1
|
Tao R, Qiao MQ, Wang B, Fan JP, Gao F, Wang SJ, Guo SY, Xia SL. Laboratory-based Biomarkers for Risk Prediction, Auxiliary Diagnosis and Post-operative Follow-up of Osteoporotic Fractures. Curr Osteoporos Rep 2025; 23:19. [PMID: 40199776 PMCID: PMC11978538 DOI: 10.1007/s11914-025-00914-5] [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] [Accepted: 03/26/2025] [Indexed: 04/10/2025]
Abstract
PURPOSE OF REVIEW Osteoporosis (OP) is characterized by degraded bone microstructure, loss of bone mass and increased risk of fragility fractures. Currently, T-score determined by dual-energy X-ray absorptiometry (DEXA) measurements has been regarded as the gold standard for the diagnosis of osteoporosis. However, multiple factors have indicated that the T-score is insufficient to identify individuals with osteoporosis at a potentially high risk of fracture, or accurately detect those who require treatment, or continuously monitor the risk of re-fracture and clinical outcomes after treatment. This review covers publications in a range of ten years and comprehensively summarizes the studies in laboratory-based biomarkers for osteoporotic fractures (OF), aiming to provide physicians and surgeons with an update of clinical research in identification, verification and application of these tools, and to provide useful information for the design of future clinical studies. RECENT FINDINGS It was found that bone formation markers (such as PINP, BGP, ECM1 and SOST), bone resorption markers (such as β-CTX, TRAcP5b, osteocalcin, RANKL, RANKL/OPG ratio, and t-PINP/β-CTX), hormonal biomarkers (such as IGF- 1, PTH, leptin, adiponectin and AMH), indicators of inflammation and oxidative stress (SII, IL- 6, LTL, FlOP_360, FlOP_400, and GGT), microRNAs (such as miR- 21, miR- 320a- 3p, miR- 491 - 5p, miR- 485 - 3p, miR- 19b- 1- 5p, miR- 203a, miR- 31 - 5p, miR- 502 - 3p, miR- 4739, miR- 497, miR- 19b, and miR- 107), other biomarkers (SAF-AGEs and glycine), adipocytokines (irisin and Omentin- 1), senescence biomarkers (RDW), and lncRNAs (MIAT) may be useful biomarkers for clinical practice. Further validation of these biomarkers and a better understanding of the underlying molecular mechanisms may help in the development and application of these biomarkers for risk prediction of OF, differential diagnosis among OP, OF and healthy individuals, as well as post-operative monitoring of re-fracture risk and treatment outcomes.
Collapse
Affiliation(s)
- Rui Tao
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New Area, Shanghai, 201203, China
| | - Mei-Qi Qiao
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
- Graduate School, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New Area, Shanghai, 201203, China
| | - Bin Wang
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Jian-Pin Fan
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Feng Gao
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Shao-Jun Wang
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Sheng-Yang Guo
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China
| | - Sheng-Li Xia
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, 1500 Zhoupu Zhouyuan Road, Pudong New Area, Shanghai, 201318, China.
- Graduate School, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New Area, Shanghai, 201203, China.
| |
Collapse
|
2
|
Liu C, Feng N, Wang Z, Zheng K, Xie Y, Wang H, Long H, Peng S. Foxk1 promotes bone formation through inducing aerobic glycolysis. Cell Death Differ 2024; 31:1650-1663. [PMID: 39232134 PMCID: PMC11618307 DOI: 10.1038/s41418-024-01371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024] Open
Abstract
Transcription factor Foxk1 can regulate cell proliferation, differentiation, metabolism, and promote skeletal muscle regeneration and cardiogenesis. However, the roles of Foxk1 in bone formation is unknown. Here, we found that Foxk1 expression decreased in the bone tissue of aged mice and osteoporosis patients. Knockdown of Foxk1 in primary murine calvarial osteoblasts suppressed osteoblast differentiation and proliferation. Conditional knockout of Foxk1 in preosteoblasts and mature osteoblasts in mice exhibited decreased bone mass and mechanical strength due to reduced bone formation. Mechanistically, we identified Foxk1 targeted the promoter region of many genes of glycolytic enzyme by CUT&Tag analysis. Lacking of Foxk1 in primary murine calvarial osteoblasts resulted in reducing aerobic glycolysis. Inhibition of glycolysis by 2DG hindered osteoblast differentiation and proliferation induced by Foxk1 overexpression. Finally, specific overexpression of Foxk1 in preosteoblasts, driven by a preosteoblast specific osterix promoter, increased bone mass and bone mechanical strength of aged mice, which could be suppressed by inhibiting glycolysis. In summary, these findings reveal that Foxk1 plays a vital role in the osteoblast metabolism regulation and bone formation stimulation, offering a promising approach for preventing age-related bone loss.
Collapse
Affiliation(s)
- Chungeng Liu
- Division of Spine, Department of Orthopedic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China
- The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Naibo Feng
- Division of Spine, Department of Orthopedic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China
- The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Zhenmin Wang
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China
| | - Kangyan Zheng
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China
| | - Yongheng Xie
- Division of Spine, Department of Orthopedic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China
| | - Hongyu Wang
- Division of Spine, Department of Orthopedic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China
| | - Houqing Long
- Division of Spine, Department of Orthopedic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China.
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, China.
| | - Songlin Peng
- Division of Spine, Department of Orthopedic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, China.
- Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, Shenzhen, China.
| |
Collapse
|
3
|
Liu XF, Liao YT, Shao JH, He DD, Fan ZH, Xu YN, Li C, Zhang X. Angelicin improves osteoporosis in ovariectomized rats by reducing ROS production in osteoclasts through regulation of the KAT6A/Nrf2 signalling pathway. Chin Med 2024; 19:91. [PMID: 38956695 PMCID: PMC11218408 DOI: 10.1186/s13020-024-00961-7] [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: 11/25/2023] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Angelicin, which is found in Psoralea, can help prevent osteoporosis by stopping osteoclast formation, although the precise mechanism remains unclear. METHODS We evaluated the effect of angelicin on the oxidative stress level of osteoclasts using ovariectomized osteoporosis model rats and RAW264.7 cells. Changes in the bone mass of the femur were investigated using H&E staining and micro-CT. ROS content was investigated by DHE fluorescence labelling. Osteoclast-related genes and proteins were examined for expression using Western blotting, immunohistochemistry, tartrate-resistant acid phosphatase staining, and real-time quantitative PCR. The influence of angelicin on osteoclast development was also evaluated using the MTT assay, double luciferin assay, chromatin immunoprecipitation, immunoprecipitation and KAT6A siRNA transfection. RESULTS Rats treated with angelicin had considerably higher bone mineral density and fewer osteoclasts. Angelicin prevented RAW264.7 cells from differentiating into osteoclasts in vitro when stimulated by RANKL. Experiments revealed reduced ROS levels and significantly upregulated intracellular KAT6A, HO-1, and Nrf2 following angelicin treatment. The expression of genes unique to osteoclasts, such as MMP9 and NFATc1, was also downregulated. Finally, KAT6A siRNA transfection increased intracellular ROS levels while decreasing KAT6A, Nrf2, and HO-1 protein expression in osteoclasts. However, in the absence of KAT6A siRNA transfection, angelicin greatly counteracted this effect in osteoclasts. CONCLUSIONS Angelicin increased the expression of KAT6A. This enhanced KAT6A expression helps to activate the Nrf2/HO-1 antioxidant stress system and decrease ROS levels in osteoclasts, thus inhibiting oxidative stress levels and osteoclast formation.
Collapse
Affiliation(s)
- Xiao-Feng Liu
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yi-Tao Liao
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jia-Hao Shao
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Dan-Dan He
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, 214071, China
| | - Zhi-Hong Fan
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye-Nan Xu
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chao Li
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, 214071, China.
| | - Xian Zhang
- Department of Spine, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, 214071, China.
| |
Collapse
|
4
|
Shi Y, Kang Q, Zhou H, Yue X, Bi Y, Luo Q. Aberrant LETM1 elevation dysregulates mitochondrial functions and energy metabolism and promotes lung metastasis in osteosarcoma. Genes Dis 2024; 11:100988. [PMID: 38292199 PMCID: PMC10825238 DOI: 10.1016/j.gendis.2023.05.005] [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: 11/23/2022] [Revised: 04/10/2023] [Accepted: 05/25/2023] [Indexed: 02/01/2024] Open
Abstract
Osteosarcoma is a differentiation-deficient disease, and despite the unique advantages and great potential of differentiation therapy, there are only a few known differentiation inducers, and little research has been done on their targets. Cell differentiation is associated with an increase in mitochondrial content and activity. The metabolism of some tumor cells is characterized by impaired oxidative phosphorylation, as well as up-regulation of aerobic glycolysis and pentose phosphate pathways. Leucine-containing zipper and EF-hand transmembrane protein 1 (LETM1) is involved in the maintenance of mitochondrial morphology and is closely associated with tumorigenesis and progression, as well as cancer cell stemness. We found that MG63 and 143B osteosarcoma cells overexpress LETM1 and exhibit abnormalities in mitochondrial structure and function. Knockdown of LETM1 partially restored the mitochondrial structure and function, inhibited the pentose phosphate pathway, promoted oxidative phosphorylation, and led to osteogenic differentiation. It also inhibited spheroid cell formation, proliferation, migration, and invasion in an in vitro model. When LETM1 was knocked down in vivo, there was reduced tumor formation and lung metastasis. These data suggest that mitochondria are aberrant in LETM1-overexpressing osteosarcoma cells, and knockdown of LETM1 partially restores the mitochondrial structure and function, inhibits the pentose phosphate pathway, promotes oxidative phosphorylation, and increases osteogenic differentiation, thereby reducing malignant biological behavior of the cells.
