1
|
Song Y, Jiao Y, Liu Y, Guo L. Role of Masticatory Force in Modulating Jawbone Immunity and Bone Homeostasis: A Review. Int J Mol Sci 2025; 26:4478. [PMID: 40429623 PMCID: PMC12111287 DOI: 10.3390/ijms26104478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Mastication exerts a significant influence on both the structural and immunological environment of the jawbone. The mechanical stress generated during chewing initiates bone remodeling through the coordinated activities of osteoclasts and osteoblasts, with these processes being modulated by immune cell responses. This review summarizes the interaction between masticatory forces and jawbone immunity, focusing on key mechanisms such as mechanotransduction in osteocytes, macrophage polarization, and the activation of T cells. The review also delves into the role of the receptor activator of nuclear factor κ-B ligand (RANKL), receptor activator of nuclear factor κ-B (RANK), and osteoprotegerin (OPG) signaling pathway, highlighting its critical function in bone resorption and immune regulation. Additionally, the review summarizes how masticatory forces modulate the immune response through changes in immune cells, particularly focusing on cytokines, and the involvement of hormonal and molecular pathways. These findings provide valuable insights into the complex interplay between mechanical forces and immune cells, with implications for bone health.
Collapse
Affiliation(s)
- Yue Song
- Department of Orthodontics (WangFuJing Campus), School of Stomatology, Capital Medical University, Scylla alley No. 11, Beijing 100069, China;
| | - Yao Jiao
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China;
| | - Yitong Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No. 4, Beijing 100050, China;
| | - Lijia Guo
- Department of Orthodontics (WangFuJing Campus), School of Stomatology, Capital Medical University, Scylla alley No. 11, Beijing 100069, China;
| |
Collapse
|
2
|
Zhao F, Zhang Y, Pei S, Wang S, Hu L, Wang L, Qian A, Yang TL, Guo Y. Mechanobiological crosstalk among bone cells and between bone and other organs. BONE CELL BIOMECHANICS, MECHANOBIOLOGY AND BONE DISEASES 2024:215-247. [DOI: 10.1016/b978-0-323-96123-3.00015-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
3
|
Chen Y, Zhang C. Role of noncoding RNAs in orthodontic tooth movement: new insights into periodontium remodeling. J Transl Med 2023; 21:101. [PMID: 36759852 PMCID: PMC9912641 DOI: 10.1186/s12967-023-03951-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Orthodontic tooth movement (OTM) is biologically based on the spatiotemporal remodeling process in periodontium, the mechanisms of which remain obscure. Noncoding RNAs (ncRNAs), especially microRNAs and long noncoding RNAs, play a pivotal role in maintaining periodontal homeostasis at the transcriptional, post-transcriptional, and epigenetic levels. Under force stimuli, mechanosensitive ncRNAs with altered expression levels transduce mechanical load to modulate intracellular genes. These ncRNAs regulate the biomechanical responses of periodontium in the catabolic, anabolic, and coupling phases throughout OTM. To achieve this, down or upregulated ncRNAs actively participate in cell proliferation, differentiation, autophagy, inflammatory, immune, and neurovascular responses. This review highlights the regulatory mechanism of fine-tuning ncRNAs in periodontium remodeling during OTM, laying the foundation for safe, precise, and personalized orthodontic treatment.
Collapse
Affiliation(s)
- Yuming Chen
- grid.284723.80000 0000 8877 7471Stomatological Hospital, Southern Medical University, Guangzhou, 510280 China
| | - Chao Zhang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
4
|
Yan L, Liao L, Su X. Role of mechano-sensitive non-coding RNAs in bone remodeling of orthodontic tooth movement: recent advances. Prog Orthod 2022; 23:55. [PMID: 36581789 PMCID: PMC9800683 DOI: 10.1186/s40510-022-00450-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/15/2022] [Indexed: 12/31/2022] Open
Abstract
Orthodontic tooth movement relies on bone remodeling and periodontal tissue regeneration in response to the complicated mechanical cues on the compressive and tensive side. In general, mechanical stimulus regulates the expression of mechano-sensitive coding and non-coding genes, which in turn affects how cells are involved in bone remodeling. Growing numbers of non-coding RNAs, particularly mechano-sensitive non-coding RNA, have been verified to be essential for the regulation of osteogenesis and osteoclastogenesis and have revealed how they interact with signaling molecules to do so. This review summarizes recent findings of non-coding RNAs, including microRNAs and long non-coding RNAs, as crucial regulators of gene expression responding to mechanical stimulation, and outlines their roles in bone deposition and resorption. We focused on multiple mechano-sensitive miRNAs such as miR-21, - 29, -34, -103, -494-3p, -1246, -138-5p, -503-5p, and -3198 that play a critical role in osteogenesis function and bone resorption. The emerging roles of force-dependent regulation of lncRNAs in bone remodeling are also discussed extensively. We summarized mechano-sensitive lncRNA XIST, H19, and MALAT1 along with other lncRNAs involved in osteogenesis and osteoclastogenesis. Ultimately, we look forward to the prospects of the novel application of non-coding RNAs as potential therapeutics for tooth movement and periodontal tissue regeneration.
