1
|
Xu L, Zhang L, Sun Q, Zhang X, Zhang J, Zhao X, Hu Z, Zhang S, Shi F. Melatonin antagonizes bone loss induced by mechanical unloading via IGF2BP1-dependent m 6A regulation. Cell Mol Life Sci 2025; 82:60. [PMID: 39849105 PMCID: PMC11757843 DOI: 10.1007/s00018-025-05588-x] [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: 08/21/2024] [Revised: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025]
Abstract
Disuse bone loss is prone to occur in individuals who lack mechanical stimulation due to prolonged spaceflight or extended bed rest, rendering them susceptible to fractures and placing an enormous burden on social care; nevertheless, the underlying molecular mechanisms of bone loss caused by mechanical unloading have not been fully elucidated. Numerous studies have focused on the epigenetic regulation of disuse bone loss; yet limited research has been conducted on the impact of RNA modification bone formation in response to mechanical unloading conditions. In this study, we discovered that m6A reader IGF2BP1 was downregulated in both osteoblasts treated with 2D clinostat and bone tissue in HLU mice. Supplementing IGF2BP1 could promote osteoblast proliferation and partially alleviate the adverse effects of mechanical unloading on bone formation. Mechanistically, IGF2BP1 inhibited the degradation of Lef1 mRNA by directly binding to its mRNA and recognizing the m6A modification. Furthermore, LEF1 promoted osteoblast proliferation by upregulating c-Myc and Cyclin D1 expression, as well as participated in mediating IGF2BP1-induced osteoblast activity under mechanical unloading. Notably, Melatonin (MT) might participate in the regulation of the IGF2BP1/LEF1 axis, thereby regulating the proliferation of osteoblasts and bone formation. Collectively, this study revealed a new insight into the regulation of the MT/IGF2BP1/LEF1 pathway in the process of unloading-induced bone loss, which could potentially contribute to establishing therapeutic strategies for disuse osteoporosis.
Collapse
Affiliation(s)
- Liqun Xu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Lijun Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
- Department of Otolaryngology Head and Neck Surgery, Bethune International Peace Hospital, Shijiazhuang, 050081, Hebei, China
| | - Quan Sun
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiaoyan Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
- Department of Otolaryngology Head and Neck Surgery, Western Theater Air Force Hospital of PLA, Chengdu, 610065, Sichuan, China
| | - Junfei Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiran Zhao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
2
|
Wu X, Shen T, Ji W, Huang M, Sima J, Li J, Song H, Xiong W, Cen M. lncRNA CASC11 regulates the progress of delayed fracture healing via sponging miR-150-3p. J Orthop Surg Res 2024; 19:757. [PMID: 39543626 PMCID: PMC11562309 DOI: 10.1186/s13018-024-05226-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) plays a pivotal role in bone regeneration by interaction with microRNAs (miRNAs) and constructing a lncRNA-miRNA regulatory network. OBJECTIVES This research aimed to elucidate the role of lncRNA CASC11 in the delayed healing process of tibial fractures and to explore its potential regulatory mechanisms. MATERIALS AND METHODS The expression levels of CASC11 and miR-150-3p in serum samples were detected and the predictive capability of CASC11 regarding delayed healing in fracture patients. Furthermore, the study confirmed the accuracy of the binding sites between CASC11 and miR-150-3p. Subsequently, overexpression/interference plasmids of CASC11, along with overexpression plasmids co-transfected with both CASC11 and miR-150-3p, were systematically introduced into MC3T3-E1 cells to investigate their effects on the expression of osteogenic marker genes, as well as their influence on cellular proliferation and apoptosis. RESULTS The expression levels of CASC11 were significantly elevated, while miR-150-3p levels were markedly decreased in individuals exhibiting delayed fracture healing (P < 0.001). CASC11 was observed to suppress the expression of osteogenic marker genes, inhibit the proliferation of MC3T3-E1 cells, and promote cell apoptosis (P < 0.05). Furthermore, the overexpression of miR-150-3p effectively countered the inhibitory impact of CASC11 on osteogenic differentiation and the promoting effect on cell apoptosis (P < 0.05). CONCLUSION The sponging effect of CASC11 on miR-150-3p led to delayed fracture healing. CASC11 emerges as a potential target for treating delayed fracture healing.
Collapse
Affiliation(s)
- Xiaoming Wu
- Orthopedics, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Tuwang Shen
- Operating Room, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, China
| | - Wenjun Ji
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, Guizhou Province, 563000, China
| | - Miao Huang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, Guizhou Province, 563000, China
| | - Jincheng Sima
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, Guizhou Province, 563000, China
| | - Jin Li
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, Guizhou Province, 563000, China
| | - Hao Song
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, Guizhou Province, 563000, China
| | - Wei Xiong
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, Guizhou Province, 563000, China.
| | - Meini Cen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, Guangxi, 533000, China.