Collapse
Affiliation(s)
- Yulu Shi
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Quan Kang
- Department of Pediatric Surgery, The Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hong Zhou
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Xiaohan Yue
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Qing Luo
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| |
Collapse
|
5
|
Yao J, Xin R, Zhao C, Yu C. MicroRNAs in osteoblast differentiation and fracture healing: From pathogenesis to therapeutic implication. Injury 2024; 55:111410. [PMID: 38359711 DOI: 10.1016/j.injury.2024.111410] [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: 10/18/2023] [Revised: 01/20/2024] [Accepted: 01/27/2024] [Indexed: 02/17/2024]
Abstract
The term "fracture" pertains to the occurrence of bones being either fully or partially disrupted as a result of external forces. Prolonged fracture healing can present a notable danger to the patient's general health and overall quality of life. The significance of osteoblasts in the process of new bone formation is widely recognized, and optimizing their function could be a desirable strategy. Therefore, the mending of bone fractures is intricately linked to the processes of osteogenic differentiation and mineralization. MicroRNAs (miRNAs) are RNA molecules that do not encode for proteins, but rather modulate the functioning of physiological processes by directly targeting proteins. The participation of microRNAs (miRNAs) in experimental investigations has been extensive, and their control functions have earned them the recognition as primary regulators of the human genome. Earlier studies have shown that modulating the expression of miRNAs, either by increasing or decreasing their levels, can initiate the differentiation of osteoblasts. This implies that miRNAs play a pivotal function in promoting osteogenesis, facilitating bone mineralization and formation, ultimately leading to an efficient healing of fractures. Hence, focusing on miRNAs can be considered a propitious therapeutic approach to accelerate the healing of fractures and forestall nonunion. In this manner, the information supplied by this investigation has the potential to aid in upcoming clinical utilization, including its possible use as biomarkers or as resources for devising innovative therapeutic tactics aimed at promoting fracture healing.
Collapse
Affiliation(s)
- Jilong Yao
- Department of surgery teaching and research section, Jiangxi Medical College, Shangrao, 334000, China
| | - Ruiwen Xin
- Department of surgery teaching and research section, Jiangxi Medical College, Shangrao, 334000, China
| | - Chao Zhao
- Department of Neurology, Shangrao municipal hospital, Shangrao, 334000, China
| | - Chunfu Yu
- Department of Neurology, Shangrao municipal hospital, Shangrao, 334000, China.
| |
Collapse
|
6
|
Chen G, Wang S, Wei R, Liu Y, Xu T, Liu Z, Tan Z, Xie Y, Yang D, Liang Z, Zhuang Y, Peng S. Circular RNA circ-3626 promotes bone formation by modulating the miR-338-3p/Runx2 axis. Joint Bone Spine 2024; 91:105669. [PMID: 38042362 DOI: 10.1016/j.jbspin.2023.105669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/11/2023] [Accepted: 11/09/2023] [Indexed: 12/04/2023]
Abstract
OBJECTIVE Disorders of bone homeostasis are the key factors leading to metabolic bone disease, such as senile osteoporosis, which is characterized by age-related bone loss. Bone marrow stromal cells (BMSCs) possess high osteogenic capacity which has been regarded as a practical approach to preventing bone loss. Previous studies have shown that the osteogenic differentiation ability of BMSCs is significantly decreased in senile osteoporosis. Recently, circular RNAs (circRNAs) have been regarded as critical regulators in controlling the osteogenic differentiation of BMSCs by sponging microRNAs (miRNAs). Our study aimed to discover new and critical osteogenesis-related circRNAs that can promote bone formation in senile osteoporosis. METHODS We detected the dysregulated circRNAs of BMSCs upon osteogenic differentiation induction and identified the critical osteogenic circRNA (circ-3626). The relationship between circ-3626 and osteoporosis was further verified in clinical bone samples and aged mice by qPCR. Moreover, circ-3626 AAV was constructed to examine the osteogenic effect of circ-3626 on bone formation via using Micro-CT, double calcein labeling, and the three-point bending tests. Bioinformatics analysis, Luciferase report gene assays, FISH, RNA pull-down, qPCR, Western Blots, and alizarin red staining assay explore the effects and mechanisms of circ-3626 on osteogenic differentiation of BMSCs. RESULTS Circ-3626 was identified as a pivotal osteogenesis-related circRNA via RNA sequencing. The results of alizarin red staining, Western blots, and qPCR assays suggest that overexpressing circ-3626 dramatically accelerates the osteogenic capability of BMSCs. Furthermore, the bone repair capability of aging mice could be significantly improved by circ-3626 AAV treatment. Micro RNA miR-338-3p was identified as the downstream target of circ-3626. Overexpression of circ-3626 increases the expression of Runx2 by sponging miR-338-3p, thereby promoting the osteogenic differentiation of BMSCs by upregulating the expression of osteogenic genes. In addition, Western blots, and qPCR assays suggest circ-3626 AAV treatment promote the expression of Runx2 and osteogenic marker genes. CONCLUSION Thus, we demonstrate that circ-3626 plays a pivotal role in promoting bone formation through the miR-338-3p/Runx2 axis and may provide new strategies for preventing and treating the bone loss of senile osteoporosis.
Collapse
Affiliation(s)
- Gaoyang Chen
- Division of Hand, Foot and Microvascular Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China.
| | - Song Wang
- Division of Spine Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Ruihong Wei
- Division of Hand, Foot and Microvascular Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Yingnan Liu
- Division of Hand, Foot and Microvascular Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Tao Xu
- Division of Hand, Foot and Microvascular Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Zhaokang Liu
- Division of Hand, Foot and Microvascular Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Zhouyong Tan
- Division of Hand, Foot and Microvascular Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Yongheng Xie
- Division of Spine Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Dazhi Yang
- Division of Spine Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China
| | - Zhen Liang
- Department of Geriatrics, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China
| | - Yongqing Zhuang
- Division of Hand, Foot and Microvascular Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China.
| | - Songlin Peng
- Division of Spine Surgery, Department of Orthopedic Surgery, Shenzhen People's Hospital (the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology), 518020 Shenzhen, China; Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration (ZDSYS20200811143752005), Shenzhen Institute for Orthopedic Research, 518020 Shenzhen, China.
| |
Collapse
|
7
|
Raimondi L, De Luca A, Gallo A, Perna F, Cuscino N, Cordaro A, Costa V, Bellavia D, Faldini C, Scilabra SD, Giavaresi G, Toscano A. Investigating the Differential Circulating microRNA Expression in Adolescent Females with Severe Idiopathic Scoliosis: A Proof-of-Concept Observational Clinical Study. Int J Mol Sci 2024; 25:570. [PMID: 38203740 PMCID: PMC10779108 DOI: 10.3390/ijms25010570] [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: 11/13/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Adolescent Idiopathic Scoliosis (AIS) is the most common form of three-dimensional spinal disorder in adolescents between the ages of 10 and 18 years of age, most commonly diagnosed in young women when severe disease occurs. Patients with AIS are characterized by abnormal skeletal growth and reduced bone mineral density. The etiology of AIS is thought to be multifactorial, involving both environmental and genetic factors, but to date, it is still unknown. Therefore, it is crucial to further investigate the molecular pathogenesis of AIS and to identify biomarkers useful for predicting curve progression. In this perspective, the relative abundance of a panel of microRNAs (miRNAs) was analyzed in the plasma of 20 AIS patients and 10 healthy controls (HC). The data revealed a significant group of circulating miRNAs dysregulated in AIS patients compared to HC. Further bioinformatic analyses evidenced a more restricted expression of some miRNAs exclusively in severe AIS females. These include some members of the miR-30 family, which are considered promising regulators for treating bone diseases. We demonstrated circulating extracellular vesicles (EVs) from severe AIS females contained miR-30 family members and decreased the osteogenic differentiation of mesenchymal stem cells. Proteomic analysis of EVs highlighted the expression of proteins associated with orthopedic disease. This study provides preliminary evidence of a miRNAs signature potentially associated with severe female AIS and suggests the corresponding vesicular component may affect cellular mechanisms crucial in AIS, opening the scenario for in-depth studies on prognostic differences related to gender and grade.
Collapse
Affiliation(s)
- Lavinia Raimondi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Angela De Luca
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Alessia Gallo
- Dipartimento di Ricerca, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Fabrizio Perna
- Ortopedia Generale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy (A.T.)
| | - Nicola Cuscino
- Dipartimento di Ricerca, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Aurora Cordaro
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Viviana Costa
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Daniele Bellavia
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Cesare Faldini
- Clinica Ortopedica e Traumatologica I, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Simone Dario Scilabra
- Fondazione Ri.MED, Dipartimento di Ricerca IRCCS ISMETT, Via Ernesto Tricomi 5, 90145 Palermo, Italy
| | - Gianluca Giavaresi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Angelo Toscano
- Ortopedia Generale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy (A.T.)
