1
|
Yalaev BI, Kaletnik EI, Karpova YS, Belaya ZE, Minniakhmetov IR, Mokrysheva NG, Khusainova RI. The Role of microRNA in the Regulation of Differentiation and the Functionality of Osteoblasts, Osteoclasts, and Their Precursors in Osteoporosis. Noncoding RNA 2025; 11:14. [PMID: 39997614 PMCID: PMC11858178 DOI: 10.3390/ncrna11010014] [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: 12/05/2024] [Revised: 01/16/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Osteoporosis is a complex disease that is affected by a variety of factors, including genetic and epigenetic influences. While DNA markers for osteoporosis have been identified, they do not fully explain the hereditary basis of the disease. Epigenetic factors, such as small microRNAs (miRNAs), may provide a missing link in understanding the molecular mechanisms underlying osteoporosis. miRNAs are a class of non-coding RNAs that play a role in the epigenetic regulation of gene expression. They are known to be involved in various biological processes, including bone formation and remodelling. Differential expression of miRNAs has been linked to the pathological decrease in bone mineral density associated with osteoporosis. It has been shown that an abnormal miRNA expression pattern leads to a decrease in osteoblast activity and an increase in osteoclast activity. Further research into the role of miRNAs in osteoporosis may help to better understand this disease and identify potential therapeutic targets for treatment. Based on these assumptions, the study of miRNA expression patterns in osteoblasts, osteoclasts, and their precursors under normal and osteoporotic conditions is a rapidly growing field of scientific research. Although the results of this research are still incomplete and sometimes contradictory, they require additional scientific analysis to better understand the complex mechanisms involved. The purpose of this paper is to review the current research on miRNAs specifically expressed in osteoblasts and osteoclasts under both normal and pathological conditions. We will also discuss the potential applications of these miRNAs as biomarkers for osteoporosis diagnosis and as targets for osteoporosis treatment.
Collapse
Affiliation(s)
- Bulat I. Yalaev
- Endocrinology Research Centre, Dmitry Ulyanov Street, 11, 117292 Moscow, Russia; (E.I.K.); (Y.S.K.); (Z.E.B.); (I.R.M.); (N.G.M.); (R.I.K.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Wu H, Liao X, Wu T, Xie B, Ding S, Chen Y, Song L, Wei B. Mechanism of MiR-145a-3p/Runx2 pathway in dexamethasone impairment of MC3T3-E1 osteogenic capacity in mice. PLoS One 2024; 19:e0309951. [PMID: 39561180 PMCID: PMC11575826 DOI: 10.1371/journal.pone.0309951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/21/2024] [Indexed: 11/21/2024] Open
Abstract
OBJECTIVE In this experiment, we screened key miRNAs involved in the dexamethasone-induced decrease in osteogenic capacity of mouse precursor osteoblasts MC3T3-E1 over and investigated their specific regulatory mechanisms. METHODS In this experiment, cell counting kit assay was utilized to act on MC3T3-E1 cells at 0, 5μM, 10μM, 15μM concentrations of dexamethasone for 24h, 48h and 72h to observe the changes in cell viability in order to select the appropriate dexamethasone concentration. Apoptosis and reactive oxygen species were detected by flow cytometry. The transcription of osteogenesis-related genes (Runx2, ALP, OCN, OPN, OPG, COL1A1) and protein expression levels (Runx2, ALP, OCN, OPN) were detected by Western Blot and qRT-PCR to validate the changes in cellular osteogenesis. The differentially expressed miRNAs related to MC3T3-E1 osteogenic differentiation after dexamethasone action were screened out. The expression levels of selected target miRNAs were verified in the experimental group and the control group by qRT-PCR. The miRNA inhibitor was transfected to knock down miRNA in dexamethasone-induced MC3T3-E1 injury. Alkaline phosphatase staining and flow cytometry were performed to detect apoptosis and reactive oxygen species changes. transcript and protein expression levels of osteogenesis-related genes in mouse MC3T3-E1 were detected by qRT-PCR and Western blot experiments. By miRNA target gene prediction, luciferase reporter gene experiments, qRT-PCR and Western blot experiments were used to verify whether the selected target miRNAs targeted the target gene. RESULTS First, it was determined that 10μM dexamethasone solution was effective in inducing a decrease in osteogenic function in mouse MC3T3-E1 by CCK8 experiments, which showed a significant decrease in alkaline phosphatase activity, a decrease in calcium nodules as shown by alizarin red staining, an increase in apoptosis and reactive oxygen species as detected by flow cytometry, as well as a decrease in the expression of osteogenesis-related genes and proteins. Five target miRNAs were identified: miR-706, miR-296-3p, miR-7011-5p, miR-145a-3p, and miR-149-3p. miR-145a-3p, which had the most pronounced and stable expression trend and was the most highly expressed miRNA, was chosen as the target of this experiment by qRT-PCR analysis. -145a-3p, as the subject of this experiment. Knockdown of miR-145a-3p in MC3T3-E1 cells after dexamethasone action significantly improved the expression of their impaired osteogenic indicators. It was shown that after knocking down the target miRNA, alkaline phosphatase staining was significantly increased compared with the dexamethasone-stimulated group and approached the level of the blank control group. Meanwhile, the expression of osteogenic function-related proteins and genes also increased in the dexamethasone-stimulated group after knocking down miR-145a-3p, and approached the level of the blank control group. A direct targeting relationship between miR-145a-3p and Runx2 was indeed confirmed by luciferase reporter gene assays, qRT-PCR and Western blot experiments. CONCLUSIONS The results indicated that dexamethasone impaired the osteogenic differentiation ability of MC3T3-E1 cells by inducing the up-regulation of miR-145a-3p expression. MiR-145a-3p inhibited the osteogenic differentiation ability of MC3T3-E1 cells by targeting and suppressing the expression level of Runx2 protein. Inhibition of miR-145a-3p levels significantly improved the osteogenic differentiation ability of MC3T3-E1 cells.
Collapse
Affiliation(s)
- Hang Wu
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xinghua Liao
- Central People's Hospital of Zhanjiang, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Tingrui Wu
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bin Xie
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Sicheng Ding
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yiren Chen
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lijun Song
- Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bo Wei
- Orthopedics Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
3
|
Xu J, Huang Z, Shi S, Xia J, Chen G, Zhou K, Zhang Y, Bian C, Shen Y, Yin X, Lu L, Gu H. Glial maturation factor-β deficiency prevents oestrogen deficiency-induced bone loss by remodelling the actin network to suppress adipogenesis of bone marrow mesenchymal stem cells. Cell Death Dis 2024; 15:829. [PMID: 39543090 PMCID: PMC11564563 DOI: 10.1038/s41419-024-07234-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
An imbalance between the adipogenesis and osteogenesis of bone marrow mesenchymal stem cells (BMSCs) is considered the basic pathogenesis of osteoporosis. Although actin cytoskeleton remodelling plays a crucial role in the differentiation of BMSCs, the role of actin cytoskeleton remodelling in the adipogenesis of BMSCs and postmenopausal osteoporosis (PMOP) has remained elusive. Glia maturation factor-beta (GMFB) has a unique role in remodelling the polymerization/depolymerization cycles of actin. We observed that GMFB expression was increased in bone tissue from both ovariectomized (OVX) rats and PMOP patients. GMFB knockout inhibited the accumulation of bone marrow adipocytes and increased bone mass in the OVX rat model. The inhibition of adipocyte differentiation in GMFB knockout BMSCs was mediated via actin cytoskeleton remodelling and the Ca2+-calcineurin-NFATc2 axis. Furthermore, we found that GMFB shRNA treatment in vivo had favourable effects on osteoporosis induced by OVX. Together, these findings suggest a pathological association of the GMFB with PMOP and highlight the potential of the GMFB as a therapeutic target for osteoporosis patients.
Collapse
Affiliation(s)
- Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Si Shi
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China
| | - Jiangni Xia
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Guangnan Chen
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Yiming Zhang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Chong Bian
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Yuqin Shen
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China.
| | - Lixia Lu
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China.
- Department of Biochemistry and Molecular Biology, Tongji University School of medicine, Shanghai, PR China.
| | - Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
4
|
Zeng X, Yuan X, Liao H, Wei Y, Wu Q, Zhu X, Li Q, Chen S, Hu M. The miR-665/SOST Axis Regulates the Phenotypes of Bone Marrow Mesenchymal Stem Cells and Osteoporotic Symptoms in Female Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2059-2075. [PMID: 39461772 DOI: 10.1016/j.ajpath.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 10/29/2024]
Abstract
Osteoporosis is a common degenerative skeletal disease among older people, especially postmenopausal women. Bone marrow mesenchymal stem cells (BMSCs), the progenitors of osteoblasts, are essential to the pathophysiology of osteoporosis. Herein, targeting miRNAs with differential expression in dysfunctional BMSCs was accomplished by bioinformatics analysis based on public databases. Target mRNAs were predicted and applied for signaling pathway and function enrichment annotations. In vitro and in vivo effects of selected miRNA on BMSC proliferation and osteogenesis were investigated, the putative binding between selected miRNA and predicted target mRNA was verified, and the co-effects of the miRNA/mRNA axis on BMSCs were determined. miRNA 665 (miR-665) was down-regulated in osteoporotic BMSCs compared with normal BMSCs and elevated in BMSCs experiencing osteogenic differentiation. In BMSCs, miR-665 overexpression promoted cell proliferation and osteogenic differentiation. miR-665 targeted the Wnt signaling inhibitor sclerostin (SOST) and inhibited SOST mRNA and protein expression. SOST overexpression inhibited BMSC cell proliferation and osteogenic differentiation. When co-transduced to BMSCs, SOST knockdown significantly reversed the effects of miR-665 on BMSCs. In ovariectomy (OVX)-induced osteoporosis model mice, OVX remarkably decreased bone mass, whereas miR-665 overexpression partially improved OVX-induced bone mass loss. miR-665 was down-regulated in osteoporotic BMSCs and up-regulated in osteogenically differentiated BMSCs. In conclusion, the miR-665/SOST axis modulates BMSC proliferation, osteogenic differentiation, and OVX-induced osteoporosis in mice, possibly through Wnt signaling.