| |
Collapse
|
3
|
Li JX, Qu YD, Xia CL, Zhang W, Wang SS, Ou SJ, Yang Y, Qi Y, Xu CP. Analysis of PANoptosis-related ceRNA network reveals lncRNA MIR17HG involved in osteogenic differentiation inhibition impaired by tumor necrosis factor-α. Mol Biol Rep 2024; 51:909. [PMID: 39145884 PMCID: PMC11327206 DOI: 10.1007/s11033-024-09810-0] [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: 04/28/2024] [Accepted: 07/19/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Inflammatory cytokines such as Interleukin 1β(IL1β), IL6,Tumor Necrosis Factor-α (TNF-α) can inhibit osteoblast differentiation and induce osteoblast apoptosis. PANoptosis, a newly identified type of programmed cell death (PCD), may be influenced by long noncoding RNA (lncRNAs) which play important roles in regulating inflammation. However, the potential role of lncRNAs in inflammation and PANoptosis during osteogenic differentiation remains unclear. This study aimed to investigate the regulatory functions of lncRNAs in inflammation and apoptosis during osteogenic differentiation. METHODS AND RESULTS High-throughput sequencing was used to identify differentially expressed genes involved in osteoblast differentiation under inflammatory conditions. Two lncRNAs associated with inflammation and PANoptosis during osteogenic differentiation were identified from sequencing data and Gene Expression Omnibus (GEO) databases. Their functionalities were analyzed using diverse bioinformatics methodologies, resulting in the construction of the lncRNA-miRNA-mRNA network. Among these, lncRNA (MIR17HG) showed a high correlation with PANoptosis. Bibliometric methods were employed to collect literature data on PANoptosis, and its components were inferred. PCR and Western Blotting experiments confirmed that lncRNA MIR17HG is related to PANoptosis in osteoblasts during inflammation. CONCLUSIONS Our data suggest that TNF-α-induced inhibition of osteogenic differentiation and PANoptosis in MC3T3-E1 osteoblasts is associated with MIR17HG. These findings highlight the critical role of MIR17HG in the interplay between inflammation, PANoptosis, and osteogenic differentiation, suggesting potential therapeutic targets for conditions involving impaired bone formation and inflammatory responses.
Collapse
Affiliation(s)
- Jia-Xuan Li
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu-Dun Qu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Chang-Liang Xia
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Wei Zhang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
- Department of Orthopaedics, Southern Medical University, Guangzhou, Guangdong, China
| | - Song-Song Wang
- School of Medicine, XiaMen University, Xiamen, Fujian, China
| | - Shuan-Ji Ou
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yang Yang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yong Qi
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| | - Chang-Peng Xu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, No. 466 Xingang Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| |
Collapse
|
4
|
Han J, Zhu Y, Zhang J, Kapilevich L, Zhang XA. Noncoding RNAs: the crucial role of programmed cell death in osteoporosis. Front Cell Dev Biol 2024; 12:1409662. [PMID: 38799506 PMCID: PMC11116712 DOI: 10.3389/fcell.2024.1409662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Osteoporosis is the most common skeletal disease characterized by an imbalance between bone resorption and bone remodeling. Osteoporosis can lead to bone loss and bone microstructural deterioration. This increases the risk of bone fragility and fracture, severely reducing patients' mobility and quality of life. However, the specific molecular mechanisms involved in the development of osteoporosis remain unclear. Increasing evidence suggests that multiple noncoding RNAs show differential expression in the osteoporosis state. Meanwhile, noncoding RNAs have been associated with an increased risk of osteoporosis and fracture. Noncoding RNAs are an important class of factors at the level of gene regulation and are mainly involved in cell proliferation, cell differentiation, and cell death. Programmed cell death is a genetically-regulated form of cell death involved in regulating the homeostasis of the internal environment. Noncoding RNA plays an important role in the programmed cell death process. The exploration of the noncoding RNA-programmed cell death axis has become an interesting area of research and has been shown to play a role in many diseases such as osteoporosis. In this review, we summarize the latest findings on the mechanism of noncoding RNA-mediated programmed cell death on bone homeostasis imbalance leading to osteoporosis. And we provide a deeper understanding of the role played by the noncoding RNA-programmed cell death axis at the gene regulatory level of osteoporosis. We hope to provide a unique opportunity to develop novel diagnostic and therapeutic approaches for osteoporosis.
Collapse
Affiliation(s)
- Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yuqing Zhu
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Jiale Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Leonid Kapilevich
- Faculty of Physical Education, Tomsk Stаte University, Tomsk, Russia
| | - Xin-an Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
5
|
Zhang Z, Jing Y, Zhang A, Liu J, Yang H, Lou X, Xu L, Liu M, Zhang Y, Gu J. Long non-coding RNA-NONMMMUT004552.2 regulates the unloading-induced bone loss through the miRNA-15b-5p/Syne1 in mice. NPJ Microgravity 2024; 10:37. [PMID: 38521778 PMCID: PMC10960867 DOI: 10.1038/s41526-024-00382-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
Exercise-induced mechanical loading can increase bone strength whilst mechanical unloading enhances bone-loss. Here, we investigated the role of lncRNA NONMMUT004552.2 in unloading-induced bone-loss. Knockout of lncRNA NONMMUT004552.2 in hindlimb-unloaded mice caused an increase in the bone formation and osteoblast activity. The silencing of lncRNA NONMMUT004552.2 also decreased the osteoblast apoptosis and expression of Bax and cleaved caspase-3, increased Bcl-2 protein expression in MC3T3-E1 cells. Mechanistic investigations demonstrated that NONMMUT004552.2 functions as a competing endogenous RNA (ceRNA) to facilitate the protein expression of spectrin repeat containing, nuclear envelope 1 (Syne1) by competitively binding miR-15b-5p and subsequently inhibits the osteoblast differentiation and bone formation in the microgravity unloading environment. These data highlight the importance of the lncRNA NONMMUT004552.2/miR-15b-5p/Syne1 axis for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Medical Engineering, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Yu Jing
- Department of Haematology, The Fifth Medical Centre of Chinese PLA General Hospital, Beijing, 100071, China
| | - Ang Zhang
- Department of Hematology, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - JiShan Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Heming Yang
- Department of General Surgery, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Xiaotong Lou
- Department of Research, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Liyan Xu
- Department of Blood Transfusion, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Min Liu
- School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Yikun Zhang
- Department of Hematology, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China.