| |
Collapse
|
8
|
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
|
9
|
Sudhakaran G, Priya PS, Jagan K, Haridevamuthu B, Meenatchi R, Arockiaraj J. Osteoporosis in polycystic ovary syndrome (PCOS) and involved mechanisms. Life Sci 2023; 335:122280. [PMID: 37981226 DOI: 10.1016/j.lfs.2023.122280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
Polycystic Ovary Syndrome (PCOS) and osteoporosis, though seemingly unrelated, exhibit intricate connections influenced by genetic and epigenetic factors. PCOS, characterized by elevated androgen levels, insulin resistance, and increased body weight, has historically been considered protective against bone fragility disorders. However, emerging research suggests that chronic inflammation, prevalent in PCOS, can adversely affect bone health. Studies have demonstrated variable bone mineral density loss in PCOS, often associated with leptin resistance and hyperinsulinemia. Key genes such as INS, IGF1, CTNNB1, AKT1, and STAT3 play pivotal roles in the complex interplay between PCOS and osteoporosis, influencing insulin signaling, oxidative stress, and inflammatory pathways. Oxidative stress, a prominent element in PCOS, can lead to osteoporosis through hormonal imbalances, chronic inflammation, insulin resistance, and lifestyle factors. The insulin signaling pathway also significantly impacts both conditions by contributing to hormonal imbalances and bone health alterations. This intricate network of genetic and epigenetic factors underscores the need for a deeper understanding of their interrelationships. Thus, this review elucidates the multifaceted genetic, epigenetic, and inflammatory connections between PCOS and osteoporosis, highlighting their implications for bone health management in individuals with PCOS.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - P Snega Priya
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Kannan Jagan
- Department of Biotechnology, SRM Arts and Science College, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - B Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Ramu Meenatchi
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
10
|
Mercier-Guery A, Millet M, Merle B, Collet C, Bagouet F, Borel O, Sornay-Rendu E, Szulc P, Vignot E, Gensburger D, Fontanges E, Croset M, Chapurlat R. Dysregulation of MicroRNAs in Adult Osteogenesis Imperfecta: The miROI Study. J Bone Miner Res 2023; 38:1665-1678. [PMID: 37715362 DOI: 10.1002/jbmr.4912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 08/23/2023] [Accepted: 09/09/2023] [Indexed: 09/17/2023]
Abstract
As epigenetic regulators of gene expression, circulating micro-RiboNucleic Acids (miRNAs) have been described in several bone diseases as potential prognostic markers. The aim of our study was to identify circulating miRNAs potentially associated with the severity of osteogenesis imperfecta (OI) in three steps. We have screened by RNA sequencing for the miRNAs that were differentially expressed in sera of a small group of OI patients versus controls and then conducted a validation phase by RT-qPCR analysis of sera of a larger patient population. In the first phase of miROI, we found 79 miRNAs that were significantly differentially expressed. We therefore selected 19 of them as the most relevant. In the second phase, we were able to validate the significant overexpression of 8 miRNAs in the larger OI group. Finally, we looked for a relationship between the level of variation of the validated miRNAs and the clinical characteristics of OI. We found a significant difference in the expression of two microRNAs in those patients with dentinogenesis imperfecta. After reviewing the literature, we found 6 of the 8 miRNAs already known to have a direct action on bone homeostasis. Furthermore, the use of a miRNA-gene interaction prediction model revealed a 100% probability of interaction between 2 of the 8 confirmed miRNAs and COL1A1 and/or COL1A2. This is the first study to establish the miRNA signature in OI, showing a significant modification of miRNA expression potentially involved in the regulation of genes involved in the physiopathology of OI. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Alexandre Mercier-Guery
- Hospices Civils de Lyon, Hôpital E. Herriot, Service de Rhumatologie et Pathologie Osseuse, Lyon, France
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| | - Marjorie Millet
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| | - Blandine Merle
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| | - Corinne Collet
- CHU Robert Debré, Université de Paris Cité, Département de Génétique, CHU Lariboisière, Paris, France
- INSERM UMR1132, CHU Lariboisière, Paris, France
| | - Flora Bagouet
- Hospices Civils de Lyon, Hôpital E. Herriot, Service de Rhumatologie et Pathologie Osseuse, Lyon, France
| | - Olivier Borel
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| | - Elisabeth Sornay-Rendu
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| | - Pawel Szulc
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| | - Emmanuelle Vignot
- Hospices Civils de Lyon, Hôpital E. Herriot, Service de Rhumatologie et Pathologie Osseuse, Lyon, France
| | - Deborah Gensburger
- Hospices Civils de Lyon, Hôpital E. Herriot, Service de Rhumatologie et Pathologie Osseuse, Lyon, France
| | - Elisabeth Fontanges
- Hospices Civils de Lyon, Hôpital E. Herriot, Service de Rhumatologie et Pathologie Osseuse, Lyon, France
| | - Martine Croset
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| | - Roland Chapurlat
- Hospices Civils de Lyon, Hôpital E. Herriot, Service de Rhumatologie et Pathologie Osseuse, Lyon, France
- Université de Lyon, Université Lyon 1, INSERM UMR 1033; LYOS Pathophysiology, Diagnosis & Treatments of Musculoskeletal Disorders, Lyon, France
| |
Collapse
|
11
|
Philippe S, Delay M, Macian N, Morel V, Pickering ME. Common miRNAs of Osteoporosis and Fibromyalgia: A Review. Int J Mol Sci 2023; 24:13513. [PMID: 37686318 PMCID: PMC10488272 DOI: 10.3390/ijms241713513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
A significant clinical association between osteoporosis (OP) and fibromyalgia (FM) has been shown in the literature. Given the need for specific biomarkers to improve OP and FM management, common miRNAs might provide promising tracks for future prevention and treatment. The aim of this review is to identify miRNAs described in OP and FM, and dysregulated in the same direction in both pathologies. The PubMed database was searched until June 2023, with a clear mention of OP, FM, and miRNA expression. Clinical trials, case-control, and cross-sectional studies were included. Gray literature was not searched. Out of the 184 miRNAs found in our research, 23 are shared by OP and FM: 7 common miRNAs are dysregulated in the same direction for both pathologies (3 up-, 4 downregulated). The majority of these common miRNAs are involved in the Wnt pathway and the cholinergic system and a possible link has been highlighted. Further studies are needed to explore this relationship. Moreover, the harmonization of technical methods is necessary to confirm miRNAs shared between OP and FM.
Collapse
Affiliation(s)
- Soline Philippe
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
| | - Marine Delay
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
- Inserm 1107, Neuro-Dol, University Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Nicolas Macian
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
| | - Véronique Morel
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
| | - Marie-Eva Pickering
- Rheumatology Department, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| |
Collapse
|
12
|
Huber J, Longaker MT, Quarto N. Circulating and extracellular vesicle-derived microRNAs as biomarkers in bone-related diseases. Front Endocrinol (Lausanne) 2023; 14:1168898. [PMID: 37293498 PMCID: PMC10244776 DOI: 10.3389/fendo.2023.1168898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/31/2023] [Indexed: 06/10/2023] Open
Abstract
MicroRNAs (miRNA) are small non-coding RNA molecules that regulate posttranscriptional gene expression by repressing messengerRNA-targets. MiRNAs are abundant in many cell types and are secreted into extracellular fluids, protected from degradation by packaging in extracellular vesicles. These circulating miRNAs are easily accessible, disease-specific and sensitive to small changes, which makes them ideal biomarkers for diagnostic, prognostic, predictive or monitoring purposes. Specific miRNA signatures can be reflective of disease status and development or indicators of poor treatment response. This is especially important in malignant diseases, as the ease of accessibility of circulating miRNAs circumvents the need for invasive tissue biopsy. In osteogenesis, miRNAs can act either osteo-enhancing or osteo-repressing by targeting key transcription factors and signaling pathways. This review highlights the role of circulating and extracellular vesicle-derived miRNAs as biomarkers in bone-related diseases, with a specific focus on osteoporosis and osteosarcoma. To this end, a comprehensive literature search has been performed. The first part of the review discusses the history and biology of miRNAs, followed by a description of different types of biomarkers and an update of the current knowledge of miRNAs as biomarkers in bone related diseases. Finally, limitations of miRNAs biomarker research and future perspectives will be presented.
Collapse
Affiliation(s)
- Julika Huber
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Plastic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
13
|
Zheng ZZ, Xu JH, Dai Y, Jiang B, Tu ZM, Li L, Li Y, Wang B. Circulating miR-107 as a diagnostic biomarker of osteoporotic vertebral compression fracture increases bone formation in vitro and in vivo. Life Sci 2023; 323:121693. [PMID: 37080350 DOI: 10.1016/j.lfs.2023.121693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/31/2023] [Accepted: 04/08/2023] [Indexed: 04/22/2023]
Abstract
AIMS This study aimed to examine the key circulating miRNAs in the plasma of patients with osteoporotic vertebral compression fracture and assess their potential role as diagnostic biomarkers and explore their function in vitro and in vivo. METHODS Weighted gene co-expression network analysis (WGCNA) was applied to identify hub miRNAs for subsequent analysis. The candidate miRNAs were tested using plasma from 144 patients and the results were applied to construct receiver operating characteristic (ROC) curves to assess their diagnostic value. In addition, the function of the target microRNA was validated in MC3T3-E1 cells, human bone marrow-derived mesenchymal stromal cells (BMSCs), and an ovariectomized (OVX) mouse model. KEY FINDINGS Seven modules were obtained by WGCNA analysis. The expression levels of circulating miR-107 in the red module were significantly lower in osteoporotic patients than in healthy controls. In addition, miR-107 provided discrimination with an AUC > 85 % by ROC analyses to differentiate women osteoporosis patients from healthy controls and differentiate women osteoporotic patients with vertebral compression fractures from osteoporotic patients without vertebral compression fractures. In vitro experiments revealed that miR-107 levels were increased in osteogenically induced MC3T3-E1 cells and BMSCs and transfection with synthetic miR-107 could promote bone formation. Lastly, the bone parameters were improved by miR-107 upregulation in OVX mice. SIGNIFICANCE Our findings show that circulating miR-107 plays an essential role in facilitating osteogenesis and may be a useful diagnostic biomarker and therapeutic target in osteoporosis.
Collapse
Affiliation(s)
- Zhen-Zhong Zheng
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin-Hong Xu
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Yuliang Dai
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Bing Jiang
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Ming Tu
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Lei Li
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Yawei Li
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Bing Wang
- Department of Spine Surgery, the Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
14
|
Sharing Circulating Micro-RNAs between Osteoporosis and Sarcopenia: A Systematic Review. Life (Basel) 2023; 13:life13030602. [PMID: 36983758 PMCID: PMC10051676 DOI: 10.3390/life13030602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Background: Osteosarcopenia, a combination of osteopenia/osteoporosis and sarcopenia, is a common condition among older adults. While numerous studies and meta-analyses have been conducted on osteoporosis biomarkers, biomarker utility in osteosarcopenia still lacks evidence. Here, we carried out a systematic review to explore and analyze the potential clinical of circulating microRNAs (miRs) shared between osteoporosis/osteopenia and sarcopenia. Methods: We performed a systematic review on PubMed, Scopus, and Embase for differentially expressed miRs (p-value < 0.05) in (i) osteoporosis and (ii) sarcopenia. Following screening for title and abstract and deduplication, 83 studies on osteoporosis and 11 on sarcopenia were identified for full-text screening. Full-text screening identified 54 studies on osteoporosis, 4 on sarcopenia, and 1 on both osteoporosis and sarcopenia. Results: A total of 69 miRs were identified for osteoporosis and 14 for sarcopenia. There were 9 shared miRs, with evidence of dysregulation (up- or down-regulation), in both osteoporosis and sarcopenia: miR-23a-3p, miR-29a, miR-93, miR-133a and b, miR-155, miR-206, miR-208, miR-222, and miR-328, with functions and targets implicated in the pathogenesis of osteosarcopenia. However, there was little agreement in the results across studies and insufficient data for miRs in sarcopenia, and only three miRs, miR-155, miR-206, and miR-328, showed the same direction of dysregulation (down-regulation) in both osteoporosis and sarcopenia. Additionally, for most identified miRs there has been no replication by more than one study, and this is particularly true for all miRs analyzed in sarcopenia. The study quality was typically rated intermediate/high risk of bias. The large heterogeneity of the studies made it impossible to perform a meta-analysis. Conclusions: The findings of this review are particularly novel, as miRs have not yet been explored in the context of osteosarcopenia. The dysregulation of miRs identified in this review may provide important clues to better understand the pathogenesis of osteosarcopenia, while also laying the foundations for further studies to lead to effective screening, monitoring, or treatment strategies.