Collapse
Affiliation(s)
- Xingxing Zeng
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Xianyu Yuan
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Hongchun Liao
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Yongfang Wei
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Qinxuan Wu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Xi Zhu
- Health Management, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qingqing Li
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China
| | - Shijie Chen
- Department of Orthopedics, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Minghua Hu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China; The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Changsha Medical University, Changsha, China.
| |
Collapse
|
5
|
Dzubanova M, Bond JM, Craige SM, Tencerova M. NOX4-reactive oxygen species axis: critical regulators of bone health and metabolism. Front Cell Dev Biol 2024; 12:1432668. [PMID: 39188529 PMCID: PMC11345137 DOI: 10.3389/fcell.2024.1432668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Bone marrow stromal cells (BMSCs) play a significant role in bone metabolism as they can differentiate into osteoblasts, bone marrow adipocytes (BMAds), and chondrocytes. BMSCs chronically exposed to nutrient overload undergo adipogenic programming, resulting in bone marrow adipose tissue (BMAT) formation. BMAT is a fat depot transcriptionally, metabolically, and morphologically distinct from peripheral adipose depots. Reactive oxygen species (ROS) are elevated in obesity and serve as important signals directing BMSC fate. ROS produced by the NADPH oxidase (NOX) family of enzymes, such as NOX4, may be responsible for driving BMSC adipogenesis at the expense of osteogenic differentiation. The dual nature of ROS as both cellular signaling mediators and contributors to oxidative stress complicates their effects on bone metabolism. This review discusses the complex interplay between ROS and BMSC differentiation in the context of metabolic bone diseases.Special attention is paid to the role of NOX4-ROS in regulating cellular processes within the bone marrow microenvironment and potential target in metabolic bone diseases.
Collapse
Affiliation(s)
- Martina Dzubanova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Jacob M. Bond
- Translational Biology, Medicine, and Health, Virginia Tech, Roanoke, VA, United States
| | - Siobhan M. Craige
- Human Nutrition, Foods and Exercise, Virginia Tech, Blacksburg, VA, United States
| | - Michaela Tencerova
- Laboratory of Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
6
|
Li M, Liu JX, Ma B, Liu JY, Chen J, Jin F, Hu CH, Xu HK, Zheng CX, Hou R. A Senescence-Associated Secretory Phenotype of Bone Marrow Mesenchymal Stem Cells Inhibits the Viability of Breast Cancer Cells. Stem Cell Rev Rep 2024; 20:1093-1105. [PMID: 38457059 DOI: 10.1007/s12015-024-10710-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] [Accepted: 02/28/2024] [Indexed: 03/09/2024]
Abstract
Breast cancer, the most prevalent malignancy in women, often progresses to bone metastases, especially in older individuals. Dormancy, a critical aspect of bone-metastasized breast cancer cells (BCCs), enables them to evade treatment and recur. This dormant state is regulated by bone marrow mesenchymal stem cells (BMMSCs) through the secretion of various factors, including those associated with senescence. However, the specific mechanisms by which BMMSCs induce dormancy in BCCs remain unclear. To address this gap, a bone-specific senescence-accelerated murine model, SAMP6, was utilized to minimize confounding systemic age-related factors. Confirming senescence-accelerated osteoporosis, distinct BMMSC phenotypes were observed in SAMP6 mice compared to SAMR1 counterparts. Notably, SAMP6-BMMSCs exhibited premature senescence primarily due to telomerase activity loss and activation of the p21 signaling pathway. Furthermore, the effects of conditioned medium (CM) derived from SAMP6-BMMSCs versus SAMR1-BMMSCs on BCC proliferation were examined. Intriguingly, only CM from SAMP6-BMMSCs inhibited BCC proliferation by upregulating p21 expression in both MCF-7 and MDA-MB-231 cells. These findings suggest that the senescence-associated secretory phenotype (SASP) of BMMSCs suppresses BCC viability by inducing p21, a pivotal cell cycle inhibitor and tumor suppressor. This highlights a heightened susceptibility of BCCs to dormancy in a senescent microenvironment, potentially contributing to the increased incidence of breast cancer bone metastasis and recurrence observed with aging.
Collapse
Affiliation(s)
- Meng Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi Clinical Research Center for Oral Diseases, Department of Prosthodontics, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jie-Xi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Bo Ma
- State Key Laboratory of Toxicology and Medical Countermeasures, Laboratory of Toxicant Analysis, Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Jin-Yu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Ji Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral Implantology, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Fang Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, 710032, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Cheng-Hu Hu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Hao-Kun Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, 710032, China.
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, China.
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, 710032, China.
| | - Rui Hou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Park JS, Kim DY, Hong HS. FGF2/HGF priming facilitates adipose-derived stem cell-mediated bone formation in osteoporotic defects. Heliyon 2024; 10:e24554. [PMID: 38304814 PMCID: PMC10831751 DOI: 10.1016/j.heliyon.2024.e24554] [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: 09/13/2023] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
Aims The activity of adipose-derived stem cells (ADSCs) is susceptible to the physiological conditions of the donor. Therefore, employing ADSCs from donors of advanced age or with diseases for cell therapy necessitates a strategy to enhance therapeutic efficacy before transplantation. This study aims to investigate the impact of supplementing Fibroblast Growth Factor 2 (FGF2) and Hepatocyte Growth Factor (HGF) on ADSC-mediated osteogenesis under osteoporotic conditions and to explore the underlying mechanisms of action. Main methods Adipose-derived stem cells (ADSCs) obtained from ovariectomized (OVX) rats were cultured ex vivo. These cells were cultured in an osteogenic medium supplemented with FGF2 and HGF and subsequently autologously transplanted into osteoporotic femur defects using Hydroxyapatite-Tricalcium Phosphate. The assessment of bone formation was conducted four weeks post-transplantation. Key findings Osteoporosis detrimentally affects the viability and osteogenic differentiation potential of ADSCs, often accompanied by a deficiency in FGF2 and HGF signaling. However, priming with FGF2 and HGF facilitated the formation of immature osteoblasts from OVX ADSCs in vitro, promoting the expression of osteoblastogenic proteins, including Runx-2, osterix, and ALP, during the early phase of osteogenesis. Furthermore, FGF2/HGF priming augmented the levels of VEGF and SDF-1α in the microenvironment of OVX ADSCs under osteogenic induction. Importantly, transplantation of OVX ADSCs primed with FGF2/HGF for 6 days significantly enhanced bone formation compared to non-primed cells. The success of bone regeneration was confirmed by the expression of type-1 collagen and osteocalcin in the bone tissue of the deficient area. Significance Our findings corroborate that priming with FGF2/HGF can improve the differentiation potential of ADSCs. This could be applied in autologous stem cell therapy for skeletal disease in the geriatric population.
Collapse
Affiliation(s)
- Jeong Seop Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Do Young Kim
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
| | - Hyun Sook Hong
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul, 02447, South Korea
- East-West Medical Research Institute, Kyung Hee University, Seoul, 02447, South Korea
- Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Medical Center, Seoul, 02447, South Korea
| |
Collapse
|
8
|
Wu Z, Zhu J, Wen Y, Lei P, Xie J, Shi H, Wu R, Lou X, Hu Y. Hmga1-overexpressing lentivirus protects against osteoporosis by activating the Wnt/β-catenin pathway in the osteogenic differentiation of BMSCs. FASEB J 2023; 37:e22987. [PMID: 37555233 DOI: 10.1096/fj.202300488r] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/26/2023] [Accepted: 05/09/2023] [Indexed: 08/10/2023]
Abstract
Postmenopausal osteoporosis is associated with bone formation inhibition mediated by the impaired osteogenic differentiation potential of bone marrow mesenchymal stem cells (BMSCs). However, identifying and confirming the essential genes in the osteogenic differentiation of BMSCs and osteoporosis remain challenging. The study aimed at revealing the key gene that regulated osteogenic differentiation of BMSCs and led to osteoporosis, thus exploring its therapeutic effect in osteoporosis. In the present study, six essential genes related to the osteogenic differentiation of BMSCs and osteoporosis were identified, namely, fibrillin 2 (Fbn2), leucine-rich repeat-containing 17 (Lrrc17), heat shock protein b7 (Hspb7), high mobility group AT-hook 1 (Hmga1), nexilin F-actin-binding protein (Nexn), and endothelial cell-specific molecule 1 (Esm1). Furthermore, the in vivo and in vitro experiments showed that Hmga1 expression was increased during the osteogenic differentiation of rat BMSCs, while Hmga1 expression was decreased in the bone tissue of ovariectomized (OVX) rats. Moreover, the expression of osteogenic differentiation-related genes, the activity of alkaline phosphatase (ALP), and the number of mineralized nodules were increased after Hmga1 overexpression, which was partially reversed by a Wnt signaling inhibitor (DKK1). In addition, after injecting Hmga1-overexpressing lentivirus into the bone marrow cavity of OVX rats, the bone loss, and osteogenic differentiation inhibition of BMSCs in OVX rats were partially reversed, while osteoclast differentiation promotion of BMSCs in OVX rats was unaffected. Taken together, the present study confirms that Hmga1 prevents OVX-induced bone loss by the Wnt signaling pathway and reveals that Hmga1 is a potential gene therapeutic target for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Zhixin Wu
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiayong Zhu
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yinxian Wen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Pengfei Lei
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Xie
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Haifei Shi
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ronghuan Wu
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xianfeng Lou
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yihe Hu
- Department of Orthopedic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Orthopedic Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Wang Y, Gao Y, Wang Y, Zhang H, Qin Q, Xu Z, Liu S, Wang X, Qu Y, Liu Y, Jiang X, He H. GDNF promotes the proliferation and osteogenic differentiation of jaw bone marrow mesenchymal stem cells via the Nr4a1/PI3K/Akt pathway. Cell Signal 2023:110721. [PMID: 37230200 DOI: 10.1016/j.cellsig.2023.110721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
How to efficiently regenerate jawbone defects caused by trauma, jaw osteomyelitis, tumors, or intrinsic genetic diseases is still challenging. Ectoderm-derived jawbone defect has been reported to be regenerated by selectively recruiting cells from its embryonic origin. Therefore, it is important to explore the strategy for promoting ectoderm-derived jaw bone marrow mesenchymal stem cells (JBMMSCs) on the repair of homoblastic jaw bone. Glial cell-derived neurotrophic factor (GDNF) is an important growth factor and is essential in the process of proliferation, migration and differentiation of nerve cells. However, whether GDNF promoting the function of JBMMSCs and the relative mechanism are not clear. Our results showed that activated astrocytes and GDNF were induced in the hippocampus after mandibular jaw defect. In addition, the expression of GDNF in the bone tissue around the injured area was also significantly increased after injury. Data from in vitro experiments demonstrated that GDNF could effectively promote the proliferation and osteogenic differentiation of JBMMSCs. Furthermore, when implanted in the defected jaw bone, JBMMSCs pretreated with GDNF exhibited enhanced repair effect compared with JBMMSCs without treatment. Mechanical studies found that GDNF induced the expression of Nr4a1 in JBMMSCs, activated PI3K/Akt signaling pathway and then enhanced the proliferation and osteogenic differentiation capacities of JBMMSCs. Our studies reveal that JBMMSCs are good candidates for repairing jawbone injury and pretreated with GDNF is an efficient strategy for enhancing bone regeneration.
