1
|
Zhang L, Zeng C, Huang J, Yan H, Jiang Y, Li R. Exploration of the miR-187-3p/CNR2 pathway in modulating osteoblast differentiation and treating postmenopausal osteoporosis through mechanical stress. FASEB J 2024; 38:e23776. [PMID: 38958998 DOI: 10.1096/fj.202400113rr] [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: 01/20/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
This study aimed to explore how mechanical stress affects osteogenic differentiation via the miR-187-3p/CNR2 pathway. To conduct this study, 24 female C57BL/6 mice, aged 8 weeks, were used and divided into four groups. The Sham and OVX groups did not undergo treadmill exercise, while the Sham + EX and OVX + EX groups received a 8-week treadmill exercise. Post-training, bone marrow and fresh femur samples were collected for further analysis. Molecular biology analysis, histomorphology analysis, and micro-CT analysis were conducted on these samples. Moreover, primary osteoblasts were cultured under osteogenic conditions and divided into GM group and CTS group. The cells in the CTS group underwent a sinusoidal stretching regimen for either 3 or 7 days. The expression of early osteoblast markers (Runx2, OPN, and ALP) was measured to assess differentiation. The study findings revealed that mechanical stress has a regulatory impact on osteoblast differentiation. The expression of miR-187-3p was observed to decrease, facilitating osteogenic differentiation, while the expression of CNR2 increased significantly. These observations suggest that mechanical stress, miR-187-3p, and CNR2 play crucial roles in regulating osteogenic differentiation. Both in vivo and in vitro experiments have confirmed that mechanical stress downregulates miR-187-3p and upregulates CNR2, which leads to the restoration of distal femoral bone mass and enhancement of osteoblast differentiation. Therefore, mechanical stress promotes osteoblasts, resulting in improved osteoporosis through the miR-187-3p/CNR2 signaling pathway. These findings have broad prospect and provide molecular biology guidance for the basic research and clinical application of exercise in the prevention and treatment of PMOP.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou, China
- Academy of Orthopedics·Guangdong Province, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Canjun Zeng
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou, China
- Academy of Orthopedics·Guangdong Province, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Juanyu Huang
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou, China
- Academy of Orthopedics·Guangdong Province, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Haohang Yan
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou, China
- Academy of Orthopedics·Guangdong Province, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yutao Jiang
- Orthopaedic Hospital of Guangdong Province, Guangzhou, China
- Academy of Orthopedics·Guangdong Province, Guangzhou, China
- Guangzhou Key Laboratory of Neuropathic Pain Mechanism at Spinal Cord Level, Guangzhou, China
- Department of Spine Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Runguang Li
- Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou, China
- Academy of Orthopedics·Guangdong Province, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| |
Collapse
|
2
|
Hu C, Yang Q, Huang X, Wang F, Zhou H, Su X. Three-Dimensional Mechanical Microenvironment Rescued the Decline of Osteogenic Differentiation of Old Human Jaw Bone Marrow Mesenchymal Stem Cells. ACS Biomater Sci Eng 2024; 10:4496-4509. [PMID: 38860704 DOI: 10.1021/acsbiomaterials.4c00680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
Resorption and atrophy of the alveolar bone, as two consequences of osteoporosis that remarkably complicate the orthodontic and prosthodontic treatments, contribute to the differentiated biological features and force-induced response of jaw bone marrow-derived mesenchymal stem cells (JBMSCs) in elderly patients. We isolated and cultured JBMSCs from adolescent and adult patients and then simulated the loading of orthodontic tension stress by constructing an in vitro three-dimensional (3D) stress loading model. The decline in osteogenic differentiation of aged JBMSCs was reversed by tensile stress stimulation. It is interesting to note that tension stimulation had a stronger effect on the osteogenic differentiation of elderly JBMSCs compared to the young ones, indicating a possible mechanism of aging rescue. High-throughput sequencing of microRNA (miRNAs) was subsequently performed before and after tension stimulation in all JBMSCs, followed by the comprehensive comparison of mechanically responsive miRNAs in the 3D strain microenvironment. The results suggested a significant reduction in the expression of miR-210-3p and miR-214-3p triggered by the 3D strain microenvironment in old-JBMSCs. Bioinformatic analysis indicated that both miRNAs participate in the regulation of critical pathways of aging and cellular senescence. Taken together, this study demonstrated that the 3D strain microenvironment efficiently rescued the cellular senescence of old-JBMSCs via modulating specific miRNAs, which provides a novel strategy for coordinating periodontal bone loss and regeneration of the elderly.
Collapse
Affiliation(s)
- Cheng Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Hospital of Stomatology & Guangdong Provincial Key Laboratory of Stomatology & Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Qiyuan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaojun Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fei Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Hong Zhou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiaoxia Su
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
3
|
Nugraha AP, Narmada IB, Winoto ER, Ardani IGAW, Triwardhani A, Alida A, Pramusita A, Nur RM, Indrastie N, Nam HY, Ihsan IS, Riawan W, Rantam FA, Nugraha AP, Noor TNEBTA. Gingiva Mesenchymal Stem Cells Normoxic or Hypoxic Preconditioned Application Under Orthodontic Mechanical Force on Osterix, Osteopontin, and ALP Expression. Eur J Dent 2024; 18:501-509. [PMID: 37995729 PMCID: PMC11132784 DOI: 10.1055/s-0043-1772699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023] Open
Abstract
OBJECTIVES The aim of this article was to investigate Osterix, ALP, and osteopontin expression in the compression and tension sides of alveolar bone after the application of normoxic/hypoxic-preconditioned GMSCs in rabbits (Oryctolagus cuniculus) induced with OMF. MATERIALS AND METHODS Forty-eight healthy, young male rabbits were divided into four groups: [-] OMF; [+] OMF; OMF with GMSCs normoxic-preconditioned; and OMF and GMSCs hypoxic-preconditioned. The central incisor and left mandibular molar in the experimental animals were moved, the mandibular first molar was moved mesially using nickel titanium (NiTi) and stainless steel ligature wire connected to a 50 g/mm2 light force closed coil spring. Allogeneic application of normoxic or hypoxic-preconditioned GMSCs was used in as many as 106 cells in a 20 µL phosphate buffered saline single dose and injected into experimental animals' gingiva after 1 day of OTM. On days 7, 14, and 28, all experimental animals were euthanized. Osterix, ALP, and osteopontin expressions were examined by immunohistochemistry. RESULTS Osterix, ALP, and osteopontin expressions were significantly different after allogeneic application of hypoxic-preconditioned GMSCs than normoxic-preconditioned GMSCs in the tension and compression of the alveolar bone side during OMF (p < 0.05). CONCLUSION Osterix, ALP, and osteopontin expressions were significantly more enhanced post-transplantation of GMSCs with hypoxic-preconditioning than after transplantation of normoxic-preconditioned GMSCs in rabbits (O. cuniculus) induced with OMF.