Collapse
|
15
|
Lombardi G, Delvin E. Micro-RNA: A Future Approach to Personalized Diagnosis of Bone Diseases. Calcif Tissue Int 2023; 112:271-287. [PMID: 35182198 DOI: 10.1007/s00223-022-00959-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/07/2022] [Indexed: 01/25/2023]
Abstract
Osteoporosis is a highly prevalent bone disease worldwide and the most studied bone-associated pathological condition. Although its diagnosis makes use of advanced and clinically relevant imaging and biochemical tools, the information suffers from several limitations and has little or no prognostic value. In this context, circulating micro-RNAs represent a potentially attractive alternative or a useful addition to the diagnostic arsenal and offer a greater prognostic potential than the conventional approaches. These short non-coding RNA molecules act as inhibitors of gene expression by targeting messenger RNAs with different degrees of complementarity, establishing a complex multilevel network, the basis for the fine modulation of gene expression that finally regulates every single activity of a cell. Micro-RNAs may passively and/or actively be released in the circulation by source cells, and being measurable in biological fluids, their concentrations may be associated to specific pathophysiological conditions. Mounting, despite debatable, evidence supports the use of micro-RNAs as markers of bone cell metabolic activity and bone diseases. Indeed, several micro-RNAs have been associated with bone mineral density, fractures and osteoporosis. However, concerns such as absence of comparability between studies and, the lack of standardization and harmonization of the methods, limit their application. In this review, we describe the pathophysiological bases of the association between micro-RNAs and the deregulation of bone cells activity and the processes that led to the identification of potential micro-RNA-based markers associated with metabolic bone diseases.
Collapse
Affiliation(s)
- Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi 4, 20161, Milano, Italy.
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Królowej Jadwigi 27/39, 61-871, Poznań, Poland.
| | - Edgard Delvin
- Ste-Justine University Hospital Research Centre & Department of Biochemistry, Université de Montreal, Montreal, QC, H3T 1C5, Canada
| |
Collapse
|
16
|
Wang J, Cui Y, Liu H, Li S, Sun S, Xu H, Peng C, Wang Y, Wu D. MicroRNA-loaded biomaterials for osteogenesis. Front Bioeng Biotechnol 2022; 10:952670. [PMID: 36199361 PMCID: PMC9527286 DOI: 10.3389/fbioe.2022.952670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
The large incidence of bone defects in clinical practice increases not only the demand for advanced bone transplantation techniques but also the development of bone substitute materials. A variety of emerging bone tissue engineering materials with osteogenic induction ability are promising strategies for the design of bone substitutes. MicroRNAs (miRNAs) are a class of non-coding RNAs that regulate intracellular protein expression by targeting the non-coding region of mRNA3′-UTR to play an important role in osteogenic differentiation. Several miRNA preparations have been used to promote the osteogenic differentiation of stem cells. Therefore, multiple functional bone tissue engineering materials using miRNA as an osteogenic factor have been developed and confirmed to have critical efficacy in promoting bone repair. In this review, osteogenic intracellular signaling pathways mediated by miRNAs are introduced in detail to provide a clear understanding for future clinical treatment. We summarized the biomaterials loaded with exogenous cells engineered by miRNAs and biomaterials directly carrying miRNAs acting on endogenous stem cells and discussed their advantages and disadvantages, providing a feasible method for promoting bone regeneration. Finally, we summarized the current research deficiencies and future research directions of the miRNA-functionalized scaffold. This review provides a summary of a variety of advanced miRNA delivery system design strategies that enhance bone regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dankai Wu
- *Correspondence: Yanbing Wang, ; Dankai Wu,
| |
Collapse
|
17
|
Li T, Zhang S, Yang Y, Zhang L, Yuan Y, Zou J. Co-regulation of circadian clock genes and microRNAs in bone metabolism. J Zhejiang Univ Sci B 2022; 23:529-546. [PMID: 35794684 DOI: 10.1631/jzus.b2100958] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mammalian bone is constantly metabolized from the embryonic stage, and the maintenance of bone health depends on the dynamic balance between bone resorption and bone formation, mediated by osteoclasts and osteoblasts. It is widely recognized that circadian clock genes can regulate bone metabolism. In recent years, the regulation of bone metabolism by non-coding RNAs has become a hotspot of research. MicroRNAs can participate in bone catabolism and anabolism by targeting key factors related to bone metabolism, including circadian clock genes. However, research in this field has been conducted only in recent years and the mechanisms involved are not yet well established. Recent studies have focused on how to target circadian clock genes to treat some diseases, such as autoimmune diseases, but few have focused on the co-regulation of circadian clock genes and microRNAs in bone metabolic diseases. Therefore, in this paper we review the progress of research on the co-regulation of bone metabolism by circadian clock genes and microRNAs, aiming to provide new ideas for the prevention and treatment of bone metabolic diseases such as osteoporosis.
Collapse
Affiliation(s)
- Tingting Li
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China.,School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Shihua Zhang
- College of Graduate Education, Jinan Sport University, Jinan 250102, China
| | - Yuxuan Yang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Lingli Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China. ,
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
18
|
Ciuffi S, Marini F, Fossi C, Donati S, Giusti F, Botta A, Masi L, Isaia G, Marcocci C, Migliaccio S, Minisola S, Nuti R, Tarantino U, Iantomasi T, Brandi ML. Circulating MicroRNAs as Biomarkers of Osteoporosis and Fragility Fractures. J Clin Endocrinol Metab 2022; 107:2267-2285. [PMID: 35532548 DOI: 10.1210/clinem/dgac293] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Measurement of circulating microRNAs (miRNAs) as potential biomarkers of fragility fracture risk has recently become a subject of investigation. OBJECTIVE Measure by next-generation sequencing (NGS), global miRNA expression in serum samples of osteoporotic subjects vs individuals with normal bone mineral density (BMD). DESIGN Samples were collected from patients with different bone phenotypes and/or fragility fractures who did not receive any antiresorptive and/or bone-forming drug at the time of blood collection. SETTING Samples and data were collected at 7 medical centers in Italy. PATIENTS NGS prescreening: 50 osteoporotic patients vs 30 individuals with normal BMD. Droplet digital polymerase chain reaction (ddPCR) validation: 213 patients with different bone phenotypes, including the NGS-analyzed cohort. RESULTS NGS identified 5 miRNAs (miR-8085, miR-320a-3p, miR-23a-3p, miR-4497, miR-145-5p) differentially expressed in osteoporosis cases without fractures vs controls. ddPCR validation confirmed lower c-miR-23a-3p expression in osteoporotic patients, with or without fracture, than in osteopenic and normal subjects and increased c-miR-320a-3p expression in osteoporotic patients with fracture and lower expression in osteoporotic patients without fracture. ddPCR analysis showed a significantly increased expression of miR-21-5p in osteoporotic patients, with or without fracture, than in osteopenic and normal subjects, not evidenced by the NGS prescreening. DISCUSSION Our study confirmed levels of c-miR-23a-3p and c-miR-21-5p as able to distinguish osteoporotic patients and subjects with normal BMD. Increased levels of c-miR-320a-3p specifically associated with fractures, independently by BMD, suggesting c-miR-320a-3p as a prognostic indicator of fracture risk in osteoporotic patients, to be confirmed in prospective studies on incident fractures.