Collapse
Affiliation(s)
- Lichao Yan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry and Engineering Research Center of Oral Translational Medicine and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry and Engineering Research Center of Oral Translational Medicine and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoxia Su
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry and Engineering Research Center of Oral Translational Medicine and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
5
|
Liu X, Zhang K, Wang L, Geng B, Liu Z, Yi Q, Xia Y. Fluid shear stress-induced down-regulation of miR-146a-5p inhibits osteoblast apoptosis via targeting SMAD4. Physiol Res 2022. [DOI: 10.33549/physiolres.934922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Fluid shear stress (FSS) plays an important role in osteoblast apoptosis. However, the role of miRNA in osteoblast apoptosis under FSS and possible molecular mechanisms remain unknown. Our aim of the study was to explore whether miR-146a-5p regulates osteoblast apoptosis under FSS and its molecular mechanisms. FSS could down-regulate the expression of miR-146a-5p in MC3T3-E1 cells. We confirm that up-regulation of miR-146a-5p promotes osteoblasts apoptosis and down-regulation of miR-146a-5p inhibits osteoblasts apoptosis. We further demonstrated that FSS inhibits osteoblast apoptosis by down-regulated miR-146a-5p. Dual-luciferase reporter assay validated that SMAD4 is a direct target gene of miR-146a-5p. In addition, mimic-146a-5p suppressed FSS-induced up-regulation of SMAD4 protein levels, which suggests that FSS elevated SMAD4 protein expression levels via regulation miR-146a-5p. Further investigations showed that SMAD4 could inhibit osteoblast apoptosis. We demonstrated that miR-146a-5p regulates osteoblast apoptosis via targeting SMAD4. Taken together, our present study showed that FSS-induced down-regulation miR-146a-5p inhibits osteoblast apoptosis via target SMAD4. These findings may provide novel mechanisms for FSS to inhibit osteoblast apoptosis, and also may provide a potential therapeutic target for osteoporosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Y Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, China
| |
Collapse
|
6
|
Han Y, Yang Q, Huang Y, Gao P, Jia L, Zheng Y, Li W. Compressive force regulates orthodontic tooth movement via activating the NLRP3 inflammasome. FASEB J 2022; 36:e22627. [PMID: 36314562 DOI: 10.1096/fj.202200447rr] [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: 03/23/2022] [Revised: 10/01/2022] [Accepted: 10/12/2022] [Indexed: 11/27/2022]
Abstract
Mechanical stress regulates various cellular functions like cell inflammation, immune responses, proliferation, and differentiation to maintain tissue homeostasis. However, the impact of mechanical signals on macrophages and the underlying mechanisms by which mechanical force regulates bone remodeling during orthodontic tooth movement remain unclear. NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome has been reported to promote osteoclastic differentiation to regulate alveolar bone resorption. But the relationship between the compressive force and NLRP3 inflammasome in macrophages remains unknown. In this study, immunohistochemical staining results showed elevated expression of NLRP3 and interleukin-1β, as well as an increased number of macrophages expressing NLRP3, on the compression side of the periodontal tissues, after force application for 7 days. Furthermore, the number of tartrate-resistant acid phosphatase-positive osteoclasts, and the mRNA and protein expression levels of osteoclast-related genes in the periodontal tissue decreased in the Nlrp3-/- mice compared to the WT mice group after orthodontic movement. In vitro mechanical force activates the NLRP3 inflammasome and inhibits autophagy. Intraperitoneal injection of the autophagy inhibitor 3-methyladenine in Nlrp3-/- mice promoted orthodontic tooth movement. This result indicates that the absence of NLRP3 inflammasome activation can be partially compensated for by autophagy inhibitors. Mechanistically, force-induced activation of the NLRP3 inflammasome in macrophages via the cGAS/P2X7R axis. In conclusion, compressive force regulates orthodontic tooth movement via activating the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Yineng Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Hangzhou, People's Republic of China.,National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, People's Republic of China
| | - Qiaolin Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, People's Republic of China
| | - Yiping Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, People's Republic of China
| | - Pengfei Gao
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, People's Republic of China.,Peking-Tsinghua Center for Life Sciences, Beijing, People's Republic of China
| | - Lingfei Jia
- National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, People's Republic of China.,Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, People's Republic of China
| | - Weiran Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.,National Center of Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing, People's Republic of China
| |
Collapse
|
7
|
Zhang K, Liu X, Tang Y, Liu Z, Yi Q, Wang L, Geng B, Xia Y. Fluid Shear Stress Promotes Osteoblast Proliferation and Suppresses Mitochondrial-Mediated Osteoblast Apoptosis Through the miR-214-3p-ATF4 Signaling Axis. Physiol Res 2022. [DOI: 10.33549/physiolres.934917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs (miRNAs) play vital roles in bone metabolism and participate in the mechanically induced bone alterations. The underlying molecular mechanisms by which fluid shear stress (FSS) regulate the proliferative and apoptotic phenotypic changes of osteoblasts remain elusive. The study aimed to investigate the regulatory effects of FSS on osteoblast proliferative and apoptotic phenotypes and the roles of miR-214-3p-ATF4 (activating transcription factor 4) signaling axis in the mechanomodulation processes. FSS promoted the proliferative activity of osteoblasts and suppressed mitochondrial-mediated osteoblast apoptosis. FSS decreased miR-214-3p expression and increased ATF4 expression in MC3T3-E1 osteoblasts. MiR-214-3p inhibited osteoblast proliferative activity and promoted mitochondrial-mediated osteoblast apoptosis. Overexpression of miR-214-3p attenuated FSS-enhanced osteoblast proliferation and FSS-suppressed mitochondrial-mediated osteoblast apoptosis. We validated that ATF4 acted as a target gene of miR-214-3p. Moreover, miR-214 3p regulated osteoblast proliferation and apoptosis through targeting ATF4. Taken together, our study proved that FSS could suppress mitochondrial-mediated osteoblast apoptosis and promote osteoblast proliferation through the miR-214-3p-ATF4 signaling axis.