| | - Jianwen Gu
- Department of Neurosurgery, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China.
| |
Collapse
|
6
|
Baniasadi M, Talebi S, Mokhtari K, Zabolian AH, Khosroshahi EM, Entezari M, Dehkhoda F, Nabavi N, Hashemi M. Role of non-coding RNAs in osteoporosis. Pathol Res Pract 2024; 253:155036. [PMID: 38134836 DOI: 10.1016/j.prp.2023.155036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/10/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023]
Abstract
Osteoporosis, a prevalent bone disorder influenced by genetic and environmental elements, significantly increases the likelihood of fractures and bone weakness, greatly affecting the lives of those afflicted. Yet, the exact epigenetic processes behind the onset of osteoporosis are still unclear. Growing research indicates that epigenetic changes could act as vital mediators that connect genetic tendencies and environmental influences, thereby increasing the risk of osteoporosis and bone fractures. Within these epigenetic factors, certain types of RNA, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been recognized as key regulatory elements. These RNA types wield significant influence on gene expression through epigenetic regulation, directing various biological functions essential to bone metabolism. This extensive review compiles current research uncovering the complex ways in which miRNAs, lncRNAs, and circRNAs are involved in the development of osteoporosis, especially in osteoblasts and osteoclasts. Gaining a more profound understanding of the roles these three RNA classes play in osteoporosis could reveal new diagnostic methods and treatment approaches for this incapacitating condition. In conclusion, this review delves into the complex domain of epigenetic regulation via non-coding RNA in osteoporosis. It sheds light on the complex interactions and mechanisms involving miRNAs, lncRNAs, and circRNAs within osteoblasts and osteoclasts, offering an in-depth understanding of the less explored aspects of osteoporosis pathogenesis. These insights not only reveal the complexity of the disease but also offer significant potential for developing new diagnostic methods and targeted treatments. Therefore, this review marks a crucial step in deciphering the elusive complexities of osteoporosis, leading towards improved patient care and enhanced quality of life.
Collapse
Affiliation(s)
- Mojtaba Baniasadi
- Department of Orthopedics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sina Talebi
- Department of Orthopedics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Khatere Mokhtari
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran; Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan,Iran
| | - Amir Hossein Zabolian
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Farshid Dehkhoda
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Noushin Nabavi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
7
|
Li W, Yu Y, Zhou G, Hu G, Li B, Ma H, Yan W, Pei H. Large-scale ORF screening based on LC-MS to discover novel lncRNA-encoded peptides responding to ionizing radiation and microgravity. Comput Struct Biotechnol J 2023; 21:5201-5211. [PMID: 37928948 PMCID: PMC10624585 DOI: 10.1016/j.csbj.2023.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 11/07/2023] Open
Abstract
In the human genome, 98% of genes can be transcribed into non-coding RNAs (ncRNAs), among which lncRNAs and their encoded peptides play important roles in regulating various aspects of cellular processes and may serve as crucial factors in modulating the biological effects induced by ionizing radiation and microgravity. Unfortunately, there are few reports in space radiation biology on lncRNA-encoded peptides below 10kD due to limitations in detection techniques. To fill this gap, we integrated a variety of methods based on genomics and peptidomics, and discovered 22 lncRNA-encoded small peptides that are sensitive to space radiation and microgravity, which have never been reported before. We concurrently validated the transmembrane helix, subcellular localization, and biological function of these small peptides using bioinformatics and molecular biology techniques. More importantly, we found that these small peptides function independently of the lncRNAs that encode them. Our findings have uncovered a previously unknown human proteome encoded by 'non-coding' genes in response to space conditions and elucidated their involvement in biological processes, providing valuable strategies for individual protection mechanisms for astronauts who carry out deep space exploration missions in space radiation environments.
Collapse
Affiliation(s)
- Wanshi Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yongduo Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guang Hu
- Department of Bioinformatics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
- Center for Systems Biology, Soochow University, Suzhou 215123, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou 215123, China
| | - Bingyan Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Hong Ma
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Wenying Yan
- Department of Bioinformatics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou 215123, China
- Center for Systems Biology, Soochow University, Suzhou 215123, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou 215123, China
| | - Hailong Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
8
|
Cai Z, Liu F, Li Y, Bai L, Feng M, Li S, Ma W, Shi S. Functional micro-RNA drugs acting as a fate manipulator in the regulation of osteoblastic death. NANOSCALE 2023; 15:12840-12852. [PMID: 37482769 DOI: 10.1039/d3nr02318d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Bone loss is prevalent in clinical pathological phenomena such as osteoporosis, which is characterized by decreased osteoblast function and number, increased osteoclast activity, and imbalanced bone homeostasis. However, current treatment strategies for bone diseases are limited. Regulated cell death (RCD) is a programmed cell death pattern activated by the expression of specific genes in response to environmental changes. Various studies have shown that RCD is closely associated with bone diseases, and manipulating the death fate of osteoblasts could contribute to effective bone treatment. Recently, microRNA-targeting therapy drugs have emerged as a potential solution because of their precise targeting, powerful curative effect, and limited side effects. Nevertheless, their clinical application is limited by their inherent instability, easy enzymatic degradation, and poor membrane penetrability. To address this challenge, a self-assembling tetrahedral DNA nanostructure (TDN)-based microRNA (Tmi) delivery system has been proposed. TDN features excellent biocompatibility, cell membrane penetrability, serum stability, and modification versatility, making it an ideal nucleic acid carrier for miRNA protection and intracellular transport. Once inside cells, Tmi can dissociate and release miRNAs to manipulate key molecules in the RCD signaling pathway, thereby regulating bone homeostasis and curing diseases caused by abnormal RCD activation. In this paper, we discuss the impact of the miRNA network on the initiation and termination of four critical RCD programs in bone tissues: apoptosis, autophagy, pyroptosis, and ferroptosis. Furthermore, we present the Tmi delivery system as a miRNA drug vector. This provides insight into the clinical translation of miRNA nucleic acid drugs and the application of miRNA drugs in bone diseases.