Collapse
Affiliation(s)
- Alexander Patera Nugraha
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ida Bagus Narmada
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ervina Restiwulan Winoto
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - I Gusti Aju Wahju Ardani
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ari Triwardhani
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Alida Alida
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Adya Pramusita
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Reyhan Mahendra Nur
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Nuraini Indrastie
- Department of Orthodontics, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Hui Yin Nam
- Nanotechnology and Catalysis Research Centre (NANOCAT), Universiti Malaya, Kuala Lumpur, Malaysia
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Igo Syaiful Ihsan
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, Indonesia
| | - Wibi Riawan
- Biomolecular Biochemistry, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Fedik Abdul Rantam
- Laboratory of Immunology and Virology Department of Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Tengku Natasha Eleena binti Tengku Ahmad Noor
- Membership of Faculty of Dental Surgery, Royal Collage of Surgeon, Edinburgh University, United Kingdom
- Malaysian Armed Forces Dental Officer, 609 Armed Forces Dental Clinic, Kem Semenggo, Kuching, Sarawak, Malaysia
| |
Collapse
|
4
|
Ren H, Zhang L, Zhang X, Yi C, Wu L. Specific lipid magnetic sphere sorted CD146-positive bone marrow mesenchymal stem cells can better promote articular cartilage damage repair. BMC Musculoskelet Disord 2024; 25:253. [PMID: 38561728 PMCID: PMC10983655 DOI: 10.1186/s12891-024-07381-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND The characteristics and therapeutic potential of subtypes of bone marrow mesenchymal stem cells (BMSCs) are largely unknown. Also, the application of subpopulations of BMSCs in cartilage regeneration remains poorly characterized. The aim of this study was to explore the regenerative capacity of CD146-positive subpopulations of BMSCs for repairing cartilage defects. METHODS CD146-positive BMSCs (CD146 + BMSCs) were sorted by self-developed CD146-specific lipid magnetic spheres (CD146-LMS). Cell surface markers, viability, and proliferation were evaluated in vitro. CD146 + BMSCs were subjected to in vitro chondrogenic induction and evaluated for chondrogenic properties by detecting mRNA and protein expression. The role of the CD146 subpopulation of BMSCs in cartilage damage repair was assessed by injecting CD146 + BMSCs complexed with sodium alginate gel in the joints of a mouse cartilage defect model. RESULTS The prepared CD146-LMS had an average particle size of 193.7 ± 5.24 nm, an average potential of 41.9 ± 6.21 mv, and a saturation magnetization intensity of 27.2 Am2/kg, which showed good stability and low cytotoxicity. The sorted CD146 + BMSCs highly expressed stem cell and pericyte markers with good cellular activity and cellular value-added capacity. Cartilage markers Sox9, Collagen II, and Aggrecan were expressed at both protein and mRNA levels in CD146 + BMSCs cells after chondrogenic induction in vitro. In a mouse cartilage injury model, CD146 + BMSCs showed better function in promoting the repair of articular cartilage injury. CONCLUSION The prepared CD146-LMS was able to sort out CD146 + BMSCs efficiently, and the sorted subpopulation of CD146 + BMSCs had good chondrogenic differentiation potential, which could efficiently promote the repair of articular cartilage injury, suggesting that the sorted CD146 + BMSCs subpopulation is a promising seed cell for cartilage tissue engineering.
Collapse
Affiliation(s)
- Hanru Ren
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China
| | - Lele Zhang
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China
| | - Xu Zhang
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China
| | - Chengqing Yi
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China.
| | - Lianghao Wu
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, No. 2800, Gongwei Road, Shanghai, 200120, China.
| |
Collapse
|
5
|
Li Z, Yang B, Yang Z, Xie X, Guo Z, Zhao J, Wang R, Fu H, Zhao P, Zhao X, Chen G, Li G, Wei F, Bian L. Supramolecular Hydrogel with Ultra-Rapid Cell-Mediated Network Adaptation for Enhancing Cellular Metabolic Energetics and Tissue Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307176. [PMID: 38295393 DOI: 10.1002/adma.202307176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/27/2023] [Indexed: 02/02/2024]
Abstract
Cellular energetics plays an important role in tissue regeneration, and the enhanced metabolic activity of delivered stem cells can accelerate tissue repair and regeneration. However, conventional hydrogels with limited network cell adaptability restrict cell-cell interactions and cell metabolic activities. In this work, it is shown that a cell-adaptable hydrogel with high network dynamics enhances the glucose uptake and fatty acid β-oxidation of encapsulated human mesenchymal stem cells (hMSCs) compared with a hydrogel with low network dynamics. It is further shown that the hMSCs encapsulated in the high dynamic hydrogels exhibit increased tricarboxylic acid (TCA) cycle activity, oxidative phosphorylation (OXPHOS), and adenosine triphosphate (ATP) biosynthesis via an E-cadherin- and AMP-activated protein kinase (AMPK)-dependent mechanism. The in vivo evaluation further showed that the delivery of MSCs by the dynamic hydrogel enhanced in situ bone regeneration in an animal model. It is believed that the findings provide critical insights into the impact of stem cell-biomaterial interactions on cellular metabolic energetics and the underlying mechanisms.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, P. R. China
| | - Boguang Yang
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, 999077, P. R. China
| | - Zhengmeng Yang
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, 999077, P. R. China
| | - Xian Xie
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, 999077, P. R. China
| | - Zhengnan Guo
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Jianyang Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Ruinan Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Hao Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P. R. China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, P. R. China
| | - Guosong Chen
- Department of Macromolecular Science, Fudan University, Shanghai, 200433, P. R. China
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, 999077, P. R. China
| | - Fuxin Wei
- Department of Orthopedic Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, P. R. China
- Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, Shenzhen, 518107, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 511442, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 511442, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 511442, P. R. China
| |
Collapse
|
6
|
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
|
7
|
Li JY, Wang TT, Ma L, Zhang Y, Zhu D. Silencing of Jumonji domain-containing 1C inhibits the osteogenic differentiation of bone marrow mesenchymal stem cells via nuclear factor-κB signaling. World J Stem Cells 2024; 16:151-162. [PMID: 38455099 PMCID: PMC10915961 DOI: 10.4252/wjsc.v16.i2.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Osteoporosis is a common metabolic bone disorder induced by an imbalance between osteoclastic activity and osteogenic activity. During osteoporosis, bone mesenchymal stem cells (BMSCs) exhibit an increased ability to differentiate into adipocytes and a decreased ability to differentiate into osteoblasts, resulting in bone loss. Jumonji domain-containing 1C (JMJD1C) has been demonstrated to suppress osteoclastogenesis. AIM To examine the effect of JMJD1C on the osteogenesis of BMSCs and the potential underlying mechanism. METHODS BMSCs were isolated from mouse bone marrow tissues. Oil Red O staining, Alizarin red staining, alkaline phosphatase staining and the expression of adipogenic and osteogenic-associated genes were assessed to determine the differentiation of BMSCs. Bone marrow-derived macrophages (BMMs) were incubated with receptor activator of nuclear factor-kappa Β ligand to induce osteoclast differentiation, and osteoclast differentiation was confirmed by tartrate-resistant acid phosphatase staining. Other related genes were measured via reverse transcription coupled to the quantitative polymerase chain reaction and western blotting. Enzyme-linked immunosorbent assays were used to measure the levels of inflammatory cytokines, including tumor necrosis factor alpha, interleukin-6 and interleukin-1 beta. RESULTS The osteogenic and adipogenic differentiation potential of BMSCs isolated from mouse bone marrow samples was evaluated. JMJD1C mRNA and protein expression was upregulated in BMSCs after osteoblast induction, while p-nuclear factor-κB (NF-κB) and inflammatory cytokines were not significantly altered. Knockdown of JMJD1C repressed osteogenic differentiation and enhanced NF-κB activation and inflammatory cytokine release in BMSCs. Moreover, JMJD1C expression decreased during BMM osteoclast differentiation. CONCLUSION The JMJD1C/NF-κB signaling pathway is potentially involved in BMSC osteogenic differentiation and may play vital roles in the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Jing-Yi Li