Collapse
Affiliation(s)
- Yadi Wang
- Medical School of Chinese PLA, Beijing 100853, China; Department of periodontology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Yang Gao
- Department of orthopaedics, The Fourth Medical Center, Chinese People's Liberation Army General Hospital, Beijing 100048, China
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Xinyuan Wang
- Medical School of Chinese PLA, Beijing 100853, China; Department of periodontology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yannv Qu
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yihan Liu
- Department of periodontology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Huixia He
- Department of periodontology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
10
|
Patel D, Thankachan S, Fawaz P P A, Venkatesh T, Prasada Kabekkodu S, Suresh PS. Deciphering the role of MitomiRs in cancer: A comprehensive review. Mitochondrion 2023; 70:118-130. [PMID: 37120081 DOI: 10.1016/j.mito.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/01/2023] [Accepted: 04/23/2023] [Indexed: 05/01/2023]
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate many metabolic and signal transduction pathways. The role of miRNAs, usually found in the cytoplasm, in regulating gene expression and cancer progression has been extensively studied in the last few decades. However, very recently, miRNAs were found to localize in the mitochondria. MiRNAs that specifically localize in the mitochondria and the cytoplasmic miRNAs associated with mitochondria that directly or indirectly modulate specific mitochondrial functions are termed as "mitomiRs". Although it is not clear about the origin of mitomiRs that are situated within mitochondria (nuclear or mitochondrial origin), it is evident that they have specific functions in modulating gene expression and regulating important mitochondrial metabolic pathways. Through this review, we aim to delineate the mechanisms by which mitomiRs alter mitochondrial metabolic pathways and influence the initiation and progression of cancer. We further discuss the functions of particular mitomiRs, which have been widely studied in the context of mitochondrial metabolism and oncogenic signaling pathways. Based on the current knowledge, we can conclude that mitomiRs contribute significantly to mitochondrial function and metabolic regulation, and that dysregulation of mitomiRs can aid the proliferation of cancer cells. Therefore, the less explored area of mitomiRs' biology can be an important topic of research investigation in the future for targeting cancer cells.
Collapse
Affiliation(s)
- Dimple Patel
- School of Biotechnology, National Institute of Technology, Calicut-673601, Kerala, India
| | - Sanu Thankachan
- School of Biotechnology, National Institute of Technology, Calicut-673601, Kerala, India
| | - Abu Fawaz P P
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipa1-576104, Karnataka, India
| | - Thejaswini Venkatesh
- Dept of Biochemistry and Molecular Biology, Central University of Kerala, Kasargod, Kerala, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipa1-576104, Karnataka, India
| | - Padmanaban S Suresh
- School of Biotechnology, National Institute of Technology, Calicut-673601, Kerala, India.
| |
Collapse
|
11
|
Agostini D, Gervasi M, Ferrini F, Bartolacci A, Stranieri A, Piccoli G, Barbieri E, Sestili P, Patti A, Stocchi V, Donati Zeppa S. An Integrated Approach to Skeletal Muscle Health in Aging. Nutrients 2023; 15:nu15081802. [PMID: 37111021 PMCID: PMC10141535 DOI: 10.3390/nu15081802] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
A decline in muscle mass and function represents one of the most problematic changes associated with aging, and has dramatic effects on autonomy and quality of life. Several factors contribute to the inexorable process of sarcopenia, such as mitochondrial and autophagy dysfunction, and the lack of regeneration capacity of satellite cells. The physiologic decline in muscle mass and in motoneuron functionality associated with aging is exacerbated by the sedentary lifestyle that accompanies elderly people. Regular physical activity is beneficial to most people, but the elderly need well-designed and carefully administered training programs that improve muscle mass and, consequently, both functional ability and quality of life. Aging also causes alteration in the gut microbiota composition associated with sarcopenia, and some advances in research have elucidated that interventions via the gut microbiota-muscle axis have the potential to ameliorate the sarcopenic phenotype. Several mechanisms are involved in vitamin D muscle atrophy protection, as demonstrated by the decreased muscular function related to vitamin D deficiency. Malnutrition, chronic inflammation, vitamin deficiencies, and an imbalance in the muscle-gut axis are just a few of the factors that can lead to sarcopenia. Supplementing the diet with antioxidants, polyunsaturated fatty acids, vitamins, probiotics, prebiotics, proteins, kefir, and short-chain fatty acids could be potential nutritional therapies against sarcopenia. Finally, a personalized integrated strategy to counteract sarcopenia and maintain the health of skeletal muscles is suggested in this review.
Collapse
Affiliation(s)
- Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Marco Gervasi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessandro Stranieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanni Piccoli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Elena Barbieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Antonino Patti
- Sport and Exercise Sciences Research Unit, Department of Psychology, Educational Science and Human Movement, University of Palermo, 90128 Palermo, Italy
| | - Vilberto Stocchi
- Department of Human Science for Promotion of Quality of Life, Università Telematica San Raffaele, 00166 Rome, Italy
| | - Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| |
Collapse
|
12
|
Cao W, Yang X, Hu XH, Li J, Tian J, OuYang R, Lin X. miR-344d-3p regulates osteogenic and adipogenic differentiation of mouse mandibular bone marrow mesenchymal stem cells. PeerJ 2023; 11:e14838. [PMID: 36815989 PMCID: PMC9936866 DOI: 10.7717/peerj.14838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/10/2023] [Indexed: 02/16/2023] Open
Abstract
Postmenopausal osteoporosis (POP) is a chronic disease of bone metabolism that occurs in middle-aged and elderly women. POP can cause abnormalities of the skeletal system in the whole body, and the jaw bone is also impacted, affecting the function of the oral and maxillofacial regions. Mandibular bone marrow mesenchymal stem cells (MBMSCs) play an important role in mandibular bone metabolism, and abnormal differentiation of MBMSCs can affect the metabolic balance between new and old bone. MicroRNAs (miRNAs) can induce the differentiation of MBMSCs. In this study, the changes in biological characteristics of mandible and MBMSCs in the bone microenvironment of postmenopausal osteoporosis were firstly analyzed, and then the key miRNAs screened from miRNAs gene chips were sorted out for verification and functional exploration. It was found that miR-344d-3p promoted the osteogenic differentiation of MC3T3-E1 and MBMSCs. It inhibited the adipogenic differentiation of 3T3-L1 and MBMSCs. In addition, Dnmt3a may be the target gene of miR-344d-3p. In conclusion, this study found new biological indicators related to bone metabolism, which are of great significance in the field of bone reconstruction.
Collapse
Affiliation(s)
- Wei Cao
- Department of Prosthodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, China
| | - Xiaohong Yang
- Department of Prosthodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, China
| | - Xiao Hua Hu
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, China
| | - Jun Li
- Department of Dental Implant, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China, Zunyi, China
| | - Jia Tian
- Department of Prosthodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, China
| | - RenJun OuYang
- Department of Prosthodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, China
| | - Xue Lin
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, China
| |
Collapse
|
13
|
Wu M, Dai M, Liu X, Zeng Q, Lu Y. lncRNA SERPINB9P1 Regulates SIRT6 Mediated Osteogenic Differentiation of BMSCs via miR-545-3p. Calcif Tissue Int 2023; 112:92-102. [PMID: 36348062 DOI: 10.1007/s00223-022-01034-3] [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: 06/15/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
Evidence has shown that the altered osteogenic differentiation of human bone marrow stromal cells (BMSCs) under pathological conditions, such as osteoporosis, lead to the imbalance of bone tissue generation and destruction. Recent studies have indicated that long noncoding RNAs may play a role in regulating BMSCs osteogenic differentiation. This contributed to our impetus to move forward with the investigation of the function of lncRNA SERPINB9P1 in osteogenic differentiation of BMSCs and the potential mechanisms involved. Osteogenic differentiation of BMSCs was induced by osteogenic medium. Relative expression of lncRNA SERPINB9P1 and miR-545-3p were tested by qRT-PCR. Osteogenic mineralization was examined by Alizarin S Red staining, ALP staining, and ALP activity assay. Expression of osteoblastic markers were detected by Western blot. RNA-binding protein immunoprecipitation and dual-luciferase reporter assays were performed to test the interaction between lncRNA SERPINB9P1 and miR-545-3p. BMSCs osteogenic differentiation resulted in LncRNA SERPINB9P1 overexpression while miR-545-3p inhibition. Functional assays suggest that knockdown of lncRNA SERPINB9P1 or overexpression of miR-545-3p both inhibit BMSC osteogenic differentiation. lncRNA SERPINB9P1 was proven to regulate the osteogenic differentiation of BMSCs by altering SIRT6 expression through its suppressive effects on miR-545-3p. lncRNA SERPINB9P1 promotes osteogenic differentiation of BMSCs through the miR-545-3p/SIRT6 pathway.