Collapse
Affiliation(s)
- Zhengwen Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Fengshuo Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Yong Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Long Bai
- The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Maogeng Feng
- The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Songhang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China
| |
Collapse
|
9
|
Meng F, Yu Y, Tian Y, Deng M, Zheng K, Guo X, Zeng B, Li J, Qian A, Yin C. A potential therapeutic drug for osteoporosis: prospect for osteogenic LncRNAs. Front Endocrinol (Lausanne) 2023; 14:1219433. [PMID: 37600711 PMCID: PMC10435887 DOI: 10.3389/fendo.2023.1219433] [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: 05/09/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Long non-coding RNAs (LncRNAs) play essential roles in multiple physiological processes including bone formation. Investigators have revealed that LncRNAs regulated bone formation through various signaling pathways and micro RNAs (miRNAs). However, several problems exist in current research studies on osteogenic LncRNAs, including sophisticated techniques, high cost for in vivo experiment, as well as low homology of LncRNAs between animal model and human, which hindered translational medicine research. Moreover, compared with gene editing, LncRNAs would only lead to inhibition of target genes rather than completely knocking them out. As the studies on osteogenic LncRNA gradually proceed, some of these problems have turned osteogenic LncRNA research studies into slump. This review described some new techniques and innovative ideas to address these problems. Although investigations on osteogenic LncRNAs still have obtacles to overcome, LncRNA will work as a promising therapeutic drug for osteoporosis in the near future.
Collapse
Affiliation(s)
- Fanjin Meng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Yang Yu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ye Tian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Meng Deng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Kaiyuan Zheng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
| | - Beilei Zeng
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jingjia Li
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| | - Chong Yin
- Department of Clinical Laboratory, Department of Oncology, Department of Rehabilitation Medicine, Ministry of Science and Technology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Department of Laboratory Medicine, Translational Medicine Research Center, North Sichuan Medical College, Nanchong, China
- Lab for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an, Shaanxi, China
| |
Collapse
|
10
|
Methyltransferase Setdb1 Promotes Osteoblast Proliferation by Epigenetically Silencing Macrod2 with the Assistance of Atf7ip. Cells 2022; 11:cells11162580. [PMID: 36010655 PMCID: PMC9406310 DOI: 10.3390/cells11162580] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/31/2022] [Accepted: 08/09/2022] [Indexed: 12/03/2022] Open
Abstract
Bone loss caused by mechanical unloading is a threat to prolonged space flight and human health. Epigenetic modifications play a crucial role in varied biological processes, but the mechanism of histone modification on unloading-induced bone loss has rarely been studied. Here, we discovered for the first time that the methyltransferase Setdb1 was downregulated under the mechanical unloading both in vitro and in vivo so as to attenuate osteoblast proliferation. Furthermore, we found these interesting processes depended on the repression of Macrod2 expression triggered by Setdb1 catalyzing the formation of H3K9me3 in the promoter region. Mechanically, we revealed that Macrod2 was upregulated under mechanical unloading and suppressed osteoblast proliferation through the GSK-3β/β-catenin signaling pathway. Moreover, Atf7ip cooperatively contributed to osteoblast proliferation by changing the localization of Setdb1 under mechanical loading. In summary, this research elucidated the role of the Atf7ip/Setdb1/Macrod2 axis in osteoblast proliferation under mechanical unloading for the first time, which can be a potential protective strategy against unloading-induced bone loss.
Collapse
|
11
|
Wang Y, Zhu X, Yang L, Hu X, He K, Yu C, Jiao S, Chen J, Guo R, Yang S. IDDLncLoc: Subcellular Localization of LncRNAs Based on a Framework for Imbalanced Data Distributions. Interdiscip Sci 2022; 14:409-420. [PMID: 35192174 DOI: 10.1007/s12539-021-00497-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
Long non-coding RNAs play a crucial role in many life processes of cell, such as genetic markers, RNA splicing, signaling, and protein regulation. Considering that identifying lncRNA's localization in the cell through experimental methods is complicated, hard to reproduce, and expensive, we propose a novel method named IDDLncLoc in this paper, which adopts an ensemble model to solve the problem of the subcellular localization. In the proposal model, dinucleotide-based auto-cross covariance features, k-mer nucleotide composition features, and composition, transition, and distribution features are introduced to encode a raw RNA sequence to vector. To screen out reliable features, feature selection through binomial distribution, and recursive feature elimination is employed. Furthermore, strategies of oversampling in mini-batch, random sampling, and stacking ensemble strategies are customized to overcome the problem of data imbalance on the benchmark dataset. Finally, compared with the latest methods, IDDLncLoc achieves an accuracy of 94.96% on the benchmark dataset, which is 2.59% higher than the best method, and the results further demonstrate IDDLncLoc is excellent on the subcellular localization of lncRNA. Besides, a user-friendly web server is available at http://lncloc.club .