- Department of Medical Cosmetology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Ting-Ting Wang
- Department of General Gerontology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Li Ma
- Department of Plastic Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yu Zhang
- Senior Department of Hematology, The Fifth Medical Centre, General Hospital of Chinese People's Liberation Army, Beijing 100071, China
| | - Di Zhu
- Department of Orthopaedic Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| |
Collapse
|
8
|
Zhu Z, Wang Z, Ma C, Zhou J, Zhang W. Isopsoralen promotes osteogenic differentiation of human jawbone marrow mesenchymal cells through Notch signaling pathway. Ann Anat 2023; 250:152156. [PMID: 37678499 DOI: 10.1016/j.aanat.2023.152156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/15/2023] [Accepted: 08/12/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The aim of this study was to investigate the effect of isopsoralen on osteogenic differentiation of human jawbone marrow mesenchymal cells and its possible mechanism. METHOD The cytotoxicity and proliferation of cells were measured by a cell counting kit 8. Alkaline phosphatase activity analysis was then used to determine the optimal concentration of isopsoralen to promote the differentiation. Western blot, qRT-PCR and Alizarin Red S staining were used to evaluate the role of Notch signaling pathway in isopsoralen-induced osteogenic differentiation. This study also investigated the anti-osteoporotic effects of ISO using in vivo osteoporosis models. RESULTS Our results showed that 1 × 10-6 mol / L isopsoralen can effectively promote the proliferation and osteogenic differentiation of cells. Moreover, we found that activation of notch signaling pathway inhibited isopsoralen-induced osteogenesis and inhibition of Notch signal promoted the differentiation of osteoblasts induced by isopsoralen. In vivo experiments revealed that ISO significantly inhibited OVX-induced bone mineral density loss and restored the impaired bone structural properties in osteoporosis model mice. CONCLUSION Our findings demonstrated that isopsoralen induced osteogenic differentiation by inhibiting Notch signaling and it might be a potential therapeutic agent for treating or preventing osteoporosis.
Collapse
Affiliation(s)
- Zhu Zhu
- Stomatology outpatient of the Air Force From Eastern Theater of PLA, Nanjing, Jiangsu, China
| | - Zitian Wang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Changyan Ma
- Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junbo Zhou
- Department of Stomatology, Nanjing Integrated Traditional Chinese and Western Medicine Hospital, Affiliated with Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, China; Department of Oral Special Consultation, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
9
|
Niu J, Feng F, Zhang S, Zhu Y, Song R, Li J, Zhao L, Wang H, Zhao Y, Zhang M. Thrombospondin-2 Couples Pressure-Promoted Chondrogenesis through NF-κB Signaling. Tissue Eng Regen Med 2023; 20:753-766. [PMID: 37219820 PMCID: PMC10352201 DOI: 10.1007/s13770-023-00548-7] [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: 12/31/2022] [Revised: 03/03/2023] [Accepted: 04/20/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Our previous studies found that the mechanical stimulation promote chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), along with up-regulation of thrombospondin-2 (TSP-2). The aim of this study was to explore the effect of thrombospondin-2 (TSP-2) on the mechanical pressure-stimulated chondrogenic differentiation of BMSCs and the possible role of NF-κB signaling in the mechano-chemical coupling regulation toward chondrogenesis. METHODS Rat BMSCs were isolated, cultured and identified. The time-dependent expressions of TSP-2 and Sox9 in BMSCs under a dynamic mechanical pressure of 0-120 kPa at 0.1 Hz for 1 h were tested by qPCR and Western blotting. The role of TSP-2 in chondrogenic differentiation of BMSCs under mechanical pressure was validated by using small interfering RNA. The impact of TSP-2 and mechanical pressure on chondrogenesis were detected and the downstream signaling molecules were explored using Western blotting. RESULTS Mechanical pressure stimulation of 0-120 kPa for 1 h significantly upregulated the expression of TSP-2 in BMSCs. The expression of the chondrogenesis markers Sox9, Aggrecan, and Col-II were all upregulated under dynamic mechanical pressure or TSP-2 stimulation. Additional exogenous TSP-2 may potentiate the chondrogenic effect of mechanical stimulation. After knock down TSP-2, the upregulation of Sox9, Aggrecan and Col-II under mechanical pressure was inhibited. The NF-κB signaling pathway responded to both dynamic pressure and TSP-2 stimulation, and the cartilage-promoting effect was blocked by an NF-κB signaling inhibitor. CONCLUSION TSP-2 plays an essential role in the chondrogenic differentiation of BMSCs under mechanical pressure. NF-κB signaling is involved in the mechano-chemical coupling of TSP-2 and mechanical pressure for the chondrogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Jing Niu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
- The College of Life Sciences and Medicine, Northwest University, Xi'an, People's Republic of China
| | - Fan Feng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Songbai Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yue Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Runfang Song
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Junrong Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Liang Zhao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Hui Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Ying Zhao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
- Department of Anesthesiology and Perioperative Medicine, Xi'an People's Hospital (Xi'an Fourth Hospital), Northwest University, Xi'an, 710004, People's Republic of China.
| | - Min Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Disease, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| |
Collapse
|
10
|
Pu P, Wu S, Zhang K, Xu H, Guan J, Jin Z, Sun W, Zhang H, Yan B. Mechanical force induces macrophage-derived exosomal UCHL3 promoting bone marrow mesenchymal stem cell osteogenesis by targeting SMAD1. J Nanobiotechnology 2023; 21:88. [PMID: 36915132 PMCID: PMC10012474 DOI: 10.1186/s12951-023-01836-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/02/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Orthodontic tooth movement (OTM), a process of alveolar bone remodelling, is induced by mechanical force and regulated by local inflammation. Bone marrow-derived mesenchymal stem cells (BMSCs) play a fundamental role in osteogenesis during OTM. Macrophages are mechanosensitive cells that can regulate local inflammatory microenvironment and promote BMSCs osteogenesis by secreting diverse mediators. However, whether and how mechanical force regulates osteogenesis during OTM via macrophage-derived exosomes remains elusive. RESULTS Mechanical stimulation (MS) promoted bone marrow-derived macrophage (BMDM)-mediated BMSCs osteogenesis. Importantly, when exosomes from mechanically stimulated BMDMs (MS-BMDM-EXOs) were blocked, the pro-osteogenic effect was suppressed. Additionally, compared with exosomes derived from BMDMs (BMDM-EXOs), MS-BMDM-EXOs exhibited a stronger ability to enhance BMSCs osteogenesis. At in vivo, mechanical force-induced alveolar bone formation was impaired during OTM when exosomes were blocked, and MS-BMDM-EXOs were more effective in promoting alveolar bone formation than BMDM-EXOs. Further proteomic analysis revealed that ubiquitin carboxyl-terminal hydrolase isozyme L3 (UCHL3) was enriched in MS-BMDM-EXOs compared with BMDM-EXOs. We went on to show that BMSCs osteogenesis and mechanical force-induced bone formation were impaired when UCHL3 was inhibited. Furthermore, mothers against decapentaplegic homologue 1 (SMAD1) was identified as the target protein of UCHL3. At the mechanistic level, we showed that SMAD1 interacted with UCHL3 in BMSCs and was downregulated when UCHL3 was suppressed. Consistently, overexpression of SMAD1 rescued the adverse effect of inhibiting UCHL3 on BMSCs osteogenesis. CONCLUSIONS This study suggests that mechanical force-induced macrophage-derived exosomal UCHL3 promotes BMSCs osteogenesis by targeting SMAD1, thereby promoting alveolar bone formation during OTM.