Collapse
Affiliation(s)
- Min Wu
- Department of Orthopedics, The Affiliated Children's Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
- Department of Orthopedics, Jiangxi Provincial Children's Hospital, Nanchang, 330006, People's Republic of China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Xuqiang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
| | - Qunqun Zeng
- Department of Orthopedics, The Affiliated Children's Hospital of Nanchang University, Nanchang, 330006, People's Republic of China
- Department of Orthopedics, Jiangxi Provincial Children's Hospital, Nanchang, 330006, People's Republic of China
| | - Yingjie Lu
- Department of Plastic Surgery, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, 330006, People's Republic of China.
| |
Collapse
|
14
|
Jiang S, Yin C, Dang K, Zhang W, Huai Y, Qian A. Comprehensive ceRNA network for MACF1 regulates osteoblast proliferation. BMC Genomics 2022; 23:695. [PMID: 36207684 PMCID: PMC9541005 DOI: 10.1186/s12864-022-08910-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Previous studies have shown that microtubule actin crosslinking factor 1 (MACF1) can regulate osteoblast proliferation and differentiation through non-coding RNA (ncRNA) in bone-forming osteoblasts. However, the role of MACF1 in targeting the competing endogenous RNA (ceRNA) network to regulate osteoblast differentiation remains poorly understood. Here, we profiled messenger RNA (mRNA), microRNA (miRNA), and long ncRNA (lncRNA) expression in MACF1 knockdown MC3TC‑E1 pre‑osteoblast cells. RESULTS In total, 547 lncRNAs, 107 miRNAs, and 376 mRNAs were differentially expressed. Significantly altered lncRNAs, miRNAs, and mRNAs were primarily found on chromosome 2. A lncRNA-miRNA-mRNA network was constructed using a bioinformatics computational approach. The network indicated that mir-7063 and mir-7646 were the most potent ncRNA regulators and mef2c was the most potent target gene. Pathway enrichment analysis showed that the fluid shear stress and atherosclerosis, p53 signaling, and focal adhesion pathways were highly enriched and contributed to osteoblast proliferation. Importantly, the fluid shear stress and atherosclerosis pathway was co-regulated by lncRNAs and miRNAs. In this pathway, Dusp1 was regulated by AK079370, while Arhgef2 was regulated by mir-5101. Furthermore, Map3k5 was regulated by AK154638 and mir-466q simultaneously. AK003142 and mir-3082-5p as well as Ak141402 and mir-446 m-3p were identified as interacting pairs that regulate target genes. CONCLUSION This study revealed the global expression profile of ceRNAs involved in the differentiation of MC3TC‑E1 osteoblasts induced by MACF1 deletion. These results indicate that loss of MACF1 activates a comprehensive ceRNA network to regulate osteoblast proliferation.
Collapse
Affiliation(s)
- Shanfeng Jiang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Chong Yin
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Department of Clinical Laboratory, Academician (expert) workstation, Lab of epigenetics and RNA therapy, Affiliated Hospital of North Sichuan Medical College, 637000, Nanchong, China
| | - Kai Dang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Wenjuan Zhang
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Ying Huai
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China
| | - Airong Qian
- Lab for Bone Metabolism, Xi'an Key Laboratory of Special Medicine and Health Engineering, Key Lab for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
- Research Center for Special Medicine and Health Systems Engineering, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
- NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, 710072, Xi'an, Shaanxi, China.
| |
Collapse
|
15
|
Rudiansyah M, El-Sehrawy AA, Ahmad I, Terefe EM, Abdelbasset WK, Bokov DO, Salazar A, Rizaev JA, Muthanna FMS, Shalaby MN. Osteoporosis treatment by mesenchymal stromal/stem cells and their exosomes: Emphasis on signaling pathways and mechanisms. Life Sci 2022; 306:120717. [PMID: 35792178 DOI: 10.1016/j.lfs.2022.120717] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/30/2022] [Accepted: 06/09/2022] [Indexed: 02/07/2023]
Abstract
Osteoporosis is the loss of bone density, which is one of the main problems in developed and developing countries and is more common in the elderly. Because this disease is often not diagnosed until a bone fracture, it can become a life-threatening disease and cause hospitalization. With the increase of older people in a population, this disease's personal and social costs increase year by year and affect different communities. Most current treatments focus on pain relief and usually do not lead to bone tissue recovery and regeneration. But today, the use of stem cell therapy is recommended to treat and improve this disease recovery, which helps restore bone tissue by improving the imbalance in the osteoblast-osteoclast axis. Due to mesenchymal stromal/stem cells (MSCs) characteristics and their exosomes, these cells and vesicles are excellent sources for treating and preventing the progression and improvement of osteoporosis. Due to the ability of MSCs to differentiate into different cells and migrate to the site of injury, these cells are used in tissue regenerative medicine. Also, due to their contents, the exosomes of these cells help regenerate and treat various tissue injuries by affecting the injury site's cells. In this article, we attempted to review new studies in which MSCs and their exosomes were used to treat osteoporosis.
Collapse
Affiliation(s)
- Mohammad Rudiansyah
- Division of Nephrology & Hypertension, Department of Internal Medicine, Faculty of Medicine, Universitas Lambung Mangkurat/Ulin Hospital, Banjarmasin, Indonesia
| | - Amr A El-Sehrawy
- Department of Internal Medicine, Mansoura Specialized Medical Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ermias Mergia Terefe
- School of pharmacy and Health science, United States International University, Nairobi, Kenya
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia; Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Dmitry Olegovich Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, 8 Trubetskaya St., bldg. 2, Moscow 119991, Russian Federation; Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, 2/14 Ustyinsky pr., Moscow 109240, Russian Federation
| | - Aleli Salazar
- Neuroimmunology Department, National Institute of Neurology and Neurosurgery "Manuel Velasco Suárez", Mexico City, Mexico
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector of Samarkand State Medical Institute, Samarkand, Uzbekistan
| | | | - Mohammed Nader Shalaby
- Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Egypt
| |
Collapse
|
16
|
Yin J, Xiao W, Zhao Q, Sun J, Zhou W, Zhao W. MicroRNA-582-3p regulates osteoporosis through regulating homeobox A10 and osteoblast differentiation. Immunopharmacol Immunotoxicol 2022; 44:421-428. [PMID: 35285389 DOI: 10.1080/08923973.2022.2052895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Jian Yin
- Department of Orthopedic, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, 830001, PR. China
| | - Wei Xiao
- Department of Orthopedic, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, 830001, PR. China
| | - Qingbin Zhao
- Department of Orthopedic, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, 830001, PR. China
| | - Jungang Sun
- Department of Orthopedic, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, 830001, PR. China
| | - Wenzheng Zhou
- Department of Orthopedic, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, 830001, PR. China
| | - Wei Zhao
- Department of Orthopedic, Xinjiang Uygur Autonomous Region People's Hospital, Urumqi, Xinjiang, 830001, PR. China
| |
Collapse
|
17
|
Protein Expression of AEBP1, MCM4, and FABP4 Differentiate Osteogenic, Adipogenic, and Mesenchymal Stromal Stem Cells. Int J Mol Sci 2022; 23:ijms23052568. [PMID: 35269711 PMCID: PMC8910760 DOI: 10.3390/ijms23052568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem cells (MSCs) gain an increasing focus in the field of regenerative medicine due to their differentiation abilities into chondrocytes, adipocytes, and osteoblastic cells. However, it is apparent that the transformation processes are extremely complex and cause cellular heterogeneity. The study aimed to characterize differences between MSCs and cells after adipogenic (AD) or osteoblastic (OB) differentiation at the proteome level. Comparative proteomic profiling was performed using tandem mass spectrometry in data-independent acquisition mode. Proteins were quantified by deep neural networks in library-free mode and correlated to the Molecular Signature Database (MSigDB) hallmark gene set collections for functional annotation. We analyzed 4108 proteins across all samples, which revealed a distinct clustering between MSCs and cell differentiation states. Protein expression profiling identified activation of the Peroxisome proliferator-activated receptors (PPARs) signaling pathway after AD. In addition, two distinct protein marker panels could be defined for osteoblastic and adipocytic cell lineages. Hereby, overexpression of AEBP1 and MCM4 for OB as well as of FABP4 for AD was detected as the most promising molecular markers. Combination of deep neural network and machine-learning algorithms with data-independent mass spectrometry distinguish MSCs and cell lineages after adipogenic or osteoblastic differentiation. We identified specific proteins as the molecular basis for bone formation, which could be used for regenerative medicine in the future.
Collapse
|
18
|
Su X, Zhang H, Lei F, Wang R, Lin T, Liao L. Epigenetic therapy attenuates oxidative stress in BMSCs during ageing. J Cell Mol Med 2021; 26:375-384. [PMID: 34874118 PMCID: PMC8743666 DOI: 10.1111/jcmm.17089] [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: 03/31/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023] Open
Abstract
Oxidative stress, a hallmark of ageing, inhibits the osteogenic differentiation of bone marrow-derived mesenchymal stem cells in long bone. The dysfunction of the cellular antioxidant defence system is a critical cause of oxidative stress, but the mechanism of the decline of antioxidant defence in senescent stem cells remains elusive. Here, we found that EZH2, an epigenetic regulator of histone methylation, acted as a suppressor of the antioxidative defence system in BMSCs from the femur. The increased EZH2 led to a decrease in the levels of antioxidant enzymes and exaggerated oxidative damage in aged BMSCs, resulting in the defect of bone formation and regeneration. Mechanistically, EZH2 enhanced the modification of H3K27me3 on the promoter of Foxo1 and suppressed its function to activate the downstream genes in antioxidant defence. Moreover, epigenetic therapy targeting EZH2-mediated H3K27me3 modification largely recovered the antioxidant defence in BMSCs and attenuate oxidative damage, leading to the recovery of the osteogenesis in old BMSCs. Taken together, our findings revealed novel crosstalk between histone epigenetic modification and oxidative stress during stem cell ageing, suggesting a possibility of epigenetic therapy in the recovery of BMSCs senescence and treatment of age-related bone disease.