Collapse
Affiliation(s)
- Simone Ciuffi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Study of Florence, Florence, Italy
| | - Francesca Marini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Study of Florence, Florence, Italy
- FirmoLab, F.I.R.M.O. Italian Foundation for the Research on Bone Diseases, Florence, Italy
| | - Caterina Fossi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Study of Florence, Florence, Italy
| | - Simone Donati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Study of Florence, Florence, Italy
| | - Francesca Giusti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Study of Florence, Florence, Italy
| | - Annalisa Botta
- Department of Biomedicine and Prevention, Medical Genetics Section, University of Rome "Tor Vergata," Rome, Italy
| | - Laura Masi
- AOU Careggi, SOD Malattie del Metabolismo Minerale ed Osseo, Florence, Italy
| | - Giancarlo Isaia
- Department of Medical Science, Gerontology Section, University of Turin, Turin, Italy
| | - Claudio Marcocci
- Department of Clinical and Experimental Medicine, Endocrinology Unit II, University of Pisa and University Hospital of Pisa, Pisa, Italy
| | - Silvia Migliaccio
- Department of Movement, Human and Health Sciences, University of "Foro Italico" of Rome, Rome, Italy
| | - Salvatore Minisola
- Dipartimento di Scienze Cliniche, Internistiche, anestesiologiche e cardiovascolari: "Sapienza," Università di Roma, Rome, Italy
| | - Ranuccio Nuti
- Department of Medicine, Surgery and Neuroscience, University of Siena, Policlinico Le Scotte, Siena, Italy
| | - Umberto Tarantino
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata" Rome, Italy
| | - Teresa Iantomasi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Study of Florence, Florence, Italy
| | - Maria Luisa Brandi
- FirmoLab, F.I.R.M.O. Italian Foundation for the Research on Bone Diseases, Florence, Italy
| |
Collapse
|
19
|
Heilmeier U, Hackl M, Schroeder F, Torabi S, Kapoor P, Vierlinger K, Eiriksdottir G, Gudmundsson EF, Harris TB, Gudnason V, Link TM, Grillari J, Schwartz AV. Circulating serum microRNAs including senescent miR-31-5p are associated with incident fragility fractures in older postmenopausal women with type 2 diabetes mellitus. Bone 2022; 158:116308. [PMID: 35066213 DOI: 10.1016/j.bone.2021.116308] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
Fragility fractures are an important hallmark of aging and an increasingly recognized complication of Type 2 diabetes (T2D). T2D individuals have been found to exhibit an increased fracture risk despite elevated bone mineral density (BMD) by dual x-ray absorptiometry (DXA). However, BMD and FRAX-scores tend to underestimate fracture risk in T2D. New, reliable biomarkers are therefore needed. MicroRNAs (miRNAs) are secreted into the circulation from cells of various tissues proportional to local disease severity. Serum miRNA-classifiers were recently found to discriminate T2D women with and without prevalent fragility fractures with high specificity and sensitivity (AUC > 0.90). However, the association of circulating miRNAs with incident fractures in T2D has not been examined yet. In 168 T2D postmenopausal women in the AGES-Reykjavik cohort, miRNAs were extracted from baseline serum and a panel of 10 circulating miRNAs known to be involved in diabetic bone disease and aging was quantified by qPCR and Ct-values extracted. Unadjusted and adjusted Cox proportional hazard models assessed the associations between serum miRNAs and incident fragility fracture. Additionally, Receiver operating curve (ROC) analyses were performed. Of the included 168 T2D postmenopausal women who were on average 77.2 ± 5.6 years old, 70 experienced at least one incident fragility fracture during the mean follow-up of 5.8 ± 2.7 years. We found that 3 serum miRNAs were significantly associated with incident diabetic fragility fracture: while low expression of miR-19b-1-5p was associated with significantly lower risk of incident fragility fracture (HR 0.84 (95% CI: 0.71-0.99, p = 0.0323)), low expression of miR-203a and miR-31-5p was each significantly associated with a higher risk of incident fragility fracture per unit increase in Ct-value (miR-203a: HR 1.29 (95% CI: 1.12-1.49), p = 0.0004, miR-31-5p HR 1.27 (95% CI: 1.06-1.52), p = 0.009). Hazard ratios of the latter two miRNAs remained significant after adjustments for age, body mass index (BMI), areal bone mineral density (aBMD), clinical FRAX or FRAXaBMD. Women with miR-203a and miR-31-5p serum levels in the lowest expression quartiles exhibited a 2.4-3.4-fold larger fracture risk than women with miR-31-5p and miR-203a serum expressions in the highest expression quartile (0.002 ≤ p ≤ 0.039). Women with both miR-203a and miR-31-5p serum levels below the median had a significantly increased fracture risk (Unadjusted HR 3.26 (95% CI: 1.57-6.78, p = 0.001) compared to those with both expression levels above the median, stable to adjustments. We next built a diabetic fragility signature consisting of the 3 miRNAs that showed the largest associations with incident fracture (miR-203a, miR-31-5p, miR-19b-1-5p). This 3-miRNA signature showed with an AUC of 0.722 comparable diagnostic accuracy in identifying incident fractures to any of the clinical parameters such as aBMD, Clinical FRAX or FRAXaBMD alone. When the 3 miRNAs were combined with aBMD, this combined 4-feature signature performed with an AUC of 0.756 (95% CI: 0.680, 0.823) significantly better than aBMD alone (AUC 0.666, 95% CI: 0.585, 0.741) (p = 0.009). Our data indicate that specific serum microRNAs including senescent miR-31-5p are associated with incident fragility fracture in older diabetic women and can significantly improve fracture risk prediction in diabetics when combined with aBMD measurements of the femoral neck.
Collapse
Affiliation(s)
- Ursula Heilmeier
- Musculoskeletal Quantitative Imaging Research Group, University of California San Francisco, San Francisco, CA, USA; Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | | | - Fabian Schroeder
- Department of Molecular Diagnostics, Austrian Institute of Technology, AIT, Vienna, Austria
| | - Soheyla Torabi
- Musculoskeletal Quantitative Imaging Research Group, University of California San Francisco, San Francisco, CA, USA
| | - Puneet Kapoor
- Musculoskeletal Quantitative Imaging Research Group, University of California San Francisco, San Francisco, CA, USA
| | - Klemens Vierlinger
- Department of Molecular Diagnostics, Austrian Institute of Technology, AIT, Vienna, Austria
| | | | | | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Bethesda, USA
| | - Vilmundur Gudnason
- The Icelandic Heart Association, Kopavogur, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Thomas M Link
- Musculoskeletal Quantitative Imaging Research Group, University of California San Francisco, San Francisco, CA, USA
| | - Johannes Grillari
- Department of Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Christian Doppler Laboratory of Biotechnology of Skin Aging, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Ann V Schwartz
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
20
|
Chen G, Long C, Wang S, Wang Z, Chen X, Tang W, He X, Bao Z, Tan B, Zhao J, Xie Y, Li Z, Yang D, Xiao G, Peng S. Circular RNA circStag1 promotes bone regeneration by interacting with HuR. Bone Res 2022; 10:32. [PMID: 35361779 PMCID: PMC8971384 DOI: 10.1038/s41413-022-00208-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 01/29/2022] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
Postmenopausal osteoporosis is a common bone metabolic disorder characterized by deterioration of the bone microarchitecture, leading to an increased risk of fractures. Recently, circular RNAs (circRNAs) have been demonstrated to play pivotal roles in regulating bone metabolism. However, the underlying functions of circRNAs in bone metabolism in postmenopausal osteoporosis remain obscure. Here, we report that circStag1 is a critical osteoporosis-related circRNA that shows significantly downregulated expression in osteoporotic bone marrow mesenchymal stem cells (BMSCs) and clinical bone tissue samples from patients with osteoporosis. Overexpression of circStag1 significantly promoted the osteogenic capability of BMSCs. Mechanistically, we found that circStag1 interacts with human antigen R (HuR), an RNA-binding protein, and promotes the translocation of HuR into the cytoplasm. A high cytoplasmic level of HuR led to the activation of the Wnt signaling pathway by stabilizing and enhancing low-density lipoprotein receptor-related protein 5/6 (Lrp5/6) and β-catenin expression, thereby stimulating the osteogenic differentiation of BMSCs. Furthermore, overexpression of circStag1 in vivo by circStag1-loaded adeno-associated virus (circStag1-AAV) promoted new bone formation, thereby preventing bone loss in ovariectomized rats. Collectively, we show that circStag1 plays a pivotal role in promoting the regeneration of bone tissue via HuR/Wnt signaling, which may provide new strategies to prevent bone metabolic disorders such as postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Gaoyang Chen
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Canling Long
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Shang Wang
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhenmin Wang
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xin Chen
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wanze Tang
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xiaoqin He
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhiteng Bao
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Baoyu Tan
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jin Zhao
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yongheng Xie
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhizhong Li
- The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Dazhi Yang
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China.
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China.
| | - Songlin Peng
- Department of Spine Surgery and Institute for Orthopaedic Research, the Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen, 518020, China.
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
21
|
Chen G, Wang S, Long C, Wang Z, Chen X, Tang W, He X, Bao Z, Tan B, Lu WW, Li Z, Yang D, Xiao G, Peng S. PiRNA-63049 inhibits bone formation through Wnt/β-catenin signaling pathway. Int J Biol Sci 2021; 17:4409-4425. [PMID: 34803507 PMCID: PMC8579447 DOI: 10.7150/ijbs.64533] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 10/13/2021] [Indexed: 12/16/2022] Open
Abstract
Bone remodeling is a dynamic process between bone formation mediated by osteoblasts and bone resorption mediated by osteoclasts. Disrupted bone remodeling is a key factor in postmenopausal osteoporosis, a metabolic disorder characterized by deteriorated bone microarchitecture and increased risk of fracture. Recent studies have shown that piwi-binding RNA (piRNA) is involved in the pathogenesis of certain diseases at the post-transcriptional level. Here, we analyzed piRNA-63049 (piR-63049), which may play an essential role in bone remodeling. The expression of piR-63049 significantly increased in both bone tissues and plasma of osteoporotic rats and postmenopausal osteoporotic patients. Overexpressing piR-63049 could inhibit the osteoblastogenesis of bone marrow stromal cells (BMSCs) while knocking down piR-63049 could promote the osteoblastogenesis of BMSCs through the Wnt2b/β-catenin signaling pathway. Moreover, knocking-down piR-63049 (piR-63049-antagonist) in vivo could attenuate the bone loss in ovariectomized rats by promoting bone formation. Taken together, the current study shows that piR-63049 inhibits bone formation through the Wnt2b/β-catenin signaling pathway. This novel piRNA may be a potential target to increase bone formation in bone loss disorders such as postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Gaoyang Chen
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Shang Wang
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Canling Long
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhenmin Wang
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xin Chen
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wanze Tang
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoqin He
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhiteng Bao
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Baoyu Tan
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - William W Lu
- Department of Orthopaedic and Traumatology, The University of Hong Kong, Hong Kong, 999077 China
| | - Zhizhong Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Dazhi Yang
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guozhi Xiao
- School of Medicine, Southern University of Science and Technology, Guangdong, Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen 518055, China
| | - Songlin Peng
- Department of Spine Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Shenzhen 518020, China.,The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
22
|
Jones TL, Esa MS, Li KHC, Krishnan SRG, Elgallab GM, Pearce MS, Young DA, Birrell FN. Osteoporosis, fracture, osteoarthritis & sarcopenia: A systematic review of circulating microRNA association. Bone 2021; 152:116068. [PMID: 34166859 DOI: 10.1016/j.bone.2021.116068] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/16/2021] [Accepted: 06/15/2021] [Indexed: 12/18/2022]
Abstract
Circulating microRNAs (c-miRs) show promise as biomarkers. This systematic review explores their potential association with age-related fracture/osteoporosis (OP), osteoarthritis (OA) and sarcopenia (SP), as well as cross-disease association. Most overlap occurred between OA and OP, suggesting potentially shared microRNA activity. There was little agreement in results across studies. Few reported receiver operating characteristic analysis (ROC) and many identified significant dysregulation in disease, but direction of effect was commonly conflicting. c-miRs with most evidence for consistency in dysregulation included miR-146a, miR-155 and miR-98 for OA (upregulated). Area under the curve (AUC) for miR-146a biomarker performance was AUC 0.92, p = 0.028. miR-125b (AUC 0.76-0.89), miR-100, miR-148a and miR-24 were consistently upregulated in OP. Insufficient evidence exists for c-miRs in SP. Study quality was typically rated intermediate/high risk of bias. Wide study heterogeneity meant meta-analysis was not possible. We provide detailed critique and recommendations for future approaches in c-miR analyses based on this review.