Collapse
Affiliation(s)
- K Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, China, Orthopaedics Key Laboratory of Gansu Province, Lanzhou Gansu, China
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Cai J, Li C, Li S, Yi J, Wang J, Yao K, Gan X, Shen Y, Yang P, Jing D, Zhao Z. A Quartet Network Analysis Identifying Mechanically Responsive Long Noncoding RNAs in Bone Remodeling. Front Bioeng Biotechnol 2022; 10:780211. [PMID: 35356768 PMCID: PMC8959777 DOI: 10.3389/fbioe.2022.780211] [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: 09/20/2021] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
Mechanical force, being so ubiquitous that it is often taken for granted and overlooked, is now gaining the spotlight for reams of evidence corroborating their crucial roles in the living body. The bone, particularly, experiences manifold extraneous force like strain and compression, as well as intrinsic cues like fluid shear stress and physical properties of the microenvironment. Though sparkled in diversified background, long noncoding RNAs (lncRNAs) concerning the mechanotransduction process that bone undergoes are not yet detailed in a systematic way. Our principal goal in this research is to highlight the potential lncRNA-focused mechanical signaling systems which may be adapted by bone-related cells for biophysical environment response. Based on credible lists of force-sensitive mRNAs and miRNAs, we constructed a force-responsive competing endogenous RNA network for lncRNA identification. To elucidate the underlying mechanism, we then illustrated the possible crosstalk between lncRNAs and mRNAs as well as transcriptional factors and mapped lncRNAs to known signaling pathways involved in bone remodeling and mechanotransduction. Last, we developed combinative analysis between predicted and established lncRNAs, constructing a pathway–lncRNA network which suggests interactive relationships and new roles of known factors such as H19. In conclusion, our work provided a systematic quartet network analysis, uncovered candidate force-related lncRNAs, and highlighted both the upstream and downstream processes that are possibly involved. A new mode of bioinformatic analysis integrating sequencing data, literature retrieval, and computational algorithm was also introduced. Hopefully, our work would provide a moment of clarity against the multiplicity and complexity of the lncRNA world confronting mechanical input.
Collapse
Affiliation(s)
- Jingyi Cai
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chaoyuan Li
- Department of Oral Implantology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, School and Hospital of Stomatology, Tongji University, Shanghai, China
| | - Shun Li
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, China
| | - Jianru Yi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ke Yao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyan Gan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Shen
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
| | - Pu Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dian Jing
- Department of Orthodontics, China Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Dian Jing, ; Zhihe Zhao,
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Dian Jing, ; Zhihe Zhao,
| |
Collapse
|
9
|
Yang H, Cao Z, Wang Y, Wang J, Gao J, Han B, Yu F, Qin Y, Guo Y. Treadmill exercise influences the microRNA profiles in the bone tissues of mice. Exp Ther Med 2021; 22:1035. [PMID: 34373721 PMCID: PMC8343800 DOI: 10.3892/etm.2021.10467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
As an important regulator involved in cell activity, microRNAs (miRNAs) are important in the process of exercise influencing bone metabolism. The present study aimed to detect and select differentially expressed miRNAs in the bone tissues of mice trained on a treadmill, predict the target genes of these differentially expressed miRNAs and lay a foundation for exploring the effect of treadmill training on bone metabolism through miRNAs. In this experiment, after the mice were trained on a treadmill for 8 weeks, the mechanical properties of mouse femur bone were assessed, and the alkaline phosphatase (ALP) activity and osteocalcin (OCN) protein levels of the bone were assayed. miRNA microarray and reverse transcription-quantitative (RT-q)PCR were performed to select and validate differentially expressed miRNAs in the bone, and the target genes of these miRNAs were predicted with bioinformatics methods. In addition, the differentially expressed miRNAs in the bone tissues were compared with those in mechanically strained osteocytes in vitro. Treadmill training improved the mechanical properties of the femur bones of mice, and elevated the ALP activity and OCN protein level in the bone. In addition, 122 differentially expressed miRNAs were detected in the bone, of which nine were validated via RT-qPCR. Among the target genes of these differentially expressed miRNAs, certain candidates were involved in bone metabolism. A total of eight miRNAs were differentially expressed in both bone tissue and osteocytes, exhibiting the same expression trends, and various target genes of these eight miRNAs were also involved in bone metabolism. Treadmill training resulted in altered miRNA expression profiles in the bones of mice (mainly in osteocytes) and the differentially expressed miRNAs may serve important roles in regulating bone metabolism and osteogenic differentiation.
Collapse
Affiliation(s)
- Huan Yang
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Zhen Cao
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Yang Wang
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China.,Department of Biomedical Engineering, Bioengineering College of Chongqing University, Chongqing 400044, P.R. China
| | - Jiahui Wang
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Jintao Gao
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Biao Han
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Fangmei Yu
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Yixiong Qin
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Yong Guo
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| |
Collapse
|
10
|
Mao L, Guo J, Hu L, Li L, Xu J, Zou J. The effects of biophysical stimulation on osteogenic differentiation and the mechanisms from ncRNAs. Cell Biochem Funct 2021; 39:727-739. [PMID: 34041775 DOI: 10.1002/cbf.3650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023]
Abstract
Ample proof showed that non-coding RNAs (ncRNAs) play a crucial role in proliferation and differentiation of osteoblasts and bone marrow stromal cells (BMSCs). Varied forms of biophysical stimuli like mechanical strain, fluid shear stress (FSS), microgravity and vibration are verified to regulate ncRNAs expression in osteogenic differentiation and influence the expression of target genes associated with osteogenic differentiation and ultimately regulate bone formation. The consequences of biophysical stimulation on osteogenic differentiation validate the prospect of exercise for the prevention and treatment of osteoporosis. In this review, we tend to summarize the studies on regulation of osteogenic differentiation by ncRNAs beneath biophysical stimulation and facilitate to reveal the regulatory mechanism of biophysical stimulation on ncRNAs, and provide an update for the prevention of bone metabolism diseases by exercise.
Collapse
Affiliation(s)
- Liwei Mao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Linghui Hu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lexuan Li
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
11
|
Iwasaki H, Ichihara Y, Morino K, Lemecha M, Sugawara L, Sawano T, Miake J, Sakurai H, Nishi E, Maegawa H, Imamura T. MicroRNA-494-3p inhibits formation of fast oxidative muscle fibres by targeting E1A-binding protein p300 in human-induced pluripotent stem cells. Sci Rep 2021; 11:1161. [PMID: 33441918 PMCID: PMC7806978 DOI: 10.1038/s41598-020-80742-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/17/2020] [Indexed: 01/29/2023] Open
Abstract
MYOD-induced microRNA-494-3p expression inhibits fast oxidative myotube formation by downregulating myosin heavy chain 2 (MYH2) in human induced pluripotent stem cells (hiPSCs) during skeletal myogenesis. However, the molecular mechanisms regulating MYH2 expression via miR-494-3p remain unknown. Here, using bioinformatic analyses, we show that miR-494-3p potentially targets the transcript of the E1A-binding protein p300 at its 3'-untranslated region (UTR). Myogenesis in hiPSCs with the Tet/ON-myogenic differentiation 1 (MYOD1) gene (MyoD-hiPSCs) was induced by culturing them in doxycycline-supplemented differentiation medium for 7 days. p300 protein expression decreased after transient induction of miR-494-3p during myogenesis. miR-494-3p mimics decreased the levels of p300 and its downstream targets MYOD and MYH2 and myotube formation efficiency. p300 knockdown decreased myotube formation efficiency, MYH2 expression, and basal oxygen consumption rate. The binding of miR-494-3p to the wild type p300 3'-UTR, but not the mutated site, was confirmed using luciferase assay. Overexpression of p300 rescued the miR-494-3p mimic-induced phenotype in MyoD-hiPSCs. Moreover, miR-494-3p mimic reduced the levels of p300, MYOD, and MYH2 in skeletal muscles in mice. Thus, miR-494-3p might modulate MYH2 expression and fast oxidative myotube formation by directly regulating p300 levels during skeletal myogenesis in MyoD-hiPSCs and murine skeletal muscle tissues.