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
- School of Artificial Intelligence, Jilin University, Changchun, China
| | - Xiaopeng Zhu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
| | - Lili Yang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, China
| | - Xuemei Hu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
| | - Kai He
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
| | - Cuinan Yu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
| | - Shaoqing Jiao
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
| | - Jiali Chen
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
| | - Rui Guo
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China
| | - Sen Yang
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, China.
| |
Collapse
|
12
|
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
|
13
|
Long noncoding RNA Lnc-DIF inhibits bone formation by sequestering miR-489-3p. iScience 2022; 25:103949. [PMID: 35265818 PMCID: PMC8898894 DOI: 10.1016/j.isci.2022.103949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 01/06/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022] Open
Abstract
Osteoporosis has become a high incident bone disease along with the aging of human population. Long noncoding RNAs (LncRNAs) play an important role in osteoporosis incidence. In this study, we screened out an LncRNA negatively correlated with osteoblast differentiation, which was therefore named Lnc-DIF (differentiation inhibiting factor). Functional analysis proved that Lnc-DIF inhibited bone formation. A special structure containing multiple 53 nucleotide repeats was found in the trailing end of Lnc-DIF. Our study suggested that this repeat sequence could sequester multiple miR-489-3p and inhibit bone formation through miR-489-3p/SMAD2 axis. Moreover, siRNA of Lnc-DIF would rescue bone formation in both aging and ovariectomized osteoporosis mice. This study revealed a kind of LncRNA that could function as a sponge and regulate multiple miRNAs. RNA therapy techniques that target these LncRNAs could manipulate its downstream miRNA-target pathway with significantly higher efficiency and specificity. This provided potential therapeutic insight for RNA-based therapy for osteoporosis. Identified LncRNA Lnc-DIF that inhibited bone formation Lnc-DIF sequestered multiple miR-489-3p by the repeat sequences on its trailing end Lnc-DIF repeat sequence inhibited bone formation via miR-489-3p/SMAD2 axis Lnc-DIF siRNA showed strong capability on rescuing osteoporosis
Collapse
|
14
|
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: 30] [Impact Index Per Article: 10.0] [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
|
15
|
Cialdai F, Bolognini D, Vignali L, Iannotti N, Cacchione S, Magi A, Balsamo M, Vukich M, Neri G, Donati A, Monici M, Capaccioli S, Lulli M. Effect of space flight on the behavior of human retinal pigment epithelial ARPE-19 cells and evaluation of coenzyme Q10 treatment. Cell Mol Life Sci 2021; 78:7795-7812. [PMID: 34714361 PMCID: PMC11073052 DOI: 10.1007/s00018-021-03989-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/20/2021] [Accepted: 10/13/2021] [Indexed: 10/20/2022]
Abstract
Astronauts on board the International Space Station (ISS) are exposed to the damaging effects of microgravity and cosmic radiation. One of the most critical and sensitive districts of an organism is the eye, particularly the retina, and > 50% of astronauts develop a complex of alterations designated as spaceflight-associated neuro-ocular syndrome. However, the pathogenesis of this condition is not clearly understood. In the current study, we aimed to explore the cellular and molecular effects induced in the human retinal pigment ARPE-19 cell line by their transfer to and 3-day stay on board the ISS in the context of an experiment funded by the Agenzia Spaziale Italiana. Treatment of cells on board the ISS with the well-known bioenergetic, antioxidant, and antiapoptotic coenzyme Q10 was also evaluated. In the ground control experiment, the cells were exposed to the same conditions as on the ISS, with the exception of microgravity and radiation. The transfer of ARPE-19 retinal cells to the ISS and their living on board for 3 days did not affect cell viability or apoptosis but induced cytoskeleton remodeling consisting of vimentin redistribution from the cellular boundaries to the perinuclear area, underlining the collapse of the network of intermediate vimentin filaments under unloading conditions. The morphological changes endured by ARPE-19 cells grown on board the ISS were associated with changes in the transcriptomic profile related to the cellular response to the space environment and were consistent with cell dysfunction adaptations. In addition, the results obtained from ARPE-19 cells treated with coenzyme Q10 indicated its potential to increase cell resistance to damage.
Collapse
Affiliation(s)
- Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, Firenze, Italy
| | - Davide Bolognini
- Department of Experimental and Clinical Medicine, Università Degli Studi Di Firenze, Firenze, Italy
| | - Leonardo Vignali
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, Firenze, Italy
| | - Nicola Iannotti
- Department of Life Sciences, Università Degli Studi Di Siena, Siena, Italy
| | - Stefano Cacchione
- Department of Biology and Biotechnology "Charles Darwin", Università Di Roma "La Sapienza", Roma, Italy
| | - Alberto Magi
- Department of Information Engineering, Università Degli Studi Di Firenze, Firenze, Italy
| | | | | | | | | | - Monica Monici
- ASAcampus Joint Laboratory, ASA Res. Div., Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, Firenze, Italy
| | - Sergio Capaccioli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, viale Morgagni 50, 50134, Firenze, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Università Degli Studi Di Firenze, viale Morgagni 50, 50134, Firenze, Italy.