Collapse
Affiliation(s)
- Xiaoxia Su
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry & Engineering Research Center of Oral Translational Medicine & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haoyu Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry & Engineering Research Center of Oral Translational Medicine & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fengzhen Lei
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Rui Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Tingting Lin
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Li Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry & Engineering Research Center of Oral Translational Medicine & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Yu L, Xie M, Zhang F, Wan C, Yao X. TM9SF4 is a novel regulator in lineage commitment of bone marrow mesenchymal stem cells to either osteoblasts or adipocytes. Stem Cell Res Ther 2021; 12:573. [PMID: 34774100 PMCID: PMC8590266 DOI: 10.1186/s13287-021-02636-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/25/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoporosis is a common bone disease in elderly population caused by imbalanced bone formation and bone resorption. Mesenchymal stem cells (MSCs) are responsible for maintaining this bone homeostasis. The phenotype of transmembrane 9 superfamily 4 (TM9SF4) knockout mice suggests a relationship between TM9SF4 proteins and bone homeostasis. But the effect of TM9SF4 in osteology has never been reported. In the present study, we investigated the function of TM9SF4 in MSC differentiation commitment, as well as its role in osteoporosis. METHODS Primary bone marrow MSCs, isolated from TM9SF4 wildtype (TM9SF4+/+) and knockout (TM9SF4-/-) mice, were induced to differentiate into osteoblasts or adipocytes, respectively. The osteogenesis was examined by qRT-PCR detection of osteogenic markers, ALP staining and Alizarin Red S staining. The adipogenesis was tested by qRT-PCR quantification of adipogenic markers and Oil Red O staining. The cytoskeletal organization of MSCs was observed under confocal microscope. The osteoporotic model was induced by ovariectomy in TM9SF4+/+ and TM9SF4-/- mice, followed by Toluidine blue and H&E staining to assess lipid accumulation in trabecular bones, as well as micro-computed tomography scanning and immunohistochemistry staining for bone mass density assessment. The experiments on signaling pathways were conducted using qRT-PCR, Western blot and Alizarin Red S staining. RESULTS We determined the role of TM9SF4 in MSC differentiation and found that TM9SF4-/- MSCs had higher potential to differentiate into osteoblasts and lower capability into adipocytes, without affecting osteoclastogenesis in vitro. In ovariectomy-induced osteoporotic model, TM9SF4-/- mice retained higher bone mass and less lipid accumulation in trabecular bones, indicating an important role of TM9SF4 in the regulation of osteoporosis. Mechanistically, TM9SF4-depleted cells showed elongated actin fibers, which may act through mTORC2/Akt/β-catenin pathway to promote their commitment into osteoblasts. Furthermore, TM9SF4-depleted cells showed higher activity of canonical Wnt pathway, suggesting the participation of Wnt/β-catenin during TM9SF4-regulated osteogenesis. CONCLUSIONS Our study demonstrates TM9SF4 as a novel regulator for MSC lineage commitment. Depletion of TM9SF4 preferentially drives MSCs into osteoblasts instead of adipocytes. Furthermore, TM9SF4-/- mice show delayed bone loss and reduced lipid accumulation during ovariectomy-induced osteoporosis. Our results indicate TM9SF4 as a promising target for the future clinical osteoporotic treatment.
Collapse
Affiliation(s)
- Libo Yu
- School of Biomedical Sciences, Heart and Vascular Institute and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China.,Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
| | - Mingxu Xie
- School of Biomedical Sciences, Heart and Vascular Institute and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China
| | - Fengjie Zhang
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Chao Wan
- MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, Heart and Vascular Institute and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China. .,Centre for Cell and Developmental Biology, State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China.
| |
Collapse
|
20
|
Echave M, Erezuma I, Golafshan N, Castilho M, Kadumudi F, Pimenta-Lopes C, Ventura F, Pujol A, Jimenez J, Camara J, Hernáez-Moya R, Iturriaga L, Sáenz Del Burgo L, Iloro I, Azkargorta M, Elortza F, Lakshminarayanan R, Al-Tel T, García-García P, Reyes R, Delgado A, Évora C, Pedraz J, Dolatshahi-Pirouz A, Orive G. Bioinspired gelatin/bioceramic composites loaded with bone morphogenetic protein-2 (BMP-2) promote osteoporotic bone repair. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112539. [DOI: 10.1016/j.msec.2021.112539] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 12/17/2022]
|
21
|
Wang Z, Li L, Gu W, Mao Y, Wang T. Resveratrol Reverses Osteogenic Decline of Bone Marrow Mesenchymal Stem Cells Via Upregulation of Yes-Associated Protein Expression in Inflammatory Environment. Stem Cells Dev 2021; 30:1202-1214. [PMID: 34598655 DOI: 10.1089/scd.2021.0195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteoporosis is an age-related bone disease, characterized by rapid boneloss, decreased bone mineral density (BMD), and consequent risk of fractures. The most prevalent form of clinically significant osteoporosis involves various inflammatory conditions, especially age-dependent osteoporosis and postmenopausal osteoporosis. Tumor necrosis factor-α (TNF-α), a pro-inflammatory cytokine, plays a critical role in the development of inflammatory, which also plays an important role in bone formation and bone loss during osteoporosis. In this report, we examined the effect of TNF-α on osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and its modulation by resveratrol (Res). We found that TNF-α can upregulate inflammatory cytokines, Il-6, Mmp-9, and Il-1β, and establish an inflammatory environment. High inflammatory cytokine expression significantly inhibited osteogenic differentiation of BMSCs by overactivating upstream Hippo kinases and decreasing the nuclear Yes-associated protein (YAP) signals. With Res treatment, decreasing inflammatory cytokine expression normalized Hippo/YAP signaling and effectively rescued YAP-mediated osteogenesis. Thus, through these studies, we present a mechanism by which TNF-α can affect BMSCs osteogenesis through modulation of Hippo/YAP signaling.
Collapse
Affiliation(s)
- Zihan Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China
| | - Le Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Wenwen Gu
- Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yuqi Mao
- Cardiovascular Disease Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Tao Wang
- Cardiovascular Disease Department, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
22
|
Zhou B, Peng K, Wang G, Chen W, Kang Y. Silencing Proteasome 26S Subunit ATPase 2 (PSMC2) Protects the Osteogenic Differentiation In Vitro and Osteogenesis In Vivo. Calcif Tissue Int 2021; 109:44-54. [PMID: 33625534 DOI: 10.1007/s00223-021-00819-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/30/2021] [Indexed: 12/17/2022]
Abstract
Osteoporosis is a commonly seen degenerative bone disorder in the elderly and postmenopausal women, with a low bone mineral density as a major risk factor. The osteogenic potential of bone marrow stromal cells (BMSCs) showed to be impaired during osteoporosis. We established a postmenopausal osteoporosis model in ovariectomized (OVX) mice and found the upregulation of proteasome 26S subunit ATPase 2 (PSMC2) in OVX mice. PSMC2 silencing improved OVX-impaired biomechanical properties of mice femur, OVX-decreased BMD, and OVX-destroyed bone structure. Histopathological analysis indicated that PSMC2 silencing improved bone trabecular structure and increased the contents of collagen fibers and newly formed bone or cartilage in OVX mice. In the meantime, PSMC2 silencing increased Runx2, PI3K, Wnt3a, and β-catenin protein contents while reduced CTSK protein. Within BMSCs isolated from OVX mice, PSMC2 silencing promoted BMSC osteogenic differentiation and elevated osteogenic markers' protein contents, including HOXA10, Runx2, OCN, OPN, and COL1A2. In conclusion, PSMC2 expression is upregulated in the postmenopausal osteoporosis model in OVX mice. PSMC2 silencing promotes the osteogenic differentiation of BMSCs in vitro, promotes bone formation, and inhibits bone resorption in vivo.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Kun Peng
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Guoqiang Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Weihua Chen
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yijun Kang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
23
|
Yu H, Wang K, Liu P, Luo P, Zhu D, Yin J, Yang Q, Huang Y, Gao J, Ai Z, Chen Y, Gao Y. miR-4286 functions in osteogenesis and angiogenesis via targeting histone deacetylase 3 and alleviates alcohol-induced bone loss in mice. Cell Prolif 2021; 54:e13054. [PMID: 33973278 PMCID: PMC8168416 DOI: 10.1111/cpr.13054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/12/2021] [Accepted: 04/17/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives Alcohol consumption is one of the leading factors contributing to premature osteopenia. MicroRNA (miRNA) coordinates a cascade of anabolic and catabolic processes in bone homeostasis and dynamic vascularization. The aim was to investigate the protective role of miR‐4286 in alcohol‐induced bone loss and its mechanism. Materials and Methods The effect of miR‐4286 and alcohol on bone mesenchymal stem cells (BMSCs) and human umbilical vein endothelial cells (HUVECs) was explored via multiple in vitro assays, including cell proliferation, QPCR, Western blot, osteogenesis, angiogenesis etc miR‐4286 directly regulated HDAC3 was investigated by luciferase reporter assay, and the function of HDAC3 was also explored in vitro. Moreover, alcohol‐induced bone loss in mice was established to reveal the preventive effect of miR‐4286 by radiographical and histopathological assays. Results In vitro, ethanol dramatically inhibited the proliferation and osteogenesis of BMSCs, and substantially impaired the proliferation and vasculogenesis of HUVECs. However, a forced overexpression of miR‐4286 within BMSCs and HUVECs could largely abolish inhibitory effects by alcohol. Furthermore, alcohol‐induced inhibition on osteogenic and vasculogenic functions was mediated by histone deacetylase 3 (HDAC3), and dual‐luciferase reporter assay showed that HDAC3 was the direct binding target of miR‐4286. In vivo, micro‐CT scanning and histology assessment revealed that miR‐4286 could prevent alcohol‐induced bone loss. Conclusions We firstly demonstrated that miR‐4286 might function via intimate osteogenesis‐angiogenesis pathway to alleviate alcohol‐induced osteopenia via targeting HDAC3.