Collapse
Affiliation(s)
- Hirotaka Iwasaki
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Yoshinori Ichihara
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Katsutaro Morino
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan.
| | - Mengistu Lemecha
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan
- Department of Molecular and Cellular Biology, City of Hope, Los Angeles, USA
| | - Lucia Sugawara
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan
| | - Tatsuya Sawano
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Junichiro Miake
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Hiroshi Maegawa
- Division of Endocrinology and Metabolism, Department of Medicine, Shiga University of Medical Science, Tsukinowa, Seta, Otsu, Shiga, 520-2192, Japan
| | - Takeshi Imamura
- Division of Pharmacology, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
12
|
Chen Z, Zhang Y, Huai Y, Zhao F, Hu L, Yang C, Tian Y, Qian A. Mechanosensitive MicroRNAs and Bone Formation. NONCODING RNAS AND BONE 2021:79-91. [DOI: 10.1007/978-981-16-2402-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
13
|
Potekhina Y, Filatova A, Tregubova E, Mokhov D. Mechanosensitivity of Cells and Its Role in the Regulation of Physiological Functions and the Implementation of Physiotherapeutic Effects (Review). Sovrem Tekhnologii Med 2020; 12:77-89. [PMID: 34795996 PMCID: PMC8596276 DOI: 10.17691/stm2020.12.4.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Indexed: 01/11/2023] Open
Abstract
Regulatory signals in the body are not limited to chemical and electrical ones. There is another type of important signals for cells: those are mechanical signals (coming from the environment or arising from within the body), which have been less known in the literature. The review summarizes new information on the mechanosensitivity of various cells of connective tissue and nervous system. Participation of mechanical stimuli in the regulation of growth, development, differentiation, and functioning of tissues is described. The data focus on bone remodeling, wound healing, neurite growth, and the formation of neural networks. Mechanotransduction, cellular organelles, and mechanosensitive molecules involved in these processes are discussed as well as the role of the extracellular matrix. The importance of mechanical characteristics of cells in the pathogenesis of diseases is highlighted. Finally, the possible role of mechanosensitivity in mediating the physiotherapeutic effects is addressed.
Collapse
Affiliation(s)
- Yu.P. Potekhina
- Professor, Department of Normal Physiology named after N.Y. Belenkov; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - A.I. Filatova
- Student, Faculty of Pediatrics; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - E.S. Tregubova
- Professor, Department of Osteopathy; North-Western State Medical University named after I.I. Mechnikov, 41 Kirochnaya St., Saint Petersburg, 191015, Russia; Associate Professor, Institute of Osteopathy; Saint Petersburg State University, 7/9 Universitetskaya naberezhnaya, Saint Petersburg, 199034, Russia
| | - D.E. Mokhov
- Head of the Department of Osteopathy; North-Western State Medical University named after I.I. Mechnikov, 41 Kirochnaya St., Saint Petersburg, 191015, Russia; Director of the Institute of Osteopathy Saint Petersburg State University, 7/9 Universitetskaya naberezhnaya, Saint Petersburg, 199034, Russia
| |
Collapse
|
14
|
Yan Y, Wang L, Ge L, Pathak JL. Osteocyte-Mediated Translation of Mechanical Stimuli to Cellular Signaling and Its Role in Bone and Non-bone-Related Clinical Complications. Curr Osteoporos Rep 2020; 18:67-80. [PMID: 31953640 DOI: 10.1007/s11914-020-00564-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Osteocytes comprise > 95% of the cellular component in bone tissue and produce a wide range of cytokines and cellular signaling molecules in response to mechanical stimuli. In this review, we aimed to summarize the molecular mechanisms involved in the osteocyte-mediated translation of mechanical stimuli to cellular signaling, and discuss their role in skeletal (bone) diseases and extra-skeletal (non-bone) clinical complications. RECENT FINDINGS Two decades before, osteocytes were assumed as a dormant cells buried in bone matrix. In recent years, emerging evidences have shown that osteocytes are pivotal not only for bone homeostasis but also for vital organ functions such as muscle, kidney, and heart. Osteocyte mechanotransduction regulates osteoblast and osteoclast function and maintains bone homeostasis. Mechanical stimuli modulate the release of osteocyte-derived cytokines, signaling molecules, and extracellular cellular vesicles that regulate not only the surrounding bone cell function and bone homeostasis but also the distant organ function in a paracrine and endocrine fashion. Mechanical loading and unloading modulate the osteocytic release of NO, PGE2, and ATPs that regulates multiple cellular signaling such as Wnt/β-catenin, RANKL/OPG, BMPs, PTH, IGF1, VEGF, sclerostin, and others. Therefore, the in-depth study of the molecular mechanism of osteocyte mechanotransduction could unravel therapeutic targets for various bone and non-bone-related clinical complications such as osteoporosis, sarcopenia, and cancer metastasis to bone.