| |
Collapse
|
16
|
Yu H, Li Y, Tang J, Lu X, Hu W, Cheng L. Long non-coding RNA RP11-84C13.1 promotes osteogenic differentiation of bone mesenchymal stem cells and alleviates osteoporosis progression via the miR-23b-3p/RUNX2 axis. Exp Ther Med 2021; 22:1340. [PMID: 34630694 PMCID: PMC8495569 DOI: 10.3892/etm.2021.10775] [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: 12/26/2020] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
The objective of the present study was to determine the role of RP11-84C13.1 in osteoporosis (OP) and its molecular mechanism. First, clinical samples were collected from OP patients and normal control patients. Human bone marrow stromal cells (hBMSCs) were extracted from femoral head tissues. Runt-related transcription factor 2 (RUNX2) and RP11-84C13.1 serum levels were assessed by reverse transcription-quantitative (RT-q)PCR. Following transfection of pcDNA-RP11-84C13.1, si-RP11-84C13.1, microRNA (miRNA)-23b-3p mimic and miRNA-23b-3p inhibitor, the expression levels of RUNX2 and RP11-84C13.1 were determined by RT-qPCR. In addition, the osteogenic ability of hBMSCs was assessed by Alizarin Red staining. The binding of RP11-84C13.1 to miRNA-23b-3p and the binding of miRNA-23b-3p to RUNX2 was confirmed by dual-luciferase reporter gene assay. Long non-coding RNA (lncRNA) RP11-84C13.1 was significantly downregulated in the serum of OP patients. The osteogenic differentiation-related genes RUNX2 and RP11-84C13.1 were markedly upregulated in a time-dependent manner, while the miRNA-23b-3p level gradually decreased in hBMSCs with the prolongation of osteogenesis. RP11-84C13.1 knockdown inhibited the osteogenic differentiation of hBMSCs. Furthermore, RP11-84C13.1 regulated RUNX2 expression by targeting miRNA-23b-3p. Overexpression of miRNA-23b-3p partially reversed the promoting effect of RP11-84C13.1 on the osteogenesis of hBMSCs. In conclusion, lncRNA RP11-84C13.1 upregulated RUNX2 by absorbing miRNA-23b-3p, and thus induced hBMSC osteogenesis to alleviate osteoporosis.
Collapse
Affiliation(s)
- Huaixi Yu
- Department of Orthopaedic Surgery, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223000, P.R. China
| | - Yunyun Li
- Department of Information Statistics Center, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223000, P.R. China
| | - Jinshan Tang
- Department of Orthopaedic Surgery, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223000, P.R. China
| | - Xiaoqing Lu
- Department of Orthopaedic Surgery, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223000, P.R. China
| | - Wen Hu
- Department of Endocrinology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223000, P.R. China
| | - Liang Cheng
- Department of Endocrinology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223000, P.R. China
| |
Collapse
|
17
|
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
|
18
|
Abstract
Osteoporosis is a common bone disease characterized by low bone mass and deterioration of bone microstructure, which predisposes to higher risks of bone fragility and bone fracture. Long non-coding RNAs (lncRNAs) are a class of RNAs with a length of > 200 nucleotides without protein-coding function, which control the expression of genes and affect multiple biological processes. Accumulating evidence suggests that lncRNAs are widely involved in the molecular mechanisms of osteoporosis. This review aims to summarize the function and underlying mechanism of lncRNAs involved in the development of osteoporosis, and how it contributes to osteoblast and osteoclast function. This knowledge will shed new light on the modulation and potential treatment of osteoporosis.
Collapse
Affiliation(s)
- Yinxi He
- Department of Orthopaedic Trauma, The Third Hospital of Shijiazhuang, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Yanxia Chen
- Department of Endocrinology, The Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, People's Republic of China.
| |
Collapse
|
19
|
Aurilia C, Donati S, Palmini G, Miglietta F, Iantomasi T, Brandi ML. The Involvement of Long Non-Coding RNAs in Bone. Int J Mol Sci 2021; 22:ijms22083909. [PMID: 33920083 PMCID: PMC8069547 DOI: 10.3390/ijms22083909] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/02/2021] [Accepted: 04/08/2021] [Indexed: 12/18/2022] Open
Abstract
A harmonious balance between osteoblast and osteoclast activity guarantees optimal bone formation and resorption, pathological conditions affecting the bone may arise. In recent years, emerging evidence has shown that epigenetic mechanisms play an important role during osteoblastogenesis and osteoclastogenesis processes, including long non-coding RNAs (lncRNAs). These molecules are a class of ncRNAs with lengths exceeding 200 nucleotides not translated into protein, that have attracted the attention of the scientific community as potential biomarkers to use for the future development of novel diagnostic and therapeutic approaches for several pathologies, including bone diseases. This review aims to provide an overview of the lncRNAs and their possible molecular mechanisms in the osteoblastogenesis and osteoclastogenesis processes. The deregulation of their expression profiles in common diseases associated with an altered bone turnover is also described. In perspective, lncRNAs could be considered potential innovative molecular biomarkers to help with earlier diagnosis of bone metabolism-related disorders and for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Cinzia Aurilia
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Simone Donati
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Francesca Miglietta
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (C.A.); (S.D.); (G.P.); (F.M.); (T.I.)
- Fondazione Italiana Ricerca sulle Malattie dell’Osso (FIRMO Onlus), 50141 Florence, Italy
- Correspondence:
| |
Collapse
|
20
|
The combined effects of simulated microgravity and X-ray radiation on MC3T3-E1 cells and rat femurs. NPJ Microgravity 2021; 7:3. [PMID: 33589605 PMCID: PMC7884416 DOI: 10.1038/s41526-021-00131-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 01/13/2021] [Indexed: 01/05/2023] Open
Abstract
Microgravity is well-known to induce Osteopenia. However, the combined effects of microgravity and radiation that commonly exist in space have not been broadly elucidated. This research investigates the combined effects on MC3T3-E1 cells and rat femurs. In MC3T3-E1 cells, simulated microgravity and X-ray radiation, alone or combination, show decreased cell activity, increased apoptosis rates by flow cytometric analysis, and decreased Runx2 and increased Caspase-3 mRNA and protein expressions. In rat femurs, simulated microgravity and X-ray radiation, alone or combination, show increased bone loss by micro-CT test and Masson staining, decreased serum BALP levels and Runx2 mRNA expressions, and increased serum CTX-1 levels and Caspase-3 mRNA expressions. The strongest effect is observed in the combined group in MC3T3-E1 cells and rat femurs. These findings suggest that the combination of microgravity and radiation exacerbates the effects of either treatment alone on MC3T3-E1 cells and rat femurs.
