore the regulatory mechanism of EID3. Here, EID3 was overexpressed or knocked down in PDLSCs by recombinant lentivirus. Then, cell proliferation activity was analyzed by colony-forming assay, EdU assay, and cell cycle assay. Cell apoptosis was detected by flow cytometry. The osteo-differentiation potential was analyzed using ALP activity assay, ALP staining, alizarin red staining, and mRNA and protein assay of osteo-differentiation related genes. The results showed that when EID3 was knocked down, the proliferation activity and osteogenic differentiation potential of PDLSCs decreased, while they increased when EID3 was overexpressed. The cell apoptosis rate decreased in PDLSCs with EID3 knockdown but increased in PDLSCs with EID3 overexpression. Moreover, EID3 inhibited the transduction of the AKT/MTOR and ERK signaling pathway. In addition, TAZ negatively regulated the expression of EID3, and the overexpression of EID3 partially reversed the promotive effects of TAZ on the osteogenic differentiation of PDLSCs. Taken together, EID3 inhibits the proliferation and osteogenic differentiation while promoting the apoptosis of PDLSCs. EID3 inhibits the transduction of the AKT/MTOR and ERK signaling pathways and mediates the regulatory effect of TAZ on PDLSC osteogenic differentiation.
Collapse
Affiliation(s)
- Linglu Jia
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China
| | - Hui Tian
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Department of Stomatology, Rizhao People's Hospital, Rizhao, Shandong, China
| | - Shaoqing Sun
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China
| | - Xingyao Hao
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China
| | - Yong Wen
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Shandong, China; Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration, Shandong, China; Shandong Provincial Clinical Research Center for Oral Diseases, Shandong, China.
| |
Collapse
|
15
|
Shi H, Yang Y, Xing H, Jia J, Xiong W, Guo S, Yang S. Exosomal non-coding RNAs: Emerging insights into therapeutic potential and mechanisms in bone healing. J Tissue Eng 2024; 15:20417314241286606. [PMID: 39371940 PMCID: PMC11456177 DOI: 10.1177/20417314241286606] [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: 08/11/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Exosomes are nano-sized extracellular vesicles (EVs) released by diverse types of cells, which affect the functions of targeted cells by transporting bioactive substances. As the main component of exosomes, non-coding RNA (ncRNA) is demonstrated to impact multiple pathways participating in bone healing. Herein, this review first introduces the biogenesis and secretion of exosomes, and elucidates the role of the main cargo in exosomes, ncRNAs, in mediating intercellular communication. Subsequently, the potential molecular mechanism of exosomes accelerating bone healing is elucidated from the following four aspects: macrophage polarization, vascularization, osteogenesis and osteoclastogenesis. Then, we systematically introduce construction strategies based on modified exosomes in bone regeneration field. Finally, the clinical trials of exosomes for bone healing and the challenges of exosome-based therapies in the biomedical field are briefly introduced, providing solid theoretical frameworks and optimization methods for the clinical application of exosomes in orthopedics.
Collapse
Affiliation(s)
- Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yang Yang
- Department of Rehabilitation, The First Hospital of China Medical University, Shenyang, China
| | - Hao Xing
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Jialin Jia
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Wei Xiong
- Department of Plastic Surgery, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, Xinjiang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
16
|
He YF, Wang XL, Deng SP, Wang YL, Huang QQ, Lin S, Lyu GR. Latest progress in low-intensity pulsed ultrasound for studying exosomes derived from stem/progenitor cells. Front Endocrinol (Lausanne) 2023; 14:1286900. [PMID: 38089611 PMCID: PMC10715436 DOI: 10.3389/fendo.2023.1286900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Stem cells have self-renewal, replication, and multidirectional differentiation potential, while progenitor cells are undifferentiated, pluripotent or specialized stem cells. Stem/progenitor cells secrete various factors, such as cytokines, exosomes, non-coding RNAs, and proteins, and have a wide range of applications in regenerative medicine. However, therapies based on stem cells and their secreted exosomes present limitations, such as insufficient source materials, mature differentiation, and low transplantation success rates, and methods addressing these problems are urgently required. Ultrasound is gaining increasing attention as an emerging technology. Low-intensity pulsed ultrasound (LIPUS) has mechanical, thermal, and cavitation effects and produces vibrational stimuli that can lead to a series of biochemical changes in organs, tissues, and cells, such as the release of extracellular bodies, cytokines, and other signals. These changes can alter the cellular microenvironment and affect biological behaviors, such as cell differentiation and proliferation. Here, we discuss the effects of LIPUS on the biological functions of stem/progenitor cells, exosomes, and non-coding RNAs, alterations involved in related pathways, various emerging applications, and future perspectives. We review the roles and mechanisms of LIPUS in stem/progenitor cells and exosomes with the aim of providing a deeper understanding of LIPUS and promoting research and development in this field.