Collapse
Affiliation(s)
- Tania L Jones
- Population Health Sciences Institute, Faculty of Medicine, Newcastle University, Sir James Spence Building, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom.
| | - Mohammed S Esa
- Population Health Sciences Institute, Faculty of Medicine, Newcastle University, Sir James Spence Building, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom.
| | - K H Christien Li
- Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - S R Gokul Krishnan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom.
| | - George M Elgallab
- Faculty of Health Sciences and Wellbeing, Sciences Complex, City Campus, Chester Road, University of Sunderland, Sunderland SR1 3SD, United Kingdom
| | - Mark S Pearce
- Population Health Sciences Institute, Faculty of Medicine, Newcastle University, Sir James Spence Building, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom.
| | - David A Young
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom.
| | - Fraser N Birrell
- Population Health Sciences Institute, Faculty of Medicine, Newcastle University, Sir James Spence Building, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, United Kingdom; Medical Research Council Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom.
| |
Collapse
|
23
|
Xu Y, Ma J, Xu G, Ma D. Recent advances in the epigenetics of bone metabolism. J Bone Miner Metab 2021; 39:914-924. [PMID: 34250565 DOI: 10.1007/s00774-021-01249-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 07/03/2021] [Indexed: 12/22/2022]
Abstract
Osteoporosis is a common form of metabolic bone disease that is costly to treat and is primarily diagnosed on the basis of bone mineral density. As the influences of genetic lesions and environmental factors are increasingly studied in the pathological development of osteoporosis, regulated epigenetics are emerging as the important pathogenesis mechanisms in osteoporosis. Recently, osteoporosis genome-wide association studies and multi-omics technologies have revealed that susceptibility loci and the misregulation of epigenetic modifiers are key factors in osteoporosis. Over the past decade, extensive studies have demonstrated epigenetic mechanisms, such as DNA methylation, histone/chromatin modifications, and non-coding RNAs, as potential contributing factors in osteoporosis that affect disease initiation and progression. Herein, we review recent advances in epigenetics in osteoporosis, with a focus on exploring the underlying mechanisms and potential diagnostic/prognostic biomarker applications for osteoporosis.
Collapse
Affiliation(s)
- Yuexin Xu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Ma
- Department of Facial Plastic and Reconstructive Surgery, ENT Institute, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Guohua Xu
- Department of Orthopedic Surgery, The Spine Surgical Center, Changzheng Hospital, Second Military Medical University, Shanghai, 20000, China.
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Xun J, Li C, Liu M, Mei Y, Zhou Q, Wu B, Xie F, Liu Y, Dai R. Serum exosomes from young rats improve the reduced osteogenic differentiation of BMSCs in aged rats with osteoporosis after fatigue loading in vivo. Stem Cell Res Ther 2021; 12:424. [PMID: 34315544 PMCID: PMC8314589 DOI: 10.1186/s13287-021-02449-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/06/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Osteoporosis is a major public health concern for the elderly population and is characterized by fatigue load resulting in bone microdamage. The ability of bone mesenchymal stem cells (BMSCs) to repair bone microdamage diminishes with age, and the accumulation of bone microdamage increases the risk of osteoporotic fracture. There is a lack of effective means to promote the repair of bone microdamage in aged patients with osteoporosis. Exosomes have been shown to be related to the osteogenic differentiation of BMSCs. Here, we aimed to evaluate the changes in the osteogenic differentiation capacity of BMSCs in aged osteoporotic rats after fatigue loading and the treatment potential of serum exosomes from young rats. METHODS The tibias of six aged osteoporotic rats were subjected to fatigue loading in vivo for 2 weeks, and the bone microdamage, microstructures, and mechanical properties were assessed. Subsequently, BMSCs were extracted to evaluate their proliferation and osteogenic differentiation abilities. In addition, the BMSCs of aged osteoporotic rats after fatigue loading were treated with serum exosomes from young rats under osteogenic induction conditions, and the expression of osteogenic-related miRNAs was quantified. The osteogenetic effects of miRNA-19b-3p in exosomes and the possible target protein PTEN was detected. RESULTS Obvious bone microdamage at the fatigue load stress point, the bone microstructure and biomechanical properties were not obviously changed. A decreased osteogenic differentiation ability of BMSCs was observed after fatigue loading, while serum exosomes from young rats highly expressing miRNA-19b-3p improved the decreased osteogenic differentiation ability of BMSCs. Transfection with miRNA-19b-3p mimic could promote osteoblastic differentiation of BMSCs and decreased the expression of PTEN. After transfection of miRNA-19b-3p inhibitor, the promotional effect of exosomes on bone differentiation was weakened. Treatment with transfected exosomes increased the expression of PTEN. CONCLUSION Serum exosomes derived from young rats can improve the decreased osteogenic differentiation ability of BMSCs in aged rats with osteoporosis after fatigue loading and can provide a new treatment strategy for the repair of bone microdamage and prevention of fractures.
Collapse
Affiliation(s)
- Jingqiong Xun
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Endocrinology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Chan Li
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Meilu Liu
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yueming Mei
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qiongfei Zhou
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bo Wu
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Fen Xie
- Department of Endocrinology, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Yuling Liu
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ruchun Dai
- National Clinical Research Center for Metabolic Diseases, Institute of Metabolism and Endocrinology, Central South University, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
25
|
Ghafouri-Fard S, Abak A, Tavakkoli Avval S, Rahmani S, Shoorei H, Taheri M, Samadian M. Contribution of miRNAs and lncRNAs in osteogenesis and related disorders. Biomed Pharmacother 2021; 142:111942. [PMID: 34311172 DOI: 10.1016/j.biopha.2021.111942] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Non-coding RNAs have been found to regulate several developmental processes among them is osteogenesis. Although these transcripts have several distinct classes, two classes i.e. microRNAs and long non-coding RNAs have attained more attention. These transcripts regulate intramembranous as well as endochondral ossification processes. The effects of microRNAs on osteogenesis are mostly mediated through modulation of Wnt/β-catenin and TGFβ/BMP pathways. Long non-coding RNAs can directly affect expression of these pathways or osteogenic transcription factors. Moreover, they can serve as a molecular sponge for miRNAs. MALAT1/miR-30, MALAt1/miR-214, LEF1-AS1/miR-24-3p, MCF2L-AS1/miR-33a, MSC-AS1/miR-140-5p and KCNQ1OT1/miR-214 are examples of such kind of interaction between lncRNAs and miRNAs in the context of osteogenesis. In the current paper, we explain these two classes of non-coding RNAs in the osteogenesis and related disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shayan Rahmani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Samadian
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Wu YZ, Huang HT, Cheng TL, Lu YM, Lin SY, Ho CJ, Lee TC, Hsu CH, Huang PJ, Huang HH, Li JY, Su YD, Chen SC, Kang L, Chen CH. Application of microRNA in Human Osteoporosis and Fragility Fracture: A Systemic Review of Literatures. Int J Mol Sci 2021; 22:ijms22105232. [PMID: 34063380 PMCID: PMC8156577 DOI: 10.3390/ijms22105232] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) could serve as ideal entry points to the deregulated pathways in osteoporosis due to their relatively simple upstream and downstream relationships with other molecules in the signaling cascades. Our study aimed to give a comprehensive review of the already identified miRNAs in osteoporosis from human blood samples and provide useful information for their clinical application. A systematic literature search for relevant studies was conducted in the Pubmed database from inception to December 2020. We set two essential inclusion criteria: human blood sampling and design of controlled studies. We sorted the results of analysis on human blood samples according to the study settings and compiled the most promising miRNAs with analyzed diagnostic values. Furthermore, in vitro and in vivo evidence for the mechanisms of the identified miRNAs was also illustrated. Based on both diagnostic value and evidence of mechanism from in vitro and in vivo experiments, miR-23b-3p, miR-140-3p, miR-300, miR-155-5p, miR-208a-3p, and miR-637 were preferred candidates in diagnostic panels and as therapeutic agents. Further studies are needed to build sound foundations for the clinical usage of miRNAs in osteoporosis.
Collapse
Affiliation(s)
- Yen-Zung Wu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Hsuan-Ti Huang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Tsung-Lin Cheng
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Yen-Mou Lu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Cheng-Jung Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Tien-Ching Lee
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Chia-Hao Hsu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Peng-Ju Huang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Han Hsiang Huang
- Department of Veterinary Medicine, National Chiayi University, Chiayi 60004, Taiwan;
| | - Jhong-You Li
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
| | - Yu-De Su
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
| | - Shih-Chieh Chen
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Department of Medical Records, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (L.K.); (C.-H.C.); Tel.: +886-7-3209-209 (C.-H.C.)