Collapse
|
21
|
Zhang K, Shi Z, Ren Y, Han X, Wang J, Hong W. [Kcnq1ot1 promotes osteogenic differentiation and suppresses osteoclast differentiation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:31-38. [PMID: 33509750 DOI: 10.12122/j.issn.1673-4254.2021.01.04] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the regulatory role of long non-coding RNA Kcnq1ot1 in osteoclast differentiation, osteogenic differentiation and osteoporosis. METHODS The expression of lnc-Kcnq1ot1, Bglap, Runx2, Alp, Bsp, Nfatc1, Mmp9, Ctsk and Oscar were detected by real-time quantitative PCR (qRT-PCR) in the femoral bones from mouse models of postmenopausal osteoporosis (ovariectomized mice, n=8), disuse osteoporosis (induced by tail suspension, n=14) and agerelated osteoporosis (18-month-old mice, n=8), and also in MC3T3-E1 cells during osteoblast differentiation and in murine bone marrow-derived macrophages (BMMs) and RAW264.7 cells during osteoclast differentiation. MC3T3-E1 cells with lncKcnq1ot1 knockdown by lentivirus infection were induced to differentiate into osteoblasts using osteogenic induction medium, and the expression of lnc-Kcnq1ot1, Alp and Bglap was detected with qRT-PCR and ALP activity was assessed with ALP staining. BMMs and RAW264.7 cells were transfected with siRNAs targeting lnc-Kcnq1ot1 and stimulated with RANKL and/or M-CSF, and the expression of lnc-Kcnq1ot1, Ctsk and Oscar was detected by qRT-PCR, and TRAP activity was assessed by TRAP staining. The subcellular localization of lnc-Kcnq1ot1 in MC3T3-E1 and RAW264.7 cells was determined using cell fractionation followed by qRT-PCR. RESULTS The expression of lnc-Kcnq1ot1 was significantly upregulated during osteoblast differentiation but downregulated in the bone tissues of osteoporotic mice and during osteoclast differentiation (P < 0.05). Silencing lnc-Kcnq1ot1 obviously decreased the expression of Bglap and Alp (P < 0.05) and attenuated osteogenic medium-induced osteoblast differentiation. Knockdown of lnc-Kcnq1ot1 also promoted the expression of Ctsk and Oscar (P < 0.05) and aggravated RANKL-induced osteoclast differentiation. The results of cell fractionation and qRT-PCR demonstrated that lnc-Kcnq1ot1 was located mainly in the nuclei of MC3T3-E1 and RAW264.7 cells. CONCLUSIONS Our data demonstrate that lnc-Kcnq1ot1 promotes osteogenic differentiation and alleviates osteoclast differentiation, suggesting the potential of lnc-Kcnq1ot1 as a therapeutic target against osteoporosis.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zhemin Shi
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yi Ren
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaohui Han
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jingzhao Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Wei Hong
- Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
22
|
Yin C, Tian Y, Yu Y, Li D, Miao Z, Su P, Zhao Y, Wang X, Pei J, Zhang K, Qian A. Long noncoding RNA AK039312 and AK079370 inhibits bone formation via miR-199b-5p. Pharmacol Res 2021; 163:105230. [PMID: 33031910 DOI: 10.1016/j.phrs.2020.105230] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/14/2022]
Abstract
Osteoporosis caused by aging and menopause had become an emerging threat to human health. The reduction of osteoblast differentiation has been considered to be an essential cause of osteoporosis. Osteoblast differentiation could be regulated by LncRNAs, and increasing evidences have proved that LncRNAs may be adopted as potential therapeutic targets for osteoporosis. However, reports on rescue effects of LncRNAs in vivo are relatively limited. In this study, two LncRNAs (AK039312 and AK079370) were screened as osteogenic related LncRNAs. Both AK039312 and AK079370 could inhibit osteoblast differentiation and bone formation through suppressing osteogenic transcription factors. This inhibitory effect was achieved via binding and sequestering miR-199b-5p, and enhanced GSK-3β which further inhibited wnt/β-catenin pathway. Moreover, the siRNAs of AK039312 and AK079370 significantly alleviated postmenopausal osteoporosis, and the combination of si-AK039312 and si-AK079370 was more efficient than applying one si-LncRNA alone. This study has provided new insights for the therapy of osteoporosis.
Collapse
Affiliation(s)
- Chong Yin
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Ye Tian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Yang Yu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dijie Li
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Zhiping Miao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Peihong Su
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Yipu Zhao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Xue Wang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Jiawei Pei
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Kewen Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, China.
| |
Collapse
|
23
|
Patil S, Dang K, Zhao X, Gao Y, Qian A. Role of LncRNAs and CircRNAs in Bone Metabolism and Osteoporosis. Front Genet 2020; 11:584118. [PMID: 33281877 PMCID: PMC7691603 DOI: 10.3389/fgene.2020.584118] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Bone is a mechanosensitive organ that provides strength and support. Many bone cells, various pathways, and signaling molecules coordinate bone metabolism and also determine the course of bone diseases, such as osteoporosis, osteonecrosis, osteopenia, etc. Osteoporosis is caused by increased bone resorption and reduced bone formation due to the changes in the level of different proteins and RNAs in osteoclast or/and osteoblasts. The available therapeutic interventions can significantly reduce bone resorption or enhance bone formation, but their prolonged use has deleterious side effects. Therefore, the use of non-coding RNAs as therapeutics has emerged as an interesting field of research. Despite advancements in the molecular field, not much is known about the role of long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) in bone homeostasis and osteoporosis. Therefore, in this article, we summarize the role of lncRNAs and circRNAs in different bone cells and osteoporosis so that it might help in the development of osteoporotic therapeutics.