Collapse
Affiliation(s)
- Panjun Pu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Shengnan Wu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Kejia Zhang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Hao Xu
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Jiani Guan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Zhichun Jin
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China
| | - Wen Sun
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China
| | - Hanwen Zhang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 210000, China.
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, 210000, China.
| | - Bin Yan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, 210000, China.
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210000, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, 210000, China.
| |
Collapse
|
11
|
Ma Q, Miri Z, Haugen HJ, Moghanian A, Loca D. Significance of mechanical loading in bone fracture healing, bone regeneration, and vascularization. J Tissue Eng 2023; 14:20417314231172573. [PMID: 37251734 PMCID: PMC10214107 DOI: 10.1177/20417314231172573] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
In 1892, J.L. Wolff proposed that bone could respond to mechanical and biophysical stimuli as a dynamic organ. This theory presents a unique opportunity for investigations on bone and its potential to aid in tissue repair. Routine activities such as exercise or machinery application can exert mechanical loads on bone. Previous research has demonstrated that mechanical loading can affect the differentiation and development of mesenchymal tissue. However, the extent to which mechanical stimulation can help repair or generate bone tissue and the related mechanisms remain unclear. Four key cell types in bone tissue, including osteoblasts, osteoclasts, bone lining cells, and osteocytes, play critical roles in responding to mechanical stimuli, while other cell lineages such as myocytes, platelets, fibroblasts, endothelial cells, and chondrocytes also exhibit mechanosensitivity. Mechanical loading can regulate the biological functions of bone tissue through the mechanosensor of bone cells intraosseously, making it a potential target for fracture healing and bone regeneration. This review aims to clarify these issues and explain bone remodeling, structure dynamics, and mechano-transduction processes in response to mechanical loading. Loading of different magnitudes, frequencies, and types, such as dynamic versus static loads, are analyzed to determine the effects of mechanical stimulation on bone tissue structure and cellular function. Finally, the importance of vascularization in nutrient supply for bone healing and regeneration was further discussed.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
- Department of Immunology, School of
Basic Medicine, Fourth Military Medical University, Xi’an, PR China
| | - Zahra Miri
- Department of Materials Engineering,
Isfahan University of Technology, Isfahan, Iran
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
| | - Amirhossein Moghanian
- Department of Materials Engineering,
Imam Khomeini International University, Qazvin, Iran
| | - Dagnjia Loca
- Rudolfs Cimdins Riga Biomaterials
Innovations and Development Centre, Institute of General Chemical Engineering,
Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga,
Latvia
- Baltic Biomaterials Centre of
Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
12
|
Bratengeier C, Bakker AD, Liszka A, Schilcher J, Fahlgren A. The release of osteoclast-stimulating factors on supraphysiological loading by osteoprogenitors coincides with expression of genes associated with inflammation and cytoskeletal arrangement. Sci Rep 2022; 12:21578. [PMID: 36517534 PMCID: PMC9751069 DOI: 10.1038/s41598-022-25567-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Supraphysiological loading induced by unstable orthopedic implants initiates osteoclast formation, which results in bone degradation. We aimed to investigate which mechanosensitive cells in the peri-implant environment produce osteoclast-stimulating factors and how the production of these factors is stimulated by supraphysiological loading. The release of osteoclast-stimulating factors by different types of isolated bone marrow-derived hematopoietic and mesenchymal stem cells from six osteoarthritic patients was analyzed after one hour of supraphysiological loading (3.0 ± 0.2 Pa, 1 Hz) by adding their conditioned medium to osteoclast precursors. Monocytes produced factors that enhanced osteoclastogenesis by 1.6 ± 0.07-fold and mesenchymal stem cells by 1.4 ± 0.07-fold. Medium from osteoprogenitors and pre-osteoblasts enhanced osteoclastogenesis by 1.3 ± 0.09-fold and 1.4 ± 0.03-fold, respectively, where medium from four patients elicited a response and two did not. Next generation sequencing analysis of osteoprogenitors revealed that genes encoding for inflammation-related pathways and cytoskeletal rearrangements were regulated differently between responders and non-responders. Our data suggest that released osteoclast-stimulating soluble factors by progenitor cells in the bone marrow after supraphysiological loading may be related to cytoskeletal arrangement in an inflammatory environment. This connection could be relevant to better understand the aseptic loosening process of orthopedic implants.
Collapse
Affiliation(s)
- Cornelia Bratengeier
- Department of Biomedical and Clinical Sciences, Division of Cell Biology, Linköping University, Linköping, Sweden.
| | - Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Aneta Liszka
- Department of Biomedical and Clinical Sciences, Division of Cell Biology, Linköping University, Linköping, Sweden
| | - Jörg Schilcher
- Department of Orthopedics and Department of Biomedical and Clinical Sciences, Faculty of Health Sciences and the Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Anna Fahlgren
- Department of Biomedical and Clinical Sciences, Division of Cell Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
13
|
Zhai M, Cui S, Li L, Cheng C, Zhang Z, Liu J, Wei F. Mechanical Force Modulates Alveolar Bone Marrow Mesenchymal Cells Characteristics for Bone Remodeling during Orthodontic Tooth Movement through Lactate Production. Cells 2022; 11:cells11233724. [PMID: 36496983 PMCID: PMC9738738 DOI: 10.3390/cells11233724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Orthodontic tooth movement (OTM) relies on mechanical force-induced bone remodeling. As a metabolic intermediate of glycolysis, lactate has recently been discovered to participate in bone remodeling by serving as a signaling molecule. However, whether lactate could respond to mechanical stimulus during OTM, as well as whether lactate has an impact on the alveolar bone remodeling during orthodontics, remain to be further elucidated. In the current study, we observed physiologically elevated production of lactate along with increased osteogenic differentiation, proliferation, and migration of alveolar bone marrow mesenchymal cells (ABMMCs) under mechanical force. Inhibition of lactate, induced by cyclic mechanical stretch by GNE-140, remarkably suppressed the osteogenic differentiation, proliferation, and migration, yet enhanced apoptosis of ABMMCs. Mechanistically, these regulatory effects of lactate were mediated by histone lactylation. Taken together, our results suggest that force-induced lactate is involved in controlling bone remodeling-related cellular activities in ABMMCs and plays a vital role in the alveolar bone remodeling during OTM. Our findings indicate that lactate might be a critical modulator for alveolar bone remodeling during OTM, providing a novel therapeutic target for the purpose of more effectively controlling tooth movement and improving the stability of orthodontic results.