Collapse
Affiliation(s)
- Yongyong Yan
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510140, China
| | - Liping Wang
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510140, China
| | - Linhu Ge
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510140, China.
| | - Janak L Pathak
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510140, China.
| |
Collapse
|
15
|
Kılıç A, Ameli A, Park JA, Kho AT, Tantisira K, Santolini M, Cheng F, Mitchel JA, McGill M, O'Sullivan MJ, De Marzio M, Sharma A, Randell SH, Drazen JM, Fredberg JJ, Weiss ST. Mechanical forces induce an asthma gene signature in healthy airway epithelial cells. Sci Rep 2020; 10:966. [PMID: 31969610 PMCID: PMC6976696 DOI: 10.1038/s41598-020-57755-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/23/2019] [Indexed: 12/27/2022] Open
Abstract
Bronchospasm compresses the bronchial epithelium, and this compressive stress has been implicated in asthma pathogenesis. However, the molecular mechanisms by which this compressive stress alters pathways relevant to disease are not well understood. Using air-liquid interface cultures of primary human bronchial epithelial cells derived from non-asthmatic donors and asthmatic donors, we applied a compressive stress and then used a network approach to map resulting changes in the molecular interactome. In cells from non-asthmatic donors, compression by itself was sufficient to induce inflammatory, late repair, and fibrotic pathways. Remarkably, this molecular profile of non-asthmatic cells after compression recapitulated the profile of asthmatic cells before compression. Together, these results show that even in the absence of any inflammatory stimulus, mechanical compression alone is sufficient to induce an asthma-like molecular signature.
Collapse
Affiliation(s)
- Ayşe Kılıç
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Asher Ameli
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Physics, Northeastern University, Boston, MA, USA
| | - Jin-Ah Park
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Alvin T Kho
- Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA
| | - Kelan Tantisira
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Marc Santolini
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Centre for Research and Interdisciplinarity (CRI), Paris, F-75014, France
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, 44106, USA
| | - Jennifer A Mitchel
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Maureen McGill
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Michael J O'Sullivan
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Margherita De Marzio
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Amitabh Sharma
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffrey M Drazen
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Jeffrey J Fredberg
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Program in Molecular Integrative Phyisological Sciences, Department of Environmental Health, Harvard TH Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
16
|
Mechanosensitive MiRs regulated by anabolic and catabolic loading of human cartilage. Osteoarthritis Cartilage 2019; 27:1208-1218. [PMID: 31009748 DOI: 10.1016/j.joca.2019.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/26/2019] [Accepted: 04/10/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Elucidation of whether miRs are involved in mechanotransduction pathways by which cartilage is maintained or disturbed has a particular importance in our understanding of osteoarthritis (OA) pathophysiology. The aim was to investigate whether mechanical loading influences global miR-expression in human chondrocytes and to identify mechanosensitive miRs responding to beneficial and non-beneficial loading regimes as potential to obtain valuable diagnostic or therapeutic targets to advance OA-treatment. METHOD Mature tissue-engineered human cartilage was subjected to two distinct loading regimes either stimulating or suppressing proteoglycan-synthesis, before global miR microarray analysis. Promising candidate miRs were selected, re-evaluated by qRT-PCR and tested for expression in human healthy vs OA cartilage samples. RESULTS After anabolic loading, miR microarray profiling revealed minor changes in miR-expression while catabolic stimulation produced a significant regulation of 80 miRs with a clear separation of control and compressed samples by hierarchical clustering. Cross-testing of selected miRs revealed that miR-221, miR-6872-3p, miR-6723-5p were upregulated by both loading conditions while others (miR-199b-5p, miR-1229-5p, miR-1275, miR-4459, miR-6891-5p, miR-7150) responded specifically after catabolic loading. Mechanosensitivity of miR-221 correlated with pERK1/2-activation induced by both loading conditions. The miR-response to loading was transient and a constitutive deregulation of mechano-miRs in OA vs healthy articular cartilage was not observed. CONCLUSIONS MiRs with broader vs narrower mechanosensitivity were discovered and the first group of mechanosensitive miRs characteristic for non-beneficial loading was defined that may shape the proteome differentially when cartilage tissue is disturbed. The findings prompt future investigations into miR-relevance for mechano-responsive pathways and the corresponding miR-target molecules.
Collapse
|
17
|
Bellavia D, De Luca A, Carina V, Costa V, Raimondi L, Salamanna F, Alessandro R, Fini M, Giavaresi G. Deregulated miRNAs in bone health: Epigenetic roles in osteoporosis. Bone 2019; 122:52-75. [PMID: 30772601 DOI: 10.1016/j.bone.2019.02.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
MicroRNA (miRNA) has shown to enhance or inhibit cell proliferation, differentiation and activity of different cell types in bone tissue. The discovery of miRNA actions and their targets has helped to identify them as novel regulations actors in bone. Various studies have shown that miRNA deregulation mediates the progression of bone-related pathologies, such as osteoporosis. The present review intends to give an exhaustive overview of miRNAs with experimentally validated targets involved in bone homeostasis and highlight their possible role in osteoporosis development. Moreover, the review analyzes miRNAs identified in clinical trials and involved in osteoporosis.
Collapse
Affiliation(s)
- D Bellavia
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - A De Luca
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - V Carina
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - V Costa
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - L Raimondi
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - F Salamanna
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - R Alessandro
- Department of Biopathology and Medical Biotechnologies, Section of Biology and Genetics, University of Palermo, Palermo 90133, Italy; Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council, Palermo, Italy
| | - M Fini
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - G Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| |
Collapse
|
18
|
Bone remodeling induced by mechanical forces is regulated by miRNAs. Biosci Rep 2018; 38:BSR20180448. [PMID: 29844019 PMCID: PMC6028748 DOI: 10.1042/bsr20180448] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/13/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022] Open
Abstract
The relationship between mechanical force and alveolar bone remodeling is an important issue in orthodontics because tooth movement is dependent on the response of bone tissue to the mechanical force induced by the appliances used. Mechanical cyclical stretch (MCS), fluid shear stress (FSS), compression, and microgravity play different roles in the cell differentiation and proliferation involved in bone remodeling. However, the underlying mechanisms are unclear, particularly the molecular pathways regulated by non-coding RNAs (ncRNAs) that play essential roles in bone remodeling. Amongst the various ncRNAs, miRNAs act as post-transcriptional regulators that inhibit the expression of their target genes. miRNAs are considered key regulators of many biologic processes including bone remodeling. Here, we review the role of miRNAs in mechanical force-induced bone metabolism.