Collapse
Affiliation(s)
- Hongping Yu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Kaiyang Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Pei Liu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Pengbo Luo
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Daoyu Zhu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Junhui Yin
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qianhao Yang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yigang Huang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Junjie Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zisheng Ai
- Department of Medical Statistics, Tongji University School of Medicine, Shanghai, China
| | - Yixuan Chen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
24
|
Zhang S, Ding X, Miao H, Wang L, Xian L, Han S, Zhang D, Li J. The Effects of MiR-320 on the Proliferation and Differentiation of Human Alveolar Bone-Derived Mesenchymal Stem Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alveolar bone-derived mesenchymal stem cells (AB-BMSCs) have a biological morphology and antigen phenotype similar to those of BMSCs. However, the intrinsic characteristics of AB-BMSCs and their underlying mechanisms, in which the involvement of micro(mi)RNAs has been reported, remain
unknown. This study shows that miR-320c expression was significantly suppressed during osteoblastic differentiation of human AB-BMSCs. The overexpression of miR-320c markedly decreased cellular proliferation, intracellular activity of alkaline phosphatase (ALP) and formation of calcium nodules;
mRNA levels of osteogenesis-related genes were significantly reduced compared to those in control cells. Calcium nodule formation in miR-320c-knockdown cells was significantly increased, and HOXA10, Runx2, and BGP mRNA levels were significantly increased compared to those in
control cells. These results indicate that miR-320c suppresss the proliferation and osteogenic differentiation of AB-BMSCs, in part by decreasing ALP activity, cellular proliferation, mineralization, and expression of several osteogenesis-related genes. These results lay the basic foundation
for the elucidation of the molecular mechanisms of alveolar bone reconstruction.
Collapse
Affiliation(s)
- Shuyue Zhang
- Department of Stomatology, Tangshan People’s Hospital, Tangshan, 063001, China
| | - Xinguo Ding
- Xiamen Haicang Hospital, Xiamen, 361026, China
| | - Haixia Miao
- Department of Stomatology, Tangshan People’s Hospital, Tangshan, 063001, China
| | - Lei Wang
- Department of Pathology, Tangshan People’s Hospital, Tangshan, 063001, China
| | - Lige Xian
- Department of Pathology, Tangshan People’s Hospital, Tangshan, 063001, China
| | - Sugui Han
- ClinicalLaboratory, Tangshan People’s Hospital, Tangshan, 063001, China
| | - Di Zhang
- North China University of Science and Technology, Tangshan, 063000, China
| | - Jian Li
- Department of Stomatology, Xiang’an Hospital of Xiamen University, Xiamen, 361101, China
| |
Collapse
|
25
|
Yu Y, Chen M, Yang S, Shao B, Chen L, Dou L, Gao J, Yang D. Osthole enhances the immunosuppressive effects of bone marrow-derived mesenchymal stem cells by promoting the Fas/FasL system. J Cell Mol Med 2021; 25:4835-4845. [PMID: 33749126 PMCID: PMC8107110 DOI: 10.1111/jcmm.16459] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/12/2021] [Accepted: 02/24/2021] [Indexed: 12/20/2022] Open
Abstract
Thanks to the advantages of easy harvesting and escape from immune rejection, autologous bone marrow-derived mesenchymal stem cells (BMSCs) are promising candidates for immunosuppressive therapy against inflammation and autoimmune diseases. However, the therapy is still challenging because the immunomodulatory properties of BMSCs are always impaired by immunopathogenesis in patients. Because of its reliable and extensive biological activities, osthole has received increased clinical attention. In this study, we found that BMSCs derived from osteoporosis donors were ineffective in cell therapy for experimental inflammatory colitis and osteoporosis. In vivo and in vitro tests showed that because of the down-regulation of Fas and FasL expression, the ability of osteoporotic BMSCs to induce T-cell apoptosis decreased. Through the application of osthole, we successfully restored the immunosuppressive ability of osteoporotic BMSCs and improved their treatment efficacy in experimental inflammatory colitis and osteoporosis. In addition, we found the immunomodulatory properties of BMSCs were enhanced after osthole pre-treatment. In this study, our data highlight a new approach of pharmacological modification (ie osthole) to improve the immune regulatory performance of BMSCs from a healthy or inflammatory microenvironment. The development of targeted strategies to enhance immunosuppressive therapy using BMSCs may be significantly improved by these findings.
Collapse
Affiliation(s)
- Yang Yu
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Meng Chen
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Shiyao Yang
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Bingyi Shao
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Liang Chen
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Lei Dou
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Jing Gao
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| | - Deqin Yang
- Northern Department of EndodonticsStomatological Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing Key Laboratory of Oral Diseases and Biomedical SciencesChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
| |
Collapse
|
26
|
Liu F, Yuan Y, Bai L, Yuan L, Li L, Liu J, Chen Y, Lu Y, Cheng J, Zhang J. LRRc17 controls BMSC senescence via mitophagy and inhibits the therapeutic effect of BMSCs on ovariectomy-induced bone loss. Redox Biol 2021; 43:101963. [PMID: 33865167 PMCID: PMC8066428 DOI: 10.1016/j.redox.2021.101963] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/06/2021] [Accepted: 03/27/2021] [Indexed: 02/08/2023] Open
Abstract
Senescence of bone marrow-derived mesenchymal stem cells (BMSCs) has been widely reported to be closely correlated with aging-related diseases, including osteoporosis (OP). Moreover, the beneficial functions of BMSCs decline with age, limiting their therapeutic efficacy in OP. In the present study, using RNA sequencing (RNA-Seq), we found that leucine-rich repeat containing 17 (LRRc17) expression in BMSCs was highly positively correlated with age. Therefore, we investigated whether LRRc17 knockdown could rejuvenate aged MSCs and increase their therapeutic efficacy in OP. Consistent with the RNA-Seq results, the protein expression of LRRc17 in senescent BMSCs was significantly increased, whereas LRRc17 knockdown inhibited cell apoptosis and reduced the expression of age-related proteins and G2 and S phase quiescence. Furthermore, LRRc17 knockdown shifted BMSCs from adipogenic to osteogenic differentiation, indicating the critical role of LRRc17 in BMSC senescence and differentiation. Additionally, similar to rapamycin (RAPA) treatment, LRRc17 knockdown activated mitophagy via inhibition of the mTOR/PI3K pathway, which consequently reduced mitochondrial dysfunction and inhibited BMSC senescence. However, the effects of LRRc17 knockdown were significantly blocked by the autophagy inhibitor hydroxychloroquine (HCQ), demonstrating that LRRc17 knockdown prevented BMSC senescence by activating mitophagy. In vivo, compared with untransfected aged mouse-derived BMSCs (O-BMSCs), O-BMSCs transfected with sh-LRRc17 showed effective amelioration of ovariectomy (OVX)-induced bone loss. Collectively, these results indicated that LRRc17 knockdown rejuvenated senescent BMSCs and thus enhanced their therapeutic efficacy in OP by activating autophagy.
Collapse
Affiliation(s)
- Fei Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Lin Bai
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China; Core Facility of West China Hospital, Sichuan University, Chengdu, PR China
| | - Longhui Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China.
| | - Jie Zhang
- Key Laboratory of Transplant Engineering and Immunology, NHFPC; Regenerative Medicine Research Center; National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, PR China.
| |
Collapse
|
27
|
Li R, Dong Y, Li F. ETS Proto-Oncogene 1 Suppresses MicroRNA-128 Transcription to Promote Osteogenic Differentiation Through the HOXA13/β-Catenin Axis. Front Physiol 2021; 12:626248. [PMID: 33746773 PMCID: PMC7965964 DOI: 10.3389/fphys.2021.626248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/09/2021] [Indexed: 12/23/2022] Open
Abstract
ETS proto-oncogene 1 (ETS1) has been implicated in osteoporosis (OP), but the exact molecular mechanisms are complex. This work focuses on the impact of ETS1 on the osteogenic differentiation and the molecules involved. A mouse pre-osteoblast cell line MC3T3-E1 was used for in vitro experiments. ETS1 was upregulated during the process of osteogenic differentiation of MC3T3-E1 cells. Overexpression of ETS1 promoted expression of osteogenic markers, alkaline phosphate concentration, and calcareous accumulation in cells. ETS1 was found to specifically bind to miR-128 promoter to suppress its transcription, while miR-128 could target homeobox A13 (HOXA13). Therefore, ETS1 suppressed miR-128 transcription to upregulate HOXA13 expression. Overexpression of HOXA13 promoted the osteogenic differentiation ability of cells and increased the protein level of β-catenin. Either overexpression of miR-128 or downregulation of β-catenin by CWP232228, a β-catenin-specific antagonist, blocked the promoting roles of ETS1 in cells. To conclude, this study provided evidence that ETS1 suppresses miR-128 transcription to activate the following HOXA13/β-catenin axis, therefore promoting osteogenic differentiation ability of MC3T3-E1 cells. This finding may offer novel ideas for OP treatment.
Collapse
Affiliation(s)
- Renyao Li
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Ying Dong
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Feipeng Li
- Naton Biotech (Beijing) Co., Ltd., Beijing, China
| |
Collapse
|
28
|
Photobiomodulation: An Effective Approach to Enhance Proliferation and Differentiation of Adipose-Derived Stem Cells into Osteoblasts. Stem Cells Int 2021; 2021:8843179. [PMID: 33833810 PMCID: PMC8012132 DOI: 10.1155/2021/8843179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 01/07/2023] Open
Abstract
Osteoporosis is regarded as the most common chronic metabolic bone condition in humans. In osteoporosis, bone mesenchymal stem cells (MSCs) have reduced cellular function. Regenerative medicine using adipose-derived stem cell (ADSC) transplantation can promote the growth and strength of new bones, improve bone stability, and reduce the risk of fractures. Various methods have been attempted to differentiate ADSCs to functioning specialized cells for prospective clinical application. However, commonly used therapies have resulted in damage to the donor site and morbidity, immune reactions, carcinogenic generation, and postoperative difficulties. Photobiomodulation (PBM) improves ADSC differentiation and proliferation along with reducing clinical difficulties such as treatment failures to common drug therapies and late initiation of treatment. PBM is a noninvasive, nonthermal treatment that encourages cells to produce more energy and to undergo self-repair by using visible green and red and invisible near-infrared (NIR) radiation. The use of PBM for ADSC proliferation and differentiation has been widely studied with multiple outcomes observed due to laser fluence and wavelength dependence. In this article, the potential for differentiating ADSCs into osteoblasts and the various methods used, including biological induction, chemical induction, and PBM, will be addressed. Likewise, the optimal laser parameters that could improve the proliferation and differentiation of ADSC, translating into clinical success, will be commented on.