Collapse
|
14
|
Limraksasin P, Nattasit P, Manokawinchoke J, Tiskratok W, Vinaikosol N, Okawa H, Limjeerajarus CN, Limjeerajarus N, Pavasant P, Osathanon T, Egusa H. Application of shear stress for enhanced osteogenic differentiation of mouse induced pluripotent stem cells. Sci Rep 2022; 12:19021. [PMID: 36347883 PMCID: PMC9643422 DOI: 10.1038/s41598-022-21479-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/27/2022] [Indexed: 11/10/2022] Open
Abstract
The self-organizing potential of induced pluripotent stem cells (iPSCs) represents a promising tool for bone tissue engineering. Shear stress promotes the osteogenic differentiation of mesenchymal stem cells, leading us to hypothesize that specific shear stress could enhance the osteogenic differentiation of iPSCs. For osteogenesis, embryoid bodies were formed for two days and then maintained in medium supplemented with retinoic acid for three days, followed by adherent culture in osteogenic induction medium for one day. The cells were then subjected to shear loading (0.15, 0.5, or 1.5 Pa) for two days. Among different magnitudes tested, 0.5 Pa induced the highest levels of osteogenic gene expression and greatest mineral deposition, corresponding to upregulated connexin 43 (Cx43) and phosphorylated Erk1/2 expression. Erk1/2 inhibition during shear loading resulted in decreased osteogenic gene expression and the suppression of mineral deposition. These results suggest that shear stress (0.5 Pa) enhances the osteogenic differentiation of iPSCs, partly through Cx43 and Erk1/2 signaling. Our findings shed light on the application of shear-stress technology to improve iPSC-based tissue-engineered bone for regenerative bone therapy.
Collapse
Affiliation(s)
- Phoonsuk Limraksasin
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan ,grid.7922.e0000 0001 0244 7875Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Rd. Pathumwan, Bangkok, 10330 Thailand
| | - Praphawi Nattasit
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Jeeranan Manokawinchoke
- grid.7922.e0000 0001 0244 7875Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Watcharaphol Tiskratok
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Naruephorn Vinaikosol
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Hiroko Okawa
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan
| | - Chalida Nakalekha Limjeerajarus
- grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Nuttapol Limjeerajarus
- grid.7922.e0000 0001 0244 7875Office of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand ,grid.512238.f0000 0004 0625 2348Research Center for Advanced Energy Technology, Faculty of Engineering, Thai-Nichi Institute of Technology, Bangkok, Thailand
| | - Prasit Pavasant
- grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Rd. Pathumwan, Bangkok, 10330 Thailand
| | - Thanaphum Osathanon
- grid.7922.e0000 0001 0244 7875Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand ,grid.7922.e0000 0001 0244 7875Center of Excellence for Regenerative Dentistry and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Rd. Pathumwan, Bangkok, 10330 Thailand
| | - Hiroshi Egusa
- grid.69566.3a0000 0001 2248 6943Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575 Japan ,grid.69566.3a0000 0001 2248 6943Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, 4-1 Seiryo-machi, Aoba-ku, Sendai-city, 980-8575 Japan
| |
Collapse
|
15
|
Zhang X, Zhang S, Wang T. How the mechanical microenvironment of stem cell growth affects their differentiation: a review. Stem Cell Res Ther 2022; 13:415. [PMID: 35964140 PMCID: PMC9375355 DOI: 10.1186/s13287-022-03070-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 07/20/2022] [Indexed: 12/18/2022] Open
Abstract
Stem cell differentiation is of great interest in medical research; however, specifically and effectively regulating stem cell differentiation is still a challenge. In addition to chemical factors, physical signals are an important component of the stem cell ecotone. The mechanical microenvironment of stem cells has a huge role in stem cell differentiation. Herein, we describe the knowledge accumulated to date on the mechanical environment in which stem cells exist, which consists of various factors, including the extracellular matrix and topology, substrate stiffness, shear stress, hydrostatic pressure, tension, and microgravity. We then detail the currently known signalling pathways that stem cells use to perceive the mechanical environment, including those involving nuclear factor-kB, the nicotinic acetylcholine receptor, the piezoelectric mechanosensitive ion channel, and hypoxia-inducible factor 1α. Using this information in clinical settings to treat diseases is the goal of this research, and we describe the progress that has been made. In this review, we examined the effects of mechanical factors in the stem cell growth microenvironment on stem cell differentiation, how mechanical signals are transmitted to and function within the cell, and the influence of mechanical factors on the use of stem cells in clinical applications.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Sibo Zhang
- China Medical University, Shenyang, China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China.
| |
Collapse
|
16
|
Liu P, Tu J, Wang W, Li Z, Li Y, Yu X, Zhang Z. Effects of Mechanical Stress Stimulation on Function and Expression Mechanism of Osteoblasts. Front Bioeng Biotechnol 2022; 10:830722. [PMID: 35252138 PMCID: PMC8893233 DOI: 10.3389/fbioe.2022.830722] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoclasts and osteoblasts play a major role in bone tissue homeostasis. The homeostasis and integrity of bone tissue are maintained by ensuring a balance between osteoclastic and osteogenic activities. The remodeling of bone tissue is a continuous ongoing process. Osteoclasts mainly play a role in bone resorption, whereas osteoblasts are mainly involved in bone remodeling processes, such as bone cell formation, mineralization, and secretion. These cell types balance and restrict each other to maintain bone tissue metabolism. Bone tissue is very sensitive to mechanical stress stimulation. Unloading and loading of mechanical stress are closely related to the differentiation and formation of osteoclasts and bone resorption function as well as the differentiation and formation of osteoblasts and bone formation function. Consequently, mechanical stress exerts an important influence on the bone microenvironment and bone metabolism. This review focuses on the effects of different forms of mechanical stress stimulation (including gravity, continuously compressive pressure, tensile strain, and fluid shear stress) on osteoclast and osteoblast function and expression mechanism. This article highlights the involvement of osteoclasts and osteoblasts in activating different mechanical transduction pathways and reports changings in their differentiation, formation, and functional mechanism induced by the application of different types of mechanical stress to bone tissue. This review could provide new ideas for further microscopic studies of bone health, disease, and tissue damage reconstruction.
Collapse
Affiliation(s)
- Pan Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Ji Tu
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Wenzhao Wang
- Department of Orthopedics, West China Hospital of Sichuan University, Chengdu, China
| | - Zheng Li
- People’s Hospital of Jiulongpo District, Chongqing, China
| | - Yao Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaoping Yu
- School of Public Health, Chengdu Medical College, Chengdu, China
- Basic Medical College of Chengdu University, Chengdu, China
- *Correspondence: Xiaoping Yu, ; Zhengdong Zhang,
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- *Correspondence: Xiaoping Yu, ; Zhengdong Zhang,
| |
Collapse
|
17
|
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
|
18
|
Liang W, Wu X, Dong Y, Chen X, Zhou P, Xu F. Mechanical stimuli-mediated modulation of bone cell function-implications for bone remodeling and angiogenesis. Cell Tissue Res 2021; 386:445-454. [PMID: 34665321 DOI: 10.1007/s00441-021-03532-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 09/21/2021] [Indexed: 12/20/2022]
Abstract
Bone remodeling, expressed as bone formation and turnover, is a complex and dynamic process closely related to its form and function. Different events, such as development, aging, and function, play a critical role in bone remodeling and metabolism. The ability of the bone to adapt to new loads and forces has been well known and has proven useful in orthopedics and insightful for research in bone and cell biology. Mechanical stimulation is one of the most important drivers of bone metabolism. Interestingly, different types of forces will have specific consequences in bone remodeling, and their beneficial effects can be traced using different biomarkers. In this narrative review, we summarize the major mediators and events in bone remodeling, focusing on the effects of mechanical stimulation on bone metabolism, cell populations, and ultimately, bone health.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan 316000, Zhejiang Province, People's Republic of China.