Collapse
|
19
|
Frith JE, Kusuma GD, Carthew J, Li F, Cloonan N, Gomez GA, Cooper-White JJ. Mechanically-sensitive miRNAs bias human mesenchymal stem cell fate via mTOR signalling. Nat Commun 2018; 9:257. [PMID: 29343687 PMCID: PMC5772625 DOI: 10.1038/s41467-017-02486-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 12/01/2017] [Indexed: 01/17/2023] Open
Abstract
Mechanotransduction is a strong driver of mesenchymal stem cell (MSC) fate. In vitro, variations in matrix mechanics invoke changes in MSC proliferation, migration and differentiation. However, when incorporating MSCs within injectable, inherently soft hydrogels, this dominance over MSC response substantially limits our ability to couple the ease of application of hydrogels with efficiently directed MSC differentiation, especially in the case of bone generation. Here, we identify differential miRNA expression in response to varying hydrogel stiffness and RhoA activity. We show that modulation of miR-100-5p and miR-143-3p can be used to bias MSC fate and provide mechanistic insight by demonstrating convergence on mTOR signalling. By modulating these mechanosensitive miRNAs, we can enhance osteogenesis in a soft 3D hydrogel. The outcomes of this study provide new understanding of the mechanisms regulating MSC mechanotransduction and differentiation, but also a novel strategy with which to drive MSC fate and significantly impact MSC-based tissue-engineering applications. Mesenchymal stem cell (MSC) fate can be mechanically regulated by substrate stiffness but this is difficult to control in a 3D hydrogel. Here the authors identify miRNAs that change expression in response to substrate stiffness and RhoA signalling and show that they can bias MSC fate in a 3D soft hydrogel.
Collapse
Affiliation(s)
- Jessica E Frith
- Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia. .,Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, 3072, Australia.
| | - Gina D Kusuma
- Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - James Carthew
- Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Fanyi Li
- Materials Science and Engineering, Monash University, Clayton, VIC, 3800, Australia
| | - Nicole Cloonan
- School of Biological Sciences, The University of Auckland, Auckland Central, Auckland, 1010, New Zealand
| | - Guillermo A Gomez
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Frome Road, Adelaide, 5000, SA, Australia.,Institute for Molecular Bioscience, Cell Biology, University of Queensland, St Lucia, QLD, 3072, Australia
| | - Justin J Cooper-White
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, 3072, Australia.,School of Chemical Engineering, University of Queensland, St Lucia, QLD, 3072, Australia.,Biomedical Manufacturing Manufacturing Flagship, CSIRO, Clayton, VIC, 3169, Australia
| |
Collapse
|
20
|
Yu W, Zheng Y, Yang Z, Fei H, Wang Y, Hou X, Sun X, Shen Y. N-AC-l-Leu-PEI-mediated miR-34a delivery improves osteogenic differentiation under orthodontic force. Oncotarget 2017; 8:110460-110473. [PMID: 29299161 PMCID: PMC5746396 DOI: 10.18632/oncotarget.22790] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/14/2017] [Indexed: 01/08/2023] Open
Abstract
Rare therapeutic genes or agents are reported to control orthodontic bone remodeling. MicroRNAs have recently been associated with bone metabolism. Here, we report the in vitro and in vivo effects of miR-34a on osteogenic differentiation under orthodontic force using an N-acetyl-L-leucine-modified polyethylenimine (N-Ac-l-Leu-PEI) carrier. N-Ac-l-Leu-PEI exhibited low cytotoxicity and high miR-34a transfection efficiency in rat bone mineral stem cells and local alveolar bone tissue. After transfection, miR-34a enhanced the osteogenic differentiation of Runx2 and ColI, Runx2 and ColI protein levels, and early osteogenesis function under orthodontic strain in vitro. MiR-34a also enhanced alveolar bone remodeling under orthodontic force in vivo, as evidenced by elevated gene and protein expression, upregulated indices of alveolar bone anabolism, and diminished tooth movement. We determined that the mechanism miR-34a in osteogenesis under orthodontic force may be associated with GSK-3β. These results suggested that miR-34a delivered by N-Ac-l-Leu-PEI could be a potential therapeutic target for orthodontic treatment.
Collapse
Affiliation(s)
- Wenwen Yu
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Yi Zheng
- Department of Periodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Zhujun Yang
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Hongbo Fei
- Department of Periodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Yang Wang
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xu Hou
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xinhua Sun
- Department of Orthodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Yuqin Shen
- Department of Periodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| |
Collapse
|
21
|
Morin MD, Frigault JJ, Lyons PJ, Crapoulet N, Boquel S, Storey KB, Morin PJ. Amplification and quantification of cold-associated microRNAs in the Colorado potato beetle (Leptinotarsa decemlineata) agricultural pest. INSECT MOLECULAR BIOLOGY 2017; 26:574-583. [PMID: 28574638 DOI: 10.1111/imb.12320] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The Colorado potato beetle [Leptinotarsa decemlineata (Say)] is an important insect pest that can inflict considerable damage to potato plants. This insect can survive extended periods of cold exposure, and yet the molecular switches underlying this phenomenon have not been fully elucidated. A better characterization of this process would highlight novel vulnerabilities associated with L. decemlineata that could serve as targets for the management of this devastating pest. Using high-throughput sequencing, the current work reveals a cold-associated signature group of microRNAs (miRNAs) in control (15 °C) and -5 °C-exposed L. decemlineata. The results show 42 differentially expressed miRNAs following cold exposure including miR-9a-3p, miR-210-3p, miR-276-5p and miR-277-3p. Functional analysis of predicted targets associated with these cold-responsive miRNAs notably linked these changes with vital metabolic and cellular processes. Overall, this study highlights the miRNAs probably responsible for facilitating cold adaptation in L. decemlineata and implicates miRNAs as a key molecular target to consider in the development of novel pest management strategies against these insects.