Collapse
|
29
|
Involvement of the long noncoding RNA H19 in osteogenic differentiation and bone regeneration. Stem Cell Res Ther 2021; 12:74. [PMID: 33478579 PMCID: PMC7819155 DOI: 10.1186/s13287-021-02149-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
Osteogenic differentiation and bone regeneration are complex processes involving multiple genes and multiple steps. In this review, we summarize the effects of the long noncoding RNA (lncRNA) H19 on osteogenic differentiation. Osteogenic differentiation includes matrix secretion and calcium mineralization as hallmarks of osteoblast differentiation and the absorption of calcium and phosphorus as hallmarks of osteoclast differentiation. Mesenchymal stem cells (MSCs) form osteoprogenitor cells, pre-osteoblasts, mature osteoblasts, and osteocytes through induction and differentiation. lncRNAs regulate the expression of coding genes and play essential roles in osteogenic differentiation and bone regeneration. The lncRNA H19 is known to have vital roles in osteogenic induction. This review highlights the role of H19 as a novel target for osteogenic differentiation and the promotion of bone regeneration.
Collapse
|
30
|
Shang F, Yu Y, Liu S, Ming L, Zhang Y, Zhou Z, Zhao J, Jin Y. Advancing application of mesenchymal stem cell-based bone tissue regeneration. Bioact Mater 2020; 6:666-683. [PMID: 33005830 PMCID: PMC7509590 DOI: 10.1016/j.bioactmat.2020.08.014] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/07/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022] Open
Abstract
Reconstruction of bone defects, especially the critical-sized defects, with mechanical integrity to the skeleton is important for a patient's rehabilitation, however, it still remains challenge. Utilizing biomaterials of human origin bone tissue for therapeutic purposes has provided a facilitated approach that closely mimics the critical aspects of natural bone tissue with regard to its properties. However, not only efficacious and safe but also cost-effective and convenient are important for regenerative biomaterials to achieve clinical translation and commercial success. Advances in our understanding of regenerative biomaterials and their roles in new bone formation potentially opened a new frontier in the fast-growing field of regenerative medicine. Taking inspiration from the role and multicomponent construction of native extracellular matrix (ECM) for cell accommodation, the ECM-mimicking biomaterials and the naturally decellularized ECM scaffolds were used to create new tissues for bone restoration. On the other hand, with the going deep in understanding of mesenchymal stem cells (MSCs), they have shown great promise to jumpstart and facilitate bone healing even in diseased microenvironments with pharmacology-based endogenous MSCs rescue/mobilization, systemic/local infusion of MSCs for cytotherapy, biomaterials-based approaches, cell-sheets/-aggregates technology and usage of subcellular vesicles of MSCs to achieve scaffolds-free or cell-free delivery system, all of them have been shown can improve MSCs-mediated regeneration in preclinical studies and several clinical trials. Here, following an overview discussed autogenous/allogenic and ECM-based bone biomaterials for reconstructive surgery and applications of MSCs-mediated bone healing and tissue engineering to further offer principles and effective strategies to optimize MSCs-based bone regeneration. Focusing on MSCs based bone regeneration. Discussed cytotherapy, cell-free therapies and cell-aggregates technology in detail. Stating the approaches of MSCs in diseased microenvironments.
Collapse
Affiliation(s)
- Fengqing Shang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Stomatology, The 306th Hospital of PLA, Beijing, 100101, China
| | - Yang Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, 250012, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Leiguo Ming
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yongjie Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, 850000, China
| | - Jiayu Zhao
- Bureau of Service for Veteran Cadres of PLA in Beijing, Beijing, 100001, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Corresponding author.
| |
Collapse
|
31
|
Epigenetic Regulation in Mesenchymal Stem Cell Aging and Differentiation and Osteoporosis. Stem Cells Int 2020; 2020:8836258. [PMID: 32963550 PMCID: PMC7501554 DOI: 10.1155/2020/8836258] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a reliable source for cell-based regenerative medicine owing to their multipotency and biological functions. However, aging-induced systemic homeostasis disorders in vivo and cell culture passaging in vitro induce a functional decline of MSCs, switching MSCs to a senescent status with impaired self-renewal capacity and biased differentiation tendency. MSC functional decline accounts for the pathogenesis of many diseases and, more importantly, limits the large-scale applications of MSCs in regenerative medicine. Growing evidence implies that epigenetic mechanisms are a critical regulator of the differentiation programs for cell fate and are subject to changes during aging. Thus, we here review epigenetic dysregulations that contribute to MSC aging and osteoporosis. Comprehending detailed epigenetic mechanisms could provide us with a novel horizon for dissecting MSC-related pathogenesis and further optimizing MSC-mediated regenerative therapies.
Collapse
|
32
|
Xu Z, He J, Zhou X, Zhang Y, Huang Y, Xu N, Yang H. Down-regulation of LECT2 promotes osteogenic differentiation of MSCs via activating Wnt/β-catenin pathway. Biomed Pharmacother 2020; 130:110593. [PMID: 32763823 DOI: 10.1016/j.biopha.2020.110593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/21/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022] Open
Abstract
Osteoporosis is a result of the imbalance between osteoblasts and osteoclasts quantities, which is closely correlated with osteogenic differentiation (OD). Leucocyte cell-derived chemotaxin 2 (LECT2) has been reported as a regulatory factor in some chronic diseases such as hepatitis through mediating downstream target gene β-catenin. Additionally, Wnt/β-catenin is also the crucial modulatory signal pathway in OD. Mesenchymal stem cells (MSC) is a kind of mesodermal stem cells; its differentiation direction is discovered affected by Wnt/β-catenin. However, the function of LECT2 in osteoporosis still remains exploration, which encourages us to lucubrate its functional effect in regulating the OD of MSCs. In this study, we found that LECT2 was expressed at low level in MSCs with osteogenic differentiation, and knockdown of LECT2 would activate Wnt/β-catenin pathway and therefore promoting OD in MSCs. It is the first time to report that LECT2 participates in regulating OD via mediating Wnt/β-catenin. Our discovery would affirmatively help provide a novel strategy for the diagnosis and therapy methods for osteoporosis.
Collapse
Affiliation(s)
- Zhonghua Xu
- Department of Orthopedics, Jintan Hospital Affiliated to Jiangsu University, Changzhou, 213200, China
| | - Jin He
- Department of Orthopedics, Jintan Hospital Affiliated to Jiangsu University, Changzhou, 213200, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China.
| | - Yi Zhang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Yong Huang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Nanwei Xu
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213003, China
| | - Haoyu Yang
- Department of Orthopedics, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, 214000, China.
| |
Collapse
|
33
|
Gu H, Shi S, Xiao F, Huang Z, Xu J, Chen G, Zhou K, Lu L, Yin X. MiR-1-3p regulates the differentiation of mesenchymal stem cells to prevent osteoporosis by targeting secreted frizzled-related protein 1. Bone 2020; 137:115444. [PMID: 32447074 DOI: 10.1016/j.bone.2020.115444] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/11/2023]
Abstract
Osteoporosis (OP) is a systemic skeletal disorder with the characteristics of bone mass reduction and microarchitecture deterioration, resulting in bone fragility and increased fracture risk. A reduction in the osteoblast-differentiation of bone marrow mesenchymal stem cells (BMSCs) is considered as a basic pathogenesis of osteoporosis. miRNAs play a substantial role in the development and differentiation of BMSCs. In the present study, we found that miR-1-3p was significantly downregulated in the bones of Chinese osteoporotic patients (n = 29). Secreted frizzled-related protein 1 (SFRP1) was predicted as a target gene of miR-1-3p via the TargetScan and PicTar softwares and validated by dual-luciferase reporter assays. The findings revealed that the expression of SFRP1 was inversely correlated with miR-1-3p in osteoporotic patients. We induced mouse MSCs (mMSCs) to osteogenesis or adipogenesis and found that miR-1-3p was upregulated during osteogenesis but downregulated during adipogenesis. The overexpression of miR-1-3p stimulated osteogenesis and inhibited adipogenesis of mMSCs. In addition, ovariectomized (OVX) mice were tested and the function of miR-1-3p in vivo was explored. Immunohistochemistry and histomorphometric assays showed that in vivo inhibition of miR-1-3p increased the expression level of SFRP1 and reduced bone formation and bone mass. Furthermore, tartrate-resistant acid phosphatase (TRAP) staining indicated that the in vivo suppression of miR-1-3p promoted osteoclast activity, suggesting that miR-1-3p may influence bone mass by regulating bone resorption. It can be concluded that miR-1-3p plays a pivotal role in the pathogenesis of osteoporosis via targeting SFRP1 and may be a potential therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Si Shi
- Department of Biochemistry and Molecular Biology, School of medicine, Tongji University, 1239 Siping Road, Shanghai 200092, PR China
| | - Fangzhu Xiao
- Department of Orthopedics, The Fifth Hospital of Xiamen, 101 Min 'an Road, Maxiang Town, Xiang 'an District, Xiamen, Fujian Province, 361101, PR China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Guangnan Chen
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China
| | - Lixia Lu
- Department of Biochemistry and Molecular Biology, School of medicine, Tongji University, 1239 Siping Road, Shanghai 200092, PR China.