| | - Xudong Wu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan 316000, Zhejiang Province, People's Republic of China
| | - Yongqiang Dong
- Department of Orthopaedics, Xinchang People's Hospital, Shaoxing, 312500, Zhejiang Province, People's Republic of China
| | - Xuerong Chen
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, 312000, Zhejiang Province, People's Republic of China
| | - Ping Zhou
- Department of Orthopaedics, Shaoxing People's Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, 312000, Zhejiang Province, People's Republic of China
| | - Fangming Xu
- Department of Orthopaedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan 316000, Zhejiang Province, People's Republic of China.
| |
Collapse
|
19
|
Polarized M2 macrophages induced by mechanical stretching modulate bone regeneration of the craniofacial suture for midfacial hypoplasia treatment. Cell Tissue Res 2021; 386:585-603. [PMID: 34568957 DOI: 10.1007/s00441-021-03533-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/21/2021] [Indexed: 01/01/2023]
Abstract
The underlying mechanism of the trans-sutural distraction osteogenesis (TSDO) technique as an effective treatment that improves the symptoms of midfacial hypoplasia syndromes is not clearly understood. Increasing findings in the orthopedics field indicate that macrophages are mechanically sensitive and their phenotypes can respond to mechanical cues. However, how macrophages respond to mechanical stretching and consequently influence osteoblast differentiation of suture-derived stem cells (SuSCs) remains unclear, particularly during the TSDO process. In the present study, we established a TSDO rat model to determine whether and how macrophages were polarized in response to stretching and consequently affected bone regeneration of the suture frontal edge. Notably, after performing immunofluorescence, RNA-sequencing, and micro-computed tomography, it was demonstrated that macrophages are first recruited by various chemokines factors and polarized to the M2 phenotype upon optimal stretching. The latter in turn regulates SuSC activity and facilitates bone regeneration in sutures. Moreover, when the activated M2 macrophages were suppressed by pharmacological manipulation, new bone microarchitecture could rarely be detected under mechanical stretching and the expansion of the sutures was clear. Additionally, macrophages achieved M2 polarization in response to the optimal mechanical stretching (10%, 0.5 Hz) and strongly facilitated SuSC osteogenic differentiation and human umbilical vein endothelial cell angiogenesis using an indirect co-culture system in vitro. Collectively, this study revealed the mechanical stimulation-immune response-bone regeneration axis and clarified at least in part how sutures achieve bone regeneration in response to mechanical force.
Collapse
|
20
|
Karoichan A, Baudequin T, Al-Jallad H, Tabrizian M. Encapsulation and differentiation of adipose-derived mesenchymal stem cells in a biomimetic purine cross-linked chitosan sponge. J Biomed Mater Res A 2021; 110:585-594. [PMID: 34545996 DOI: 10.1002/jbm.a.37311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/19/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells derived from adipose tissue have become a widely investigated cell source to use in tissue engineering applications. However, an optimal delivery scaffold for these cells is still needed. A rapidly gelling, injectable chitosan sponge was proposed in this study as a potential candidate for a suitable delivery scaffold. The results demonstrated the ability to encapsulate the stem cells at a 97.6% encapsulation efficiency and that the cells maintain their viability within the sponge. With the potential of using this scaffold for bone tissue engineering, ALP activity assay and fluorescent imaging for osteocalcin proved the ability to differentiate the encapsulated cells into the osteogenic lineage. Furthermore, co-encapsulation of pyrophosphatase within the sponge was investigated as a method to overcome the inhibitory effects that the sponge degradation by-products have on mineralization. Alizarin Red S staining demonstrated the beneficial effects of adding pyrophosphatase, where a significant increase in mineralization levels was achieved.
Collapse
Affiliation(s)
- Antoine Karoichan
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospitals for Children, Montreal, Quebec, Canada
| | - Timothée Baudequin
- Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
| | - Hadil Al-Jallad
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Shriners Hospitals for Children, Montreal, Quebec, Canada.,Department of Experimental Surgery, McGill University, Montreal, Quebec, Canada
| | - Maryam Tabrizian
- Faculty of Dentistry, McGill University, Montreal, Quebec, Canada.,Biomedical Engineering Department, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
21
|
Gli1 + Cells Residing in Bone Sutures Respond to Mechanical Force via IP 3R to Mediate Osteogenesis. Stem Cells Int 2021; 2021:8138374. [PMID: 34434241 PMCID: PMC8380501 DOI: 10.1155/2021/8138374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/26/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Early orthodontic correction of skeletal malocclusion takes advantage of mechanical force to stimulate unclosed suture remodeling and to promote bone reconstruction; however, the underlying mechanisms remain largely unclear. Gli1+ cells in maxillofacial sutures have been shown to participate in maxillofacial bone development and damage repair. Nevertheless, it remains to be investigated whether these cells participate in mechanical force-induced bone remodeling during orthodontic treatment of skeletal malocclusion. In this study, rapid maxillary expansion (RME) mouse models and mechanical stretch loading cell models were established using two types of transgenic mice which are able to label Gli1+ cells, and we found that Gli1+ cells participated in mechanical force-induced osteogenesis both in vivo and in vitro. Besides, we found mechanical force-induced osteogenesis through inositol 1,4,5-trisphosphate receptor (IP3R), and we observed for the first time that inhibition of Gli1 suppressed an increase in mechanical force-induced IP3R overexpression, suggesting that Gli1+ cells participate in mechanical force-induced osteogenesis through IP3R. Taken together, this study is the first to demonstrate that Gli1+ cells in maxillofacial sutures are involved in mechanical force-induced bone formation through IP3R during orthodontic treatment of skeletal malocclusion. Furthermore, our results provide novel insights regarding the mechanism of orthodontic treatments of skeletal malocclusion.
Collapse
|
22
|
Zhu G, Zhang T, Chen M, Yao K, Huang X, Zhang B, Li Y, Liu J, Wang Y, Zhao Z. Bone physiological microenvironment and healing mechanism: Basis for future bone-tissue engineering scaffolds. Bioact Mater 2021; 6:4110-4140. [PMID: 33997497 PMCID: PMC8091181 DOI: 10.1016/j.bioactmat.2021.03.043] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023] Open
Abstract
Bone-tissue defects affect millions of people worldwide. Despite being common treatment approaches, autologous and allogeneic bone grafting have not achieved the ideal therapeutic effect. This has prompted researchers to explore novel bone-regeneration methods. In recent decades, the development of bone tissue engineering (BTE) scaffolds has been leading the forefront of this field. As researchers have provided deep insights into bone physiology and the bone-healing mechanism, various biomimicking and bioinspired BTE scaffolds have been reported. Now it is necessary to review the progress of natural bone physiology and bone healing mechanism, which will provide more valuable enlightenments for researchers in this field. This work details the physiological microenvironment of the natural bone tissue, bone-healing process, and various biomolecules involved therein. Next, according to the bone physiological microenvironment and the delivery of bioactive factors based on the bone-healing mechanism, it elaborates the biomimetic design of a scaffold, highlighting the designing of BTE scaffolds according to bone biology and providing the rationale for designing next-generation BTE scaffolds that conform to natural bone healing and regeneration.