Collapse
Affiliation(s)
- Yi-fang He
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xia-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| | - Shuang-ping Deng
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-li Wang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qing-qing Huang
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia
| | - Guo-rong Lyu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Departments of Medical Imaging, Quanzhou Medical College, Quanzhou, China
| |
Collapse
|
17
|
Deng S, Cao H, Cui X, Fan Y, Wang Q, Zhang X. Optimization of exosome-based cell-free strategies to enhance endogenous cell functions in tissue regeneration. Acta Biomater 2023; 171:68-84. [PMID: 37730080 DOI: 10.1016/j.actbio.2023.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023]
Abstract
Exosomes, nanoscale extracellular vesicles, play a crucial role in intercellular communication, owing to their biologically active cargoes such as RNAs and proteins. In recent years, they have emerged as a promising tool in the field of tissue regeneration, with the potential to initiate a new trend in cell-free therapy. However, it's worth noting that not all types of exosomes derived from cells are appropriate for tissue repair. Thus, selecting suitable cell sources is critical to ensure their efficacy in specific tissue regeneration processes. Current therapeutic applications of exosomes also encounter several limitations, including low-specific content for targeted diseases, non-tissue-specific targeting, and short retention time due to rapid clearance in vivo. Consequently, this review paper focuses on exosomes from diverse cell sources with functions specific to tissue regeneration. It also highlights the latest engineering strategies developed to overcome the functional limitations of natural exosomes. These strategies encompass the loading of specific therapeutic contents into exosomes, the endowment of tissue-specific targeting capability on the exosome surface, and the incorporation of biomaterials to extend the in vivo retention time of exosomes in a controlled-release manner. Collectively, these innovative approaches aim to synergistically enhance the therapeutic effects of natural exosomes, optimizing exosome-based cell-free strategies to boost endogenous cell functions in tissue regeneration. STATEMENT OF SIGNIFICANCE: Exosome-based cell-free therapy has recently emerged as a promising tool for tissue regeneration. This review highlights the characteristics and functions of exosomes from different sources that can facilitate tissue repair and their contributions to the regeneration process. To address the functional limitations of natural exosomes in therapeutic applications, this review provides an in-depth understanding of the latest engineering strategies. These strategies include optimizing exosomal contents, endowing tissue-specific targeting capability on the exosome surface, and incorporating biomaterials to extend the in vivo retention time of exosomes in a controlled-release manner. This review aims to explore and discuss innovative approaches that can synergistically improve endogenous cell functions in advanced exosome-based cell-free therapies for a broad range of tissue regeneration.
Collapse
Affiliation(s)
- Siyan Deng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Hongfu Cao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiaolin Cui
- School of medicine, the Chinese University of Hong Kong, Shenzhen, China; Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610065, China; College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan 610065, China
| |
Collapse
|
18
|
Review of Basic Research about Ossification of the Spinal Ligaments Focusing on Animal Models. J Clin Med 2023; 12:jcm12051958. [PMID: 36902744 PMCID: PMC10003841 DOI: 10.3390/jcm12051958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Ossification of the posterior longitudinal ligament (OPLL) is a heterotopic ossification that may cause spinal cord compression. With the recent development of computed tomography (CT) imaging, it is known that patients with OPLL often have complications related to ossification of other spinal ligaments, and OPLL is now considered part of ossification of the spinal ligaments (OSL). OSL is known to be a multifactorial disease with associated genetic and environmental factors, but its pathophysiology has not been clearly elucidated. To elucidate the pathophysiology of OSL and develop novel therapeutic strategies, clinically relevant and validated animal models are needed. In this review, we focus on animal models that have been reported to date and discuss their pathophysiology and clinical relevance. The purpose of this review is to summarize the usefulness and problems of existing animal models and to help further the development of basic research on OSL.
Collapse
|
19
|
Chen Z, Zhai J, Ma J, Chen P, Lin W, Zhang W, Xiong J, Zhang C, Wei H. Melatonin-Primed Mesenchymal Stem Cells-Derived Small Extracellular Vesicles Alleviated Neurogenic Erectile Dysfunction by Reversing Phenotypic Modulation. Adv Healthc Mater 2023; 12:e2203087. [PMID: 36652551 DOI: 10.1002/adhm.202203087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/10/2023] [Indexed: 01/19/2023]
Abstract
Erectile dysfunction (ED) is an adverse side effect of pelvic surgery with no effective treatment. In this study, it is explored whether melatonin could improve the therapeutic effects of small extracellular vesicles (sEVs), derived from mesenchymal stem cells (MSCs), on cavernous nerve injury (CNI) ED, and the underlying mechanisms are investigated. The sEVs from melatonin-pretreated MSCs (MT-EVs) and MSCs (NC-EVs) are isolated and applied to CNI ED. Transplantation of MT-EVs remarkably increases erectile function and reduces phenotypic modulation in CNI ED rats. The therapeutic effects of MT-EVs are superior to those of NC-EVs. Sequencing implies that miR-10a-3p is enriched in MT-EVs, and directly targets the protein kinase inhibitor α (PKIA). After the suppression of miR-10a-3p, the therapeutic actions of MT-EVs are abolished, but are rescued by PKIA. Similarly, RhoA/ROCK is inhibited by MT-EVs, but this action is reversed by suppressing miR-10a-3p, accompanied by corresponding changes in PKIA. In conclusion, transplantation of MT-EVs could significantly alleviate CNI ED. MT-EVs may relieve the phenotypic modulation of the corpora cavernosum smooth muscle cells via the miR-10a-3p/PKIA/RhoA/ROCK signaling axis. These nanovesicles should be potential therapeutic vectors or bioactive materials for CNI ED.
Collapse
Affiliation(s)
- Zehong Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Jiancheng Zhai
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Jiahui Ma
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Peng Chen
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Weishun Lin
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Weipeng Zhang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Jiaming Xiong
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Chaowei Zhang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| | - Hongbo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Tianhe Road 600, Guangzhou, 510630, China
| |
Collapse
|