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80420, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
- Correspondence: (L.K.); (C.-H.C.); Tel.: +886-7-3209-209 (C.-H.C.)
| |
Collapse
|
27
|
Use of Omics Data in Fracture Prediction; a Scoping and Systematic Review in Horses and Humans. Animals (Basel) 2021; 11:ani11040959. [PMID: 33808497 PMCID: PMC8065418 DOI: 10.3390/ani11040959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Despite many recent advances in imaging and epidemiological data analysis, musculoskeletal injuries continue to be a welfare issue in racehorses. Omics studies describe the study of protein, genetic material (both DNA and RNA, including microRNAs—small non-coding ribonucleic acids) and metabolites that may provide insights into the pathophysiology of disease or opportunities to monitor response to treatment when measured in bodily fluids. As these fields of study are scientifically complex and highly specialised, it is timely to perform a review of the current literature to allow for the design of robust studies that allow for repeatable work. Systematic reviews have been introduced into the medical literature and are a methodological way of searching for relevant papers followed by critical review of the content and a detection of biases. The objectives of the current systematic review were to identify and critically appraise the literature pertaining to microRNA (miRNA) and their target genes that are correlated with stress fractures in racehorses and humans. The object was to define a panel of miRNAs and their target genes as potential biomarkers in either horses or human subjects. The online scientific databases were searched and a reviewed was performed according to preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. MicroRNA profiling studies in horses continue to emerge, but as of yet, no miRNA profile can reliably predict the occurrence of fractures. It is very important that future studies are well designed to mitigate the effects of variation in sample size, exercise and normalisation methods. Abstract Despite many recent advances in imaging and epidemiological data analysis, musculoskeletal injuries continue to be a welfare issue in racehorses. Peptide biomarker studies have failed to consistently predict bone injury. Molecular profiling studies provide an opportunity to study equine musculoskeletal disease. A systematic review of the literature was performed using preferred reporting items for systematic reviews and meta-analyses protocols (PRISMA-P) guidelines to assess the use of miRNA profiling studies in equine and human musculoskeletal injuries. Data were extracted from 40 papers between 2008 and 2020. Three miRNA studies profiling equine musculoskeletal disease were identified, none of which related to equine stress fractures. Eleven papers studied miRNA profiles in osteoporotic human patients with fractures, but differentially expressed miRNAs were not consistent between studies. MicroRNA target prediction programmes also produced conflicting results between studies. Exercise affected miRNA profiles in both horse and human studies (e.g., miR-21 was upregulated by endurance exercise and miR-125b was downregulated by exercise). MicroRNA profiling studies in horses continue to emerge, but as yet, no miRNA profile can reliably predict the occurrence of fractures. It is very important that future studies are well designed to mitigate the effects of variation in sample size, exercise and normalisation methods.
Collapse
|
28
|
Wang L, Dong J, Ma J, Lu Q, Shan B, Han T, Xie P, Zuo X. By inhibiting ADCY5, miR-18a-3p promotes osteoporosis and possibly contributes to spinal fracture. Biochem Biophys Res Commun 2021; 550:49-55. [PMID: 33684620 DOI: 10.1016/j.bbrc.2021.02.118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/23/2023]
Abstract
To investigate the influence of miR-18a-3p and ADCY5 on OP and osteogenic differentiation of human Mesenchymal stem cell (hBMSCs) and its possible mechanism. Samples were collected from osteoporotic patients with or without vertebral compression fracture, and without OP volunteers. MiR-18a-3p and ADCY5 mRNA expression levels in the tissue samples and hBMSCs during osteogenic differentiation were detected。MiR-18a-3p mimic and OE-ADCY5 were introduced into hBMSCs to research the effects of miR-18a-3p and ADCY5 on osteogenesis differentiation of hBMSCs. Dual luciferase reporter system and RNA pull-down were applied to determine whether ADCY5 was a target gene of miR-18a-3p. Compared with the control group, ADCY5 expression level was down-regulated in patients with OP-no-Frx and OP-Frx, but that of miR-18a-3p was up-regulated. In addition, ADCY5 increased during osteogenesis differentiation of hBMSCs, whereas miR-18a-3p did not. OE-ADCY5 significantly facilitated calcium deposition, ALP activity, osteoblast protein expression (OSX, ALP and EUNX2), miR-18a-3p mimic inhibited osteogenic differentiation, and partially reversed the effect of OE-ADCY5 on osteogenic differentiation. In general, miR-18a-3p targets ADCY5 to promote OP and may be involved in spinal fracturs.
Collapse
Affiliation(s)
- Lei Wang
- Department of Anesthesiology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huaian, 223001, Jiangsu, China
| | - Junli Dong
- Department of Pain Management, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China
| | - Jun Ma
- Department of Radiology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huaian, 223001, Jiangsu, China
| | - Qin Lu
- Department of Ultrasonography, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huaian, 223001, Jiangsu, China
| | - Ben Shan
- Department of Radiology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huaian, 223001, Jiangsu, China
| | - Tingting Han
- Department of Radiology, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huaian, 223001, Jiangsu, China
| | - Peng Xie
- Department of Neurosurgery, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huai'an, 223002, China.
| | - Xiaohua Zuo
- Department of Pain Management, The Affiliated Huai'an Hospital of Xuzhou Medical University and the Second People's Hospital of Huai'an, Huaian, 223001, Jiangsu, China.
| |
Collapse
|
29
|
Zhi F, Ding Y, Wang R, Yang Y, Luo K, Hua F. Exosomal hsa_circ_0006859 is a potential biomarker for postmenopausal osteoporosis and enhances adipogenic versus osteogenic differentiation in human bone marrow mesenchymal stem cells by sponging miR-431-5p. Stem Cell Res Ther 2021; 12:157. [PMID: 33648601 PMCID: PMC7923524 DOI: 10.1186/s13287-021-02214-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/07/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND As one of the most common chronic diseases in the world, osteoporosis occurs especially in postmenopausal women. Circular RNAs (circRNAs) are emerging as major drivers in human disease. The aim of the present study was to analyse circRNA expression profiles in osteoporosis and to explore the clinical significance and the regulatory molecular mechanism of hsa_circ_0006859 during osteoporosis. METHODS Exosomes were isolated from clinically collected serum samples. A circRNA microarray was performed to screen differentially expressed circRNAs. Quantitative real-time PCR (qRT-PCR) and western blot were performed to analyse target gene mRNA expression and protein expression. Alizarin red staining (ARS) was performed to evaluate the mineralization ability of human bone marrow mesenchymal stem cells (hBMSCs). Oil Red O staining was performed to evaluate the lipid droplet formation ability of hBMSCs. Bioinformatics analysis and the luciferase reporter assay were performed to investigate the interaction between two genes. RESULTS Hsa_circ_0006859 was identified as one of the most upregulated circRNAs in the microarray analysis. Hsa_circ_0006859 in exosomes was upregulated in osteoporosis patients compared to healthy controls. Hsa_circ_0006859 differentiated osteopenia or osteoporosis patients from healthy controls with high sensitivity and specificity. Hsa_circ_0006859 suppressed osteoblastic differentiation and promoted adipogenic differentiation of hBMSCs. Hsa_circ_0006859 directly bound to miR-431-5p, and ROCK1 was identified as a novel target gene of miR-431-5p. Hsa_circ_0006859 is a competing endogenous RNA (ceRNA) of miR-431-5p that promotes ROCK1 expression. Hsa_circ_0006859 suppressed osteogenesis and promoted adipogenesis by sponging miR-431-5p to upregulate ROCK1. CONCLUSIONS Exosomal hsa_circ_0006859 is a potential biomarker for postmenopausal osteoporosis and controls the balance between osteogenesis and adipogenesis in hBMSCs by sponging miR-431-5p.
Collapse
Affiliation(s)
- Feng Zhi
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou City, 213003, Jiangsu, China.
| | - Yi Ding
- Department of Geriatrics, Third Affiliated Hospital of Soochow University, Changzhou City, 213003, Jiangsu, China
| | - Rong Wang
- Department of Neurosurgery, Third Affiliated Hospital of Soochow University, Changzhou City, 213003, Jiangsu, China
| | - Yujiao Yang
- Department of Geriatrics, Third Affiliated Hospital of Soochow University, Changzhou City, 213003, Jiangsu, China
| | - Kaiming Luo
- Department of Endocrinology, Third Affiliated Hospital of Soochow University, Changzhou City, 213003, Jiangsu, China
| | - Fei Hua
- Department of Endocrinology, Third Affiliated Hospital of Soochow University, Changzhou City, 213003, Jiangsu, China.
| |
Collapse
|
30
|
Xiao P, Zhu Z, Du C, Zeng Y, Liao J, Cheng Q, Chen H, Zhao C, Huang W. Silencing Smad7 potentiates BMP2-induced chondrogenic differentiation and inhibits endochondral ossification in human synovial-derived mesenchymal stromal cells. Stem Cell Res Ther 2021; 12:132. [PMID: 33588941 PMCID: PMC7885459 DOI: 10.1186/s13287-021-02202-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/31/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Bone morphogenetic protein 2 (BMP2) is a promising chondrogenic growth factor for cartilage tissue-engineering, but it also induces robust endochondral ossification. Human synovial-derived mesenchymal stromal cells (hSMSCs) have attracted great interest due to their poor potential for differentiation into osteogenic lineages. Smad7 plays a significant in the endochondral ossification. In this study, we explored a new method to amplify the BMP2-induced chondrogenic differentiation of hSMSCs by downregulating Smad7 and applying a cellular scaffold. METHODS hSMSCs were isolated from human knee joint synovium from 3 donors through adhesion growth. In vitro and in vivo models of the chondrogenic differentiation of hSMSCs were established. Transgenic expression of BMP2 and silencing of Smad7 and Smad7 was achieved by adenoviral vectors. The osteogenic differentiation was detected by alkaline phosphatase staining, alizarin red staining, and RT-PCR analysis of the osteogenic genes RUNX2, Osterix, and Osteocalcin. The chondrogenic differentiation was detected by Alcian blue staining and RT-PCR analysis of the chondrogenic genes SOX9, COL2, and aggrecan. Hypertrophic differentiation was detected by the markers COL10 and MMP13. A subcutaneous stem cell implantation model was established with polyethylene glycol citrate-co-N-isopropylacrylamide (PPCN) scaffolds and athymic nude mice (3/group, 4-6 week-old female) and evaluated by micro-CT, H&E staining, and Alcian blue staining. An immunohistochemistry assay was used to detected COL1 and COL2, and an immunofluorescence assay was used to detect COL10 and MMP13. RESULTS These hSMSCs identified by flow cytometry. These hSMSCs exhibited lower osteo-differentiation potential than iMads and C3H10T1/2-cells. When Smad7 was silenced in BMP2-induced hSMSCs, the chondrogenic differentiation genes SOX9, COL2, and aggrecan were enhanced in vitro. Additionally, it silencing Smad7 led to a decrease in the hypertrophic differentiation genes COL10 and MMP13. In subcutaneous stem cell implantation assays, immunofluorescence and immunohistochemical staining demonstrated that silencing Smad7 increased the number of COL2-positive cells and decreased the expression of COL1, COL10, and MMP13. CONCLUSION This study suggests that the application of hSMSCs, cell scaffolds, and silencing Smad7 can potentiate BMP2-induced chondrogenic differentiation and inhibit endochondral ossification. Thus, inhibiting the expression of Smad7 in BMP2-induced hSMSC differentiation may be a new strategy for cartilage tissue-engineering.