Collapse
Affiliation(s)
- Guanyin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Ke Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yazhen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| |
Collapse
|
23
|
Manokawinchoke J, Pavasant P, Limjeerajarus CN, Limjeerajarus N, Osathanon T, Egusa H. Mechanical loading and the control of stem cell behavior. Arch Oral Biol 2021; 125:105092. [PMID: 33652301 DOI: 10.1016/j.archoralbio.2021.105092] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/08/2021] [Accepted: 02/21/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Mechanical stimulation regulates many cell responses. The present study describes the effects of different in vitro mechanical stimulation approaches on stem cell behavior. DESIGN The narrative review approach was performed. The articles published in English language that addressed the effects of mechanical force on stem cells were searched on Pubmed and Scopus database. The effects of extrinsic mechanical force on stem cell response was reviewed and discussed. RESULTS Cells sense mechanical stimuli by the function of mechanoreceptors and further transduce force stimulation into intracellular signaling. Cell responses to mechanical stimuli depend on several factors including type, magnitude, and duration. Further, similar mechanical stimuli exhibit distinct cell responses based on numerous factors including cell type and differentiation stage. Various mechanical applications modulate stemness maintenance and cell differentiation toward specific lineages. CONCLUSIONS Mechanical force application modulates stemness maintenance and differentiation. Modification of force regimens could be utilized to precisely control appropriate stem cell behavior toward specific applications.
Collapse
Affiliation(s)
- Jeeranan Manokawinchoke
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Chalida Nakalekha Limjeerajarus
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Nuttapol Limjeerajarus
- Research Center for Advanced Energy Technology, Faculty of Engineering, Thai-Nichi Institute of Technology, Bangkok, 10250, Thailand.
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand; Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan.
| |
Collapse
|
24
|
Shiroud Heidari B, Ruan R, De-Juan-Pardo EM, Zheng M, Doyle B. Biofabrication and Signaling Strategies for Tendon/Ligament Interfacial Tissue Engineering. ACS Biomater Sci Eng 2021; 7:383-399. [PMID: 33492125 DOI: 10.1021/acsbiomaterials.0c00731] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Tendons and ligaments (TL) have poor healing capability, and for serious injuries like tears or ruptures, surgical intervention employing autografts or allografts is usually required. Current tissue replacements are nonideal and can lead to future problems such as high retear rates, poor tissue integration, or heterotopic ossification. Alternatively, tissue engineering strategies are being pursued using biodegradable scaffolds. As tendons connect muscle and bone and ligaments attach bones, the interface of TL with other tissues represent complex structures, and this intricacy must be considered in tissue engineered approaches. In this paper, we review recent biofabrication and signaling strategies for biodegradable polymeric scaffolds for TL interfacial tissue engineering. First, we discuss biodegradable polymeric scaffolds based on the fabrication techniques as well as the target tissue application. Next, we consider the effect of signaling factors, including cell culture, growth factors, and biophysical stimulation. Then, we discuss human clinical studies on TL tissue healing using commercial synthetic scaffolds that have occurred over the past decade. Finally, we highlight the challenges and future directions for biodegradable scaffolds in the field of TL and interface tissue engineering.
Collapse
Affiliation(s)
- Behzad Shiroud Heidari
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia.,School of Engineering, The University of Western Australia, Perth, Western Australia 6009, Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Australia
| | - Rui Ruan
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Elena M De-Juan-Pardo
- School of Engineering, The University of Western Australia, Perth, Western Australia 6009, Australia.,T3mPLATE, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia.,Science and Engineering Faculty, Queensland University of Technology, Brisbane, Queensland, 4000, Australia
| | - Minghao Zheng
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia 6009, Australia.,Perron Institute for Neurological and Translational Science, Nedlands, Western Australia 6009, Australia
| | - Barry Doyle
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia 6009, Australia.,School of Engineering, The University of Western Australia, Perth, Western Australia 6009, Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Australia.,BHF Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH9 3FD, United Kingdom
| |
Collapse
|
25
|
Wang D, Cai J, Zeng Z, Gao X, Shao X, Ding Y, Feng X, Jing D. The interactions between mTOR and NF-κB: A novel mechanism mediating mechanical stretch-stimulated osteoblast differentiation. J Cell Physiol 2020; 236:4592-4603. [PMID: 33289098 DOI: 10.1002/jcp.30184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022]
Abstract
Mechanical stretch is known to promote osteoblast differentiation in vitro and accelerate bone regeneration in vivo, whereas the relevant mechanism remains unclear. Recent studies have shown the importance of reciprocal interactions between mammalian target of rapamycin (mTOR) and nuclear factor kappa B (NF-κB; two downstream molecules of Akt) in the regulation of tumor cells. Thus, we hypothesize that mTOR and NF-κB as well as their interconnection play a critical role in mediating stretch-induced osteogenic differentiation in osteoblasts. We herein found that mechanical stretch (10% elongation at six cycles/min) significantly promoted the expression of osteoblast differentiation-related markers (including ALP, BMP2, Col1α, OCN, and Runx2) in osteoblast-like MG-63 cells, accompanied by increased mTOR phosphorylation and NF-κB p65 phosphorylation and nuclear translocation. Blockade of mTOR by antagonist or small interfering RNA suppressed osteogenesis-related gene expression in response to mechanical stretch, whereas inhibition of NF-κB further increased stretch-induced osteoblast differentiation. Moreover, inhibition of mTOR decreased the phosphorylation of NF-κB, and blockade of NF-κB reduced the mTOR activation in MG63 cells under mechanical stretch. Coinhibition of mTOR and NF-κB abolishes the alteration of osteogenic differentiation induced by single mTOR or NF-κB inhibition under mechanical stretch, which is equivalent to the noninhibition level for osteoblasts under mechanical stretch. The expression levels of osteogenic differentiation in osteoblasts after inhibition of Akt were similar to those after co-inhibition of mTOR and NF-κB under mechanical stretch. This study for the first time reveals the reciprocal interconnection between mTOR and NF-κB in osteoblasts under mechanical stretch and indicates that mTOR and NF-κB as well as their interactions play a key role in the regulation of cellular homeostasis of osteoblasts in response to mechanical stretch. These findings are helpful for enriching our basic knowledge of the molecular mechanisms of osteoblast mechanotransduction, and also providing insight into the clinical therapeutic modality associated with mechanical stretch (e.g., distraction osteogenesis).