Collapse
Affiliation(s)
- M D Morin
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
| | - J J Frigault
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
| | - P J Lyons
- Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - N Crapoulet
- Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - S Boquel
- Agriculture and Agri-Food Canada, Fredericton Research and Development Centre, Fredericton, NB, Canada
| | - K B Storey
- Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
| | - P Jr Morin
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
| |
Collapse
|
22
|
Song S, Lin F, Zhu P, Wu C, Zhao S, Han Q, Li X. Extract of Spatholobus suberctus Dunn ameliorates ischemia-induced injury by targeting miR-494. PLoS One 2017; 12:e0184348. [PMID: 28880896 PMCID: PMC5589225 DOI: 10.1371/journal.pone.0184348] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/22/2017] [Indexed: 11/28/2022] Open
Abstract
Cerebral stroke is a leading cause of death and permanent disability. The current therapeutic outcome of ischemic stroke (>85% of all strokes) is very poor, thus novel therapeutic drug is urgently needed. In vitro cell model of ischemia was established by oxygen-glucose deprivation (OGD) and in vivo animal model of ischemia was established by middle cerebral artery occlusion (MCAO). The effects of Spatholobus suberctus Dunn extract (SSCE) on OGD-induced cell injury, MCAO-induced neural injury and miR-494 level were all evaluated. The possible target genes were virtually screened utilizing bioinformatics and verified by luciferase assay. Subsequently, the effects of abnormally expressed miR-494 on OGD-induced cell injury and target gene expression were determined. Additionally, whether SSCE affected target gene expression through modulation of miR-494 was studied. Finally, the effects of aberrantly expressed Sox8 on OGD-induced injury and signaling pathways were estimated. SSCE reduced OGD-induced cell injury and ameliorated MCAO-induced neuronal injury, along with down-regulation of miR-494. Then, OGD-induced cell injury was increased by miR-494 overexpression but decreased by miR-494 silence. Sox8 was a target gene of miR-494, and SSCE could up-regulate Sox8 expression via down-regulating miR-494. Afterwards, OGD-induced cell injury was proved to be increased by Sox8 inhibition but reduced by Sox8 overexpression. Finally, OGD-induced inhibition of PI3K/AKT/mTOR and MAPK pathways was further inhibited by Sox8 silence but activated by Sox8 overexpression. SSCE ameliorates ischemia-induced injury both in vitro and in vivo by miR-494-mediated modulation of Sox8, involving activations of PI3K/AKT/mTOR and MAPK pathways.
Collapse
Affiliation(s)
- Shiqing Song
- Yantai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Faliang Lin
- Yantai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Pengyan Zhu
- Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Changyan Wu
- Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Shuling Zhao
- Yantai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Qiao Han
- Yantai Blood Center, Yantai, Shandong, China
| | - Xiaomei Li
- Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| |
Collapse
|
23
|
Mechanosensitive miRNAs and Bone Formation. Int J Mol Sci 2017; 18:ijms18081684. [PMID: 28767056 PMCID: PMC5578074 DOI: 10.3390/ijms18081684] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/25/2017] [Accepted: 07/28/2017] [Indexed: 12/18/2022] Open
Abstract
Mechanical stimuli are required for the maintenance of skeletal integrity and bone mass. An increasing amount of evidence indicates that multiple regulators (e.g., hormone, cytoskeleton proteins and signaling pathways) are involved in the mechanical stimuli modulating the activities of osteogenic cells and the process of bone formation. Significantly, recent studies have showed that several microRNAs (miRNAs) were sensitive to various mechanical stimuli and played a crucial role in osteogenic differentiation and bone formation. However, the functional roles and further mechanisms of mechanosensitive miRNAs in bone formation are not yet completely understood. This review highlights the roles of mechanosensitive miRNAs in osteogenic differentiation and bone formation and underlines their potential therapeutic application for bone loss induced by the altering of mechanical stimuli.
Collapse
|
24
|
Zhang L, Tan W, Zhou J, Xu M, Yuan G. Investigation of G-quadruplex formation in the FGFR2 promoter region and its transcriptional regulation by liensinine. Biochim Biophys Acta Gen Subj 2017; 1861:884-891. [PMID: 28132898 DOI: 10.1016/j.bbagen.2017.01.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/21/2017] [Accepted: 01/25/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Fibroblast growth factor receptor 2 (FGFR2) is overexpressed in breast cancer tissues and cells, and has been shown to be a susceptibility factor for breast cancer. In this study, we found that the G-rich sequences in the FGFR2 promoter region can form G-quadruplexes, which could be the target and inhibitor of the FGFR2 gene. METHODS Initially, the formation of G-quadruplexes was confirmed by ESI-MS and CD, and DMS footprinting experiments gave the folding pattern of the G-quadruplexes. After luciferase reporter assays revealed that the G-quadruplex could inhibit the activity of the FGFR2 promoter, MS and SPR showed binding affinity and selectivity of the ligand. Then cell culture experiments and ChIP assay showed the bioactivity of the ligand. RESULTS We found that three G-rich sequences (S1-S3) in the FGFR2 promoter region can form G-quadruplex structures. And a natural alkaloid, liensinine, was found to bind to the S1 G-quadruplex with relative high affinity and selectivity. Cell culture experiments showed that liensinine inhibits FGFR2 activity at both the transcriptional and translational levels. Moreover, chromatin immunoprecipitation assay (ChIP) results showed that liensinine blocks the binding of E2F1 at the transcription factor binding site (TFBS) in the S1 sequence, which is the mechanism through which liensinine inhibits the FGFR2 gene. CONCLUSIONS A natural alkaloid was discovered to selectively bind to the S1 G-quadruplex with relative high affinity, and therefore inhibited FGFR2 transcription and translation. GENERAL SIGNIFICANCE Our discovery offers a useful strategy to inhibit FGFR2 transcription, i.e., targeting the G-quadruplex with a natural alkaloid.