Collapse
Affiliation(s)
- Suryaji Patil
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Kai Dang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xin Zhao
- School of Pharmacy, Shaanxi Institute of International Trade & Commerce, Xi'an, China
| | - Yongguang Gao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Department of Chemistry, Tangshan Normal University, Tangshan, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
24
|
Yin C, Tian Y, Yu Y, Yang C, Su P, Zhao Y, Wang X, Zhang K, Pei J, Li D, Chen Z, Zhang Y, Miao Z, Qian A. miR-129-5p Inhibits Bone Formation Through TCF4. Front Cell Dev Biol 2020; 8:600641. [PMID: 33240893 PMCID: PMC7681249 DOI: 10.3389/fcell.2020.600641] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis is a frequently occurring bone disease in middle-aged and aged men and women. However, current therapies on this disease are still not ideal. MicroRNAs (miRNAs) are a class of endogenous non-protein-coding RNA with a length of 18–25 nucleotides. miRNAs have been identified as important regulators for development, metabolism, carcinogenesis, and bone formation. miR-129-5p has been reported as a regulator of cancer and neuroscience, whereas studies about its function on bone formation is still limited. In this study, we investigated the function and mechanism of miR-129-5p on osteoblast differentiation and bone formation. We have assessed the expression of miRNAs in bone mesenchymal stem cells from aging and menopause osteoporosis C57BL6 mice. The expression of miR-129-5p was altered in all osteoporosis models. Besides, the expression of miR-129-5p was negatively correlated with osteoblastic differentiation markers in the femur tissues of C57BL/6 mice of different ages. We further demonstrated that overexpression of miR-129-5p inhibited osteoblast differentiation in MC3T3-E1 cell line, as well as bone formation of C57BL/6 mice. On the other hand, down-regulation of miR-129-5p enhanced osteoblast differentiation and bone formation. We also found that miR-129-5p inhibited Wnt/β-catenin pathway in osteoblast. The target gene of miR-129-5p has been forecasted and proved as Tcf4. We further found that plasmid containing Tcf4–3′ UTR sequence enhanced osteoblast differentiation, as well as Wnt/β-catenin pathway in MC3T3-E1 cells. To further investigate the rescue effect of miR-129-5p inhibitor, we manufactured bioengineered novel recombinant miR-129-5p inhibitor through Escherichia coli system and then tested its function. The results showed that the novel recombinant miR-129-5p inhibitor promoted osteoblast differentiation and greatly ameliorated menopause osteoporosis in C57BL6 mice. In conclusion, we have discovered miR-129-5p as an inhibitor of bone formation. miR-129-5p inhibited downstream transcription factors of Wnt/β-catenin pathway through targeting Tcf4. Moreover, novel recombinant miR-129-5p inhibitor showed rescue effect on osteoporosis. This study has revealed a new mechanism of osteogenic differentiation and provided novel therapeutic strategies for treatment of skeletal disorders.
Collapse
Affiliation(s)
- Chong Yin
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Ye Tian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yang Yu
- Tianjin Key Laboratory on Technologies Enabling Development Clinical Therapeutics and Diagnostics (Theranostics), School of pharmacy, Tianjin Medical University, Tianjin, China
| | - Chaofei Yang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Peihong Su
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yipu Zhao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Xue Wang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Kewen Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Jiawei Pei
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Dijie Li
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Zhihao Chen
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Yan Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Zhiping Miao
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
25
|
Zhao W, Wang G, Zhou C, Zhao Q. The regulatory roles of long noncoding RNAs in osteoporosis. Am J Transl Res 2020; 12:5882-5907. [PMID: 33042467 PMCID: PMC7540091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
Osteoporosis is a common metabolic bone disease characterized by low bone mineral density (BMD) and microarchitectural deterioration of bone tissue, which leads to decreased bone strength and increased fracture risk. Osteoporosis mainly results from a disruption of the balance between bone formation mediated by osteoblasts and bone resorption mediated by osteoclasts. At present, the molecular mechanisms underlying osteoporosis are still not fully understood. Long noncoding RNAs (lncRNAs) are RNA molecules that exceed 200 nucleotides (nt) in length and have limited or no protein-coding capacity. Over the past decade, numerous lncRNAs have been demonstrated to participate in multiple biological processes and to play essential roles in the pathogenesis of various diseases. In this review, we summarize recent progress in research on lncRNAs in osteoporosis and mainly focus on their regulatory roles in osteogenesis and osteoclastogenesis. Moreover, we briefly discuss the potential clinical applications of lncRNAs in osteoporosis.
Collapse
Affiliation(s)
- Weisong Zhao
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200080, China
- First Clinical College, Xinxiang Medical UniversityXinxiang 453000, Henan, China
| | - Gangyang Wang
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200080, China
| | - Chenghao Zhou
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200080, China
| | - Qinghua Zhao
- Department of Orthopaedics, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200080, China
| |
Collapse
|