Collapse
Affiliation(s)
- Dan Wang
- Laboratory of Tissue Engineering, Faculty of Life Sciences, Northwest University, Xi'an, China
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Jing Cai
- Department of Diagnostics, College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zhaobin Zeng
- Department of Stomatology, General Hospital of Northern Theater Command (Formerly General Hospital of Shenyang Military Area), Shenyang, China
| | - Xue Gao
- Laboratory of Tissue Engineering, Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Xi Shao
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Yuanjun Ding
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| | - Xue Feng
- Department of Cell Biology, School of Medicine, Northwest University, Xi'an, China
| | - Da Jing
- Department of Biomedical Engineering, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
26
|
Park JH, Kang YH, Hwang SC, Oh SH, Byun JH. Parthenolide Has Negative Effects on In Vitro Enhanced Osteogenic Phenotypes by Inflammatory Cytokine TNF-α via Inhibiting JNK Signaling. Int J Mol Sci 2020; 21:ijms21155433. [PMID: 32751648 DOI: 10.3390/ijms21155433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor kappa B (NF-κB) regulates inflammatory gene expression and represents a likely target for novel disease treatment approaches, including skeletal disorders. Several plant-derived sesquiterpene lactones can inhibit the activation of NF-κB. Parthenolide (PTL) is an abundant sesquiterpene lactone, found in Mexican Indian Asteraceae family plants, with reported anti-inflammatory activity, through the inhibition of a common step in the NF-κB activation pathway. This study examined the effects of PTL on the enhanced, in vitro, osteogenic phenotypes of human periosteum-derived cells (hPDCs), mediated by the inflammatory cytokine tumor necrosis factor (TNF)-α. PTL had no significant effects on hPDC viability or osteoblastic activities, whereas TNF-α had positive effects on the in vitro osteoblastic differentiation of hPDCs. c-Jun N-terminal kinase (JNK) signaling played an important role in the enhanced osteoblastic differentiation of TNF-α-treated hPDCs. Treatment with 1 µM PTL did not affect TNF-α-treated hPDCs; however, 5 and 10 µM PTL treatment decreased the histochemical detection and activity of alkaline phosphatase (ALP), alizarin red-positive mineralization, and the expression of ALP and osteocalcin mRNA. JNK phosphorylation decreased significantly in TNF-α-treated hPDCs pretreated with PTL. These results suggested that PTL exerts negative effects on the increased osteoblastic differentiation of TNF-α-treated hPDCs by inhibiting JNK signaling.
Collapse
Affiliation(s)
- Jin-Ho Park
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
| | - Young-Hoon Kang
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Changwon Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery, Institute of Health Sciences, School of Medicine, Gyeongsang National University, Jinju 52727, Korea
| | - Se Heang Oh
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Korea
| | - June-Ho Byun
- Department of Oral and Maxillofacial Surgery, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Institute of Health Sciences, Gyeongsang National University, Jinju 52727, Korea
| |
Collapse
|
27
|
Wei L, Mousawi F, Li D, Roger S, Li J, Yang X, Jiang LH. Adenosine Triphosphate Release and P2 Receptor Signaling in Piezo1 Channel-Dependent Mechanoregulation. Front Pharmacol 2019; 10:1304. [PMID: 31780935 PMCID: PMC6853025 DOI: 10.3389/fphar.2019.01304] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Organs and tissues and their constituent cells are physiologically submitted to diverse types of mechanical forces or stress, one common sequence of which is release of intracellular ATP into extracellular space. Extracellular ATP is a well-established autocrine or paracrine signaling molecule that regulates multiple cell functions and mediates cell-to-cell communications via activating the purinergic P2 receptors, more specifically, ligand-gated ion channel P2X receptors and some of the G-protein-coupled P2Y receptors. The molecular mechanisms that sense mechanical and transduce forces to trigger ATP release are poorly understood. The Piezo1, a newly identified mechanosensing ion channel, shows widespread expression and confers mechanosensitivity in many different types of cells. In this mini-review, we briefly introduce the Piezo1 channel and discuss the evidence that supports its important role in the mechanoregulation of diverse cell functions and, more specifically, critical engagement of ATP release and subsequent P2 receptor activation in Piezo1 channel-dependent mechanoregulation. Such ATP release-mediated coupling of the Piezo1 channel and P2 receptors may serve a signaling mechanism that is more common than we currently understand in transducing mechanical information to regulation of the attendant cell functions in various organs and tissues.
Collapse
Affiliation(s)
- Linyu Wei
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Fatema Mousawi
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Dongliang Li
- Department of Physiology, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Sébastien Roger
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| | - Jing Li
- Lingnan Medical Research Centre, School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuebin Yang
- Department of Oral Biology, Faculty of Medicine and Health, University of Leeds, Leeds, United Kingdom
| | - Lin-Hua Jiang
- Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- EA4245, Transplantation, Immunology and Inflammation, Faculty of Medicine, University of Tours, Tours, France
| |
Collapse
|
28
|
Liu Y, Huang X, Yu H, Yang J, Li Y, Yuan X, Guo Q. HIF-1α-TWIST pathway restrains cyclic mechanical stretch-induced osteogenic differentiation of bone marrow mesenchymal stem cells. Connect Tissue Res 2019; 60:544-554. [PMID: 30938209 DOI: 10.1080/03008207.2019.1601185] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Aim: Mechanical strain plays a crucial role in bone formation and remodeling. Hypoxia-inducible factor (HIF)-1α and TWIST are upstream of master regulators of osteogenesis, including runt-related transcription factor 2 (RUNX2) and bone morphogenetic proteins (BMPs). This study investigated the effect of the HIF-1α-TWIST pathway on cyclic mechanical stretch-induced osteogenic differentiation of rat bone marrow mesenchymal stem cells (BMSCs) and the underlying mechanism. Materials and Methods: BMSCs were isolated from bone marrow derived from the femurs and humeri of Sprague-Dawley rats. Osteogenic differentiation of BMSCs was induced by applying cyclic mechanical stretch using the Flexcell Tension System. HIF-1α and TWIST were knocked down using recombinant lentiviral vectors. Osteogenic differentiation was evaluated by real-time qPCR, western blotting, and the alkaline phosphatase (ALP) activity assay. Results: Cyclic mechanical stretch increased ALP activity and expression of HIF-1α and TWIST in BMSCs. Knockdown of HIF-1α decreased TWIST expression in stretched BMSCs. Moreover, knockdown of HIF-1α or TWIST enhanced cyclic mechanical stretch-induced osteogenic differentiation of BMSCs. In addition, knockdown of TWIST increased expression of RUNX2 and BMP2 in stretched BMSCs. Conclusions: The HIF-1α-TWIST signaling pathway inhibits cyclic mechanical stretch-induced osteogenic differentiation of BMSCs. This finding may facilitate cell and tissue engineering for clinical applications.
Collapse
Affiliation(s)
- Ying Liu
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China.,Department of Orthodontics, Stomatology College of Qingdao University , Qingdao , Shandong , China.,Department of Stomatology, The Affiliated Qingdao Municipal Hospital, Qingdao University , Qingdao , Shandong , China
| | - Xia Huang
- Department of Nursing and Hospital Infection Management, The Affiliated Hospital of Qingdao University , Qingdao , China
| | - Haibo Yu
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Jing Yang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Yazhen Li
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Xiao Yuan
- Department of Orthodontics, The Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Qingyuan Guo
- Department of Stomatology, The Affiliated Qingdao Municipal Hospital, Qingdao University , Qingdao , Shandong , China.,Department of Stomatology, Chinese PLA General Hospital , Beijing , China
| |
Collapse
|
29
|
Jing H, Zhang X, Gao M, Luo K, Fu W, Yin M, Wang W, Zhu Z, Zheng J, He X. Kartogenin preconditioning commits mesenchymal stem cells to a precartilaginous stage with enhanced chondrogenic potential by modulating JNK and β‐catenin–related pathways. FASEB J 2019; 33:5641-5653. [DOI: 10.1096/fj.201802137rrr] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hui Jing
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiaoyang Zhang
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Manchen Gao
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Kai Luo
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Wei Fu
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Meng Yin
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Wei Wang
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Zhongqun Zhu
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Jinghao Zheng
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiaomin He
- Department of Cardiothoracic SurgeryShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|