Collapse
Affiliation(s)
- Lulu Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Wei Tan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jiang Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China; Analytical Instrumentation Center, Peking University, Beijing 100871, China.
| | - Ming Xu
- Institute of Vascular Medicine, Department of Cardiology, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Beijing 100191, China.
| | - Gu Yuan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| |
Collapse
|
25
|
Yuan Y, Zhang L, Tong X, Zhang M, Zhao Y, Guo J, Lei L, Chen X, Tickner J, Xu J, Zou J. Mechanical Stress Regulates Bone Metabolism Through MicroRNAs. J Cell Physiol 2016; 232:1239-1245. [PMID: 27861865 DOI: 10.1002/jcp.25688] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 11/11/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Yu Yuan
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
- School of Pathology and Laboratory Medicine; the University of Western Australia; Perth Western Australia Australia
| | - Lingli Zhang
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
| | - Xiaoyang Tong
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
| | - Miao Zhang
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
| | - Yilong Zhao
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
| | - Jianming Guo
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
| | - Le Lei
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
| | - Xi Chen
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
- School of Sports Science; Wenzhou Medical University; Wenzhou China
| | - Jennifer Tickner
- School of Pathology and Laboratory Medicine; the University of Western Australia; Perth Western Australia Australia
| | - Jiake Xu
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
- School of Pathology and Laboratory Medicine; the University of Western Australia; Perth Western Australia Australia
| | - Jun Zou
- School of Kinesiology; Shanghai University of Sport; Shanghai P. R. China
| |
Collapse
|
26
|
Hadjiargyrou M, Zhi J, Komatsu DE. Identification of the microRNA transcriptome during the early phases of mammalian fracture repair. Bone 2016; 87:78-88. [PMID: 27058875 DOI: 10.1016/j.bone.2016.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/10/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Fracture repair is a complex process that involves multiple biological processes requiring spatiotemporal expression of thousands of genes. The molecular regulation of this process is not completely understood. MicroRNAs (miRNAs) regulate gene expression by promoting mRNA degradation or blocking translation. To identify miRNAs expressed during fracture repair, we generated murine bone fractures and isolated miRNA-enriched RNA from intact and post-fracture day (PFD) 1, 3, 5, 7, 11, and 14 femurs. RNA samples were individually hybridized to mouse miRNA microarrays. Results indicated that 959 (51%) miRNAs were absent while 922 (49%) displayed expression in at least one sample. Of the 922 miRNAs, 306 (33.2%) and 374 (40.6%) were up- and down-regulated, respectively, in the calluses in comparison to intact bone. Additionally, 20 (2.2%) miRNAs displayed combined up- and down-regulated expression within the time course and the remaining 222 (24%) miRNAs did not exhibit any changes between calluses and intact bone. Quantitative-PCR validated the expression of several miRNAs. Further, we identified 2048 and 4782 target genes that were unique to the up- and down-regulated miRNAs, respectively. Gene ontology and pathway enrichment analyses indicated relevant biological processes. These data provide the first complete analysis of the miRNA transcriptome during the early phases of fracture repair.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, Theobald Science Center, Room 420, New York Institute of Technology, Old Westbury, NY 11568-8000, USA.
| | - Jizu Zhi
- Bioinformatics Core Facility, Stony Brook University, Stony Brook, NY 11794, USA.
| | - David E Komatsu
- Department of Orthopaedics, HSC T18 Room 85, Stony Brook University, Stony Brook, NY 11794-8181, USA.
| |
Collapse
|
27
|
Tay J, Tiao J, Hughes Q, Gilmore G, Baker R. Therapeutic Potential of miR-494 in Thrombosis and Other Diseases: A Review. Aust J Chem 2016. [DOI: 10.1071/ch16020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Functional nucleic acids, such as microRNAs (miRNAs), have been implicated in the pathophysiology of many diseases. The miRNA expression profiles of various cancers including haematological malignancies are well defined, but the role of miRNAs in haemostasis and the regulation of coagulation is poorly understood. We identified that miR-494 is oestrogen responsive and directly targets the anticoagulant protein, Protein S, as a mechanism for acquiring Protein S deficiency under high oestrogenic conditions such as during pregnancy and oral contraceptive use. Furthermore, previous studies have also characterised miR-494 to be involved in many biological processes. This paper reviews the current knowledge in the role of miRNAs in regulating haemostatic proteins and the known biological functions of miR-494, highlighting miR-494 as an emerging therapeutic target, with an overview of the strategy we have employed in identifying functional nucleic acids such as miRNAs that target haemostatic factors and the therapeutic potential of miR-494-directed therapy for the treatment of thrombotic disorders.
Collapse
|
28
|
Ji Z, Dong F, Wang G, Hou L, Liu Z, Chao T, Wang J. miR-135a Targets and Regulates Prolactin Receptor Gene in Goat Mammary Epithelial Cells. DNA Cell Biol 2015; 34:534-40. [PMID: 26102062 DOI: 10.1089/dna.2015.2904] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mammary gland development and lactation are typical traits controlled by multiple genes, hormones, and regulatory factors. Prolactin receptor (PRLR), a specific receptor of prolactin, has been reported to have important physiological functions in regulating mammogenesis and lactogenesis. However, the post-transcriptional regulation mechanisms of PRLR expression have not yet been shown in detail. In this study, the expression of miR-135a and PRLR at different development stages of Laoshan dairy goat mammary gland tissues was investigated. After overexpression and silencing expression of miR-135a in cultured primary mammary epithelial cells, the regulatory relationship between miR-135a and PRLR was examined through dual-luciferase reporter assay, and the expression of PRLR at both mRNA and protein levels was examined by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot. Collectively, our results suggested that PRLR is a direct target gene of miR-135a, miR-135a is a novel regulator of PRLR, and it might play an essential role in the regulation of animal mammary gland development and lactation.
Collapse
Affiliation(s)
- Zhibin Ji
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University , Taian, China
| | - Fei Dong
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University , Taian, China
| | - Guizhi Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University , Taian, China
| | - Lei Hou
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University , Taian, China
| | - Zhaohua Liu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University , Taian, China
| | - Tianle Chao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University , Taian, China
| | - Jianmin Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University , Taian, China
| |
Collapse
|