Collapse
Affiliation(s)
- Pengcheng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhenglin Zhu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengcheng Du
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yongsheng Zeng
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junyi Liao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qiang Cheng
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chen Zhao
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
31
|
Circulating MicroRNAs as Novel Biomarkers for Osteoporosis and Fragility Fracture Risk: Is There a Use in Assessment Risk? Int J Mol Sci 2020; 21:ijms21186927. [PMID: 32967246 PMCID: PMC7555752 DOI: 10.3390/ijms21186927] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
Osteoporosis is a multifactorial skeletal disease that is associated with both bone mass decline and microstructure damage. The fragility fractures-especially those affecting the femur-that embody the clinical manifestation of this pathology continue to be a great medical and socioeconomic challenge worldwide. The currently available diagnostic tools, such as dual energy X-ray absorptiometry, Fracture Risk Assessment Tool (FRAX) score, and bone turnover markers, show limited specificity and sensitivity; therefore, the identification of alternative approaches is necessary. As a result of their advantageous features, such as non-invasiveness, biofluid stability, and easy detection, circulating cell-free miRs are promising new potential biomarkers for the diagnosis of osteoporosis and low-traumatic fracture risk assessment. However, due to the absence of both standardized pre-analytical, analytical, and post-analytical protocols for their measurement and universally accepted guidelines for diagnostic use, their clinical utility is limited. The aim of this review was to record all the data currently available in the literature concerning the use of circulating microRNAs as both potential biomarkers for osteoporosis diagnosis and fragility fracture risk evaluation, and group them according to the experimental designs, in order to support a more conscious choice of miRs for future research in this field.
Collapse
|
32
|
Donati S, Ciuffi S, Palmini G, Brandi ML. Circulating miRNAs: A New Opportunity in Bone Fragility. Biomolecules 2020; 10:biom10060927. [PMID: 32570976 PMCID: PMC7355961 DOI: 10.3390/biom10060927] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis, one of the leading causes of bone fractures, is characterized by low bone mass and structural deterioration of bone tissue, which are associated with a consequent increase in bone fragility and predisposition to fracture. Current screening tools are limited in estimating the proper assessment of fracture risk, highlighting the need to discover novel more suitable biomarkers. Genetic and environmental factors are both implicated in this disease. Increasing evidence suggests that epigenetics and, in particular, miRNAs, may represent a link between these factors and an increase of fracture risk. miRNAs are a class of small noncoding RNAs that negatively regulate gene expression. In the last decade, several miRNAs have been associated with the development of osteoporosis and bone fracture risk, opening up new possibilities in precision medicine. Recently, these molecules have been identified in several biological fluids, and the possible existence of a circulating miRNA (c-miRNA) signature years before the fracture occurrence is suggested. The aim of this review is to provide an overview of the c-miRNAs suggested as promising biomarkers for osteoporosis up until now, which could be helpful for early diagnosis and monitoring of treatment response, as well as fracture risk assessment, in osteoporotic patients.
Collapse
Affiliation(s)
- Simone Donati
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Study of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (S.D.); (S.C.); (G.P.)
| | - Simone Ciuffi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Study of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (S.D.); (S.C.); (G.P.)
| | - Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Study of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (S.D.); (S.C.); (G.P.)
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Study of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (S.D.); (S.C.); (G.P.)
- Unit of Bone and Mineral Diseases, University Hospital of Florence, Largo Palagi 1, 50139 Florence, Italy
- Correspondence: ; Tel.: +39-055-7946304; Fax: +39-055-7946303
| |
Collapse
|
33
|
Zhou L, Qiu M, Yang L, Yang L, Zhang Y, Mu S, Song H. MicroRNA-1-3p enhances osteoblast differentiation of MC3T3-E1 cells by interacting with hypoxia-inducible factor 1 α inhibitor (HIF1AN). Mech Dev 2020; 162:103613. [PMID: 32387587 DOI: 10.1016/j.mod.2020.103613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/19/2023]
Abstract
Studies have proved that miRNAs participate in the regulation of osteoblast differentiation (OD), and abnormal expression of miRNAs is related with various states of OD. In this study, we investigated the role of miRNA-1-3p in OD using MC3T3-E1 cells. BMP2 is used to induce OD of MC3T3-E1 cells. MiRNA-1-3p mimics or miRNA-1-3p inhibitor was transfected to MC3T3-E1 cells with BMP2. The expression levels of miRNA-1-3p were determined by qRT-PCR. The expression of Runx2, OSX, OPN, and OCN was detected by Western blotting. ALP assay was performed to measure alkaline phosphatase activity. Calcium nodules were evaluated by alizarin red staining. Over-expression of hypoxia-inducible factor 1-alpha inhibitor (HIF1AN) was performed and miRNA-1-3p rescue experiments were carried out. Over-expression of miRNA-1-3p promoted osteogenic differentiations and calcifications, as demonstrated by increased ALP, calcification and osteogenic markers. Knock-down of miRNA-1-3p generated the opposite results. HIF1AN was identified to be directly targeted by miRNA-1-3p. Over-expression of HIF1AN suppressed OD and calcifications, and miRNA-1-3p reversed the effect. Our results demonstrated that miRNA-1-3p could enhance OD of MC3T3-E1 cells through interacting with HIF1AN, which might be employed as therapeutic applications for bone formation and regeneration.
Collapse
Affiliation(s)
- Long Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Min Qiu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Lei Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Liyu Yang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Yiqi Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Shuai Mu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Hanyi Song
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China.
| |
Collapse
|
34
|
Zhang W, Xu H, Zhao X, Tang X, Yang S, Yu L, Zhao S, Chang K, Chen M. 3D DNA nanonet structure coupled with target-catalyzed hairpin assembly for dual-signal synergistically amplified electrochemical sensing of circulating microRNA. Anal Chim Acta 2020; 1122:39-47. [PMID: 32503742 DOI: 10.1016/j.aca.2020.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/30/2020] [Accepted: 05/01/2020] [Indexed: 12/18/2022]
Abstract
DNA nanomaterials are reliable and powerful tools in the development of a variety of biosensors owing to their notable self-assembly ability and precise recognition capability. Here, we propose a DNA nanomaterial-based system for the dual-amplified electrochemical sensing of circulating microRNAs by a coupled cascade of catalyzed hairpin assembly (CHA) and three-dimensional (3D) DNA nanonet structure. In the target-assisted CHA process, the stable hairpin structures H1 and H2 act as probes for the recognition and recycling of circulating microRNAs, leading to the formation of abundant H1-H2 duplexes with tails. Subsequently, a 3D DNA nanonet structure was introduced, which was assembled using three DNA strands constructed X-DNA monomers as the building blocks, and hybridized to the tails of H1-H2 duplexes. The successful integration of target-assisted CHA and 3D DNA nanonet structure induced the second signal amplification. The designed biosensor performed under optimized experimental conditions, and exposed admirable analytical performance for the detection of circulating miR-21, with a wide linear range from 10 fM to 1 nM, high sensitivity of limit of detection (LOD) of 3.6083 fM, good specificity in the face of single nucleotides and other microRNAs, satisfactory stability and reproducibility for practical analysis. Furthermore, the clinical applicability for circulating miR-21 detection was verified in human serum samples without additional treatment. We hope that this elaborated biosensor will provide new opportunities for bioassays based on DNA nanomaterials.
Collapse
Affiliation(s)
- Wenqing Zhang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Huan Xu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xianxian Zhao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Xiaoqi Tang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Sha Yang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Lianyu Yu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Shuang Zhao
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China
| | - Kai Chang
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.
| | - Ming Chen
- Department of Clinical Laboratory Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China; College of Pharmacy and Laboratory Medicine, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China; State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), 30 Gaotanyan, Shapingba District, Chongqing, 400038, China.
| |
Collapse
|
35
|
Bottani M, Banfi G, Lombardi G. The Clinical Potential of Circulating miRNAs as Biomarkers: Present and Future Applications for Diagnosis and Prognosis of Age-Associated Bone Diseases. Biomolecules 2020; 10:E589. [PMID: 32290369 PMCID: PMC7226497 DOI: 10.3390/biom10040589] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/01/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis, related fracture/fragility, and osteoarthritis are age-related pathologies that, over recent years, have seen increasing incidence and prevalence due to population ageing. The diagnostic approaches to these pathologies suffer from limited sensitivity and specificity, also in monitoring the disease progression or treatment. For this reason, new biomarkers are desirable for improving the management of osteoporosis and osteoarthritis patients. The non-coding RNAs, called miRNAs, are key post-transcriptional factors in bone homeostasis, and promising circulating biomarkers for pathological conditions in which to perform a biopsy can be problematic. In fact, miRNAs can easily be detected in biological fluids (i.e., blood, serum, plasma) using methods with elevated sensitivity and specificity (RT-qPCR, microarray, and NGS). However, the analytical phases required for miRNAs' evaluation still present some practical issues that limit their use in clinical practice. This review reveals miRNAs' potential as circulating biomarkers for evaluating predisposition, diagnosis, and prognosis of osteoporosis (postmenopausal or idiopathic), bone fracture/fragility, and osteoarthritis, with a focus on pre-analytical, analytical, and post-analytical protocols used for their validation and thus on their clinical applicability. These evidences may support the definition of early diagnostic tools based on circulating miRNAs for bone diseases and osteoarthritis as well as for monitoring the effects of specific treatments.
Collapse
Affiliation(s)
- Michela Bottani
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi 4, 20161 Milano, Italy; (M.B.); (G.B.)
| | - Giuseppe Banfi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi 4, 20161 Milano, Italy; (M.B.); (G.B.)
- Vita-Salute San Raffaele University, 20132 Milano, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Riccardo Galeazzi 4, 20161 Milano, Italy; (M.B.); (G.B.)
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Królowej Jadwigi 27/39, 61-871 Poznań, Poland
| |
Collapse
|
36
|
Zhang Z, Li X, Li A, Wu G. miR-485-5p suppresses Schwann cell proliferation and myelination by targeting cdc42 and Rac1. Exp Cell Res 2020; 388:111803. [DOI: 10.1016/j.yexcr.2019.111803] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 10/25/2022]
|