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Hospital, Fudan University, 170 Xin Song Road, Shanghai 201199, PR China.
| |
Collapse
|
34
|
Castaño IM, Raftery RM, Chen G, Cavanagh B, Quinn B, Duffy GP, O'Brien FJ, Curtin CM. Rapid bone repair with the recruitment of CD206 +M2-like macrophages using non-viral scaffold-mediated miR-133a inhibition of host cells. Acta Biomater 2020; 109:267-279. [PMID: 32251781 DOI: 10.1016/j.actbio.2020.03.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 01/01/2023]
Abstract
microRNAs offer vast therapeutic potential for multiple disciplines. From a bone perspective, inhibition of miR-133a may offer potential to enhance Runx2 activity and increase bone repair. This study aims to assess the therapeutic capability of antagomiR-133a delivery from collagen-nanohydroxyapatite (coll-nHA) scaffolds following cell-free implantation in rat calvarial defects (7 mm diameter). This is, to the best of our knowledge, the first report of successful in vivo antagomiR uptake in host cells of fully immunocompetent animals without distribution to other off-target tissues. Our results demonstrate the localized release of antagomiR-133a to the implant site at 1 week post-implantation with increased calcium deposits already evident in the antagomiR-133a loaded scaffolds at this early timepoint. This was followed by an approximate 2-fold increase in bone volume versus antagomiR-free scaffolds and a significant 10-fold increase over the empty defect controls, after just 4 weeks. An increase in host CD206+ cells suggests an accelerated pro-remodeling response by M2-like macrophages accompanying bone repair with this treatment. Overall, this non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the addition of exogenous cells - and underlines the role of M2 macrophage-like cells in directing accelerated bone repair. Expanding the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a variety of therapeutic indications. STATEMENT OF SIGNIFICANCE: microRNAs, small non-coding RNA molecules involved in gene regulation, may have potential as a new class of bone healing therapeutics as they can enhance the regenerative capacity of bone-forming cells. We developed a collagen-nanohydroxyapatite-microRNA scaffold system to investigate whether miR133a inhibition can enhance osteogenesis in rat MSCs and ultimately accelerate endogenous bone repair by host cells in vivo without pre-seeding cells prior to implantation. Overall, this off-the-shelf, non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the requirement of exogenous cells - and highlights the role of CD206+M2 macrophage-like cells in guiding accelerated bone repair. Translating the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a vast myriad of therapeutic indications.
Collapse
|
35
|
Wang K, Wang Y, Hu Z, Zhang L, Li G, Dang L, Tan Y, Cao X, Shi F, Zhang S, Zhang G. Bone-targeted lncRNA OGRU alleviates unloading-induced bone loss via miR-320-3p/Hoxa10 axis. Cell Death Dis 2020; 11:382. [PMID: 32427900 PMCID: PMC7237470 DOI: 10.1038/s41419-020-2574-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 01/13/2023]
Abstract
Unloading-induced bone loss is a threat to human health and can eventually result in osteoporotic fractures. Although the underlying molecular mechanism of unloading-induced bone loss has been broadly elucidated, the pathophysiological role of long noncoding RNAs (lncRNAs) in this process is unknown. Here, we identified a novel lncRNA, OGRU, a 1816-nucleotide transcript with significantly decreased levels in bone specimens from hindlimb-unloaded mice and in MC3T3-E1 cells under clinorotation-unloading conditions. OGRU overexpression promoted osteoblast activity and matrix mineralization under normal loading conditions, and attenuated the suppression of MC3T3-E1 cell differentiation induced by clinorotation unloading. Furthermore, this study found that supplementation of pcDNA3.1(+)–OGRU via (DSS)6–liposome delivery to the bone-formation surfaces of hindlimb-unloaded (HLU) mice partially alleviated unloading-induced bone loss. Mechanistic investigations demonstrated that OGRU functions as a competing endogenous RNA (ceRNA) to facilitate the protein expression of Hoxa10 by competitively binding miR-320-3p and subsequently promote osteoblast differentiation and bone formation. Taken together, the results of our study provide the first clarification of the role of lncRNA OGRU in unloading-induced bone loss through the miR-320-3p/Hoxa10 axis, suggesting an efficient anabolic strategy for osteoporosis treatment.
Collapse
Affiliation(s)
- Ke Wang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Yixuan Wang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Zebing Hu
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Lijun Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Gaozhi Li
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Lei Dang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xinsheng Cao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China
| | - Fei Shi
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China.
| | - Shu Zhang
- The Key Laboratory of Aerospace Medicine, Ministry of Education, Air Force Medical University, 710032, Xi'an, Shaanxi, China.
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
36
|
Lotz EM, Cohen DJ, Schwartz Z, Boyan BD. Titanium implant surface properties enhance osseointegration in ovariectomy induced osteoporotic rats without pharmacologic intervention. Clin Oral Implants Res 2020; 31:374-387. [PMID: 31953969 PMCID: PMC7771214 DOI: 10.1111/clr.13575] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/03/2019] [Accepted: 01/04/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVES This study determined whether implant surfaces that promote osseointegration in normal rats can promote osseointegration in osteoporotic rats without pharmacologic intervention. MATERIALS AND METHODS Virgin female 8-month-old CD Sprague Dawley rats (N = 25) were ovariectomized. At 6 weeks, microstructured/non-nanostructured/hydrophobic, microstructured/nanostructured/hydrophobic, or microstructured/nanostructured/hydrophilic Ti implants (Ø2.5 × 3.5 mm; Institut Straumann AG, Basel, Switzerland) were placed in the distal metaphysis of each femur. At 28 days, bone quality and implant osseointegration were assessed using microCT, histomorphometrics, and removal torque values (RTVs). Calvarial osteoblasts were isolated and cultured for 7 days on Ø15 mm Ti disks processed to exhibit similar surface characteristics as the implants used for the in vivo studies. The phenotype was assessed by measuring the production of osteocalcin, osteoprotegerin, osteopontin, BMP2, VEGF, and RANKL. RESULTS Microstructured/nanostructured/hydrophilic implants promoted increased bone-to-implant contact and RTVs in vivo and increased osteoblastic marker production in vitro compared to microstructured/non-nanostructured/hydrophobic and microstructured/nanostructured/hydrophobic implants, suggesting that osseointegration occurs in osteoporotic animals, and implant surface properties improve its rate. CONCLUSIONS Although all modified implants were able to osseointegrate in rats with OVX-induced osteoporosis without pharmacologic intervention, the degree of osseointegration was greater around microstructured/nanostructured/hydrophilic implant surfaces. These results suggest that when appropriate microstructure is present, hydrophilicity has a greater influence on Ti implant osseointegration compared to nanostructures. Moreover, modified implant surfaces can exert their control over the altered bone turnover observed in osteoporotic patients to stimulate functional osseointegration. These results provide critical insight for developing implants with improved osseointegration in patients with metabolic disorders of bone remodeling.
Collapse
Affiliation(s)
- Ethan M. Lotz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - David J. Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Barbara D. Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
37
|
Xiang LX, Ran Q, Chen L, Xiang Y, Li FJ, Zhang XM, Xiao YN, Zou LY, Zhong JF, Li SC, Li ZJ. CR6-interacting factor-1 contributes to osteoclastogenesis by inducing receptor activator of nuclear factor κB ligand after radiation. World J Stem Cells 2020; 12:222-240. [PMID: 32266053 PMCID: PMC7118287 DOI: 10.4252/wjsc.v12.i3.222] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/29/2020] [Accepted: 03/15/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Radiation induces rapid bone loss and enhances bone resorption and adipogenesis, leading to an increased risk of bone fracture. There is still a lack of effective preventive or therapeutic method for irradiation-induced bone injury. Receptor activator of nuclear factor κB ligand (RANKL) provides the crucial signal to induce osteoclast differentiation and plays an important role in bone resorption. However, the mechanisms of radiation-induced osteoporosis are not fully understood.
AIM To investigate the role of CR6-interacting factor-1 (Crif1) in osteoclastogenesis after radiation and its possible mechanism.
METHODS C57BL/6 mice were exposed to Co-60 gamma rays and received 5 Gy of whole-body sublethal irradiation at a rate of 0.69 Gy/min. For in vitro study, mouse bone marrow mesenchymal stem/stromal cells (BM-MSCs) were irradiated with Co-60 at a single dose of 9 Gy. For osteoclast induction, monocyte-macrophage RAW264.7 cells were cocultured with mouse BM-MSCs for 7 d. ClusPro and InterProSurf were used to investigate the interaction interface in Crif1 and protein kinase cyclic adenosine monophosphate (cAMP)-activited catalytic subunit alpha complex. Virtual screening using 462608 compounds from the Life Chemicals database around His120 of Crif1 was carried out using the program Autodock_vina. A tetrazolium salt (WST-8) assay was carried out to study the toxicity of compounds to different cells, including human BM-MSCs, mouse BM-MSCs, and Vero cells.
RESULTS Crif1 expression increased in bone marrow cells after radiation in mice. Overexpression of Crif1 in mouse BM-MSCs and radiation exposure could increase RANKL secretion and promote osteoclastogenesis in vitro. Deletion of Crif1 in BM-MSCs could reduce both adipogenesis and RANKL expression, resulting in the inhibition of osteoclastogenesis. Deletion of Crif1 in RAW264.7 cells did not affect the receptor activator of nuclear factor κB expression or osteoclast differentiation. Following treatment with protein kinase A (PKA) agonist (forskolin) and inhibitor (H-89) in mouse BM-MSCs, Crif1 induced RANKL secretion via the cAMP/PKA pathway. Moreover, we identified the Crif1-protein kinase cyclic adenosine monophosphate-activited catalytic subunit alpha interaction interface by in silico studies and shortlisted interface inhibitors through virtual screening on Crif1. Five compounds dramatically suppressed RANKL secretion and adipogenesis by inhibiting the cAMP/PKA pathway.
CONCLUSION Crif1 promotes RANKL expression via the cAMP/PKA pathway, which induces osteoclastogenesis by binding to receptor activator of nuclear factor κB on monocytes-macrophages in the mouse model. These results suggest a role for Crif1 in modulating osteoclastogenesis and provide insights into potential therapeutic strategies targeting the balance between osteogenesis and adipogenesis for radiation-induced bone injury.
Collapse
Affiliation(s)
- Li-Xin Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qian Ran
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Li Chen
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yang Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Feng-Jie Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xiao-Mei Zhang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yan-Ni Xiao
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ling-Yun Zou
- Bioinformatics Center, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Jiang F Zhong
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Shengwen Calvin Li
- CHOC Children’s Research Institute, Children’s Hospital of Orange County, University of California, Irvine, CA 92868, United States
| | - Zhong-Jun Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| |
Collapse
|
38
|
Yan G, Yuan Y, He M, Gong R, Lei H, Zhou H, Wang W, Du W, Ma T, Liu S, Xu Z, Gao M, Yu M, Bian Y, Pang P, Li X, Yu S, Yang F, Cai B, Yang L. m 6A Methylation of Precursor-miR-320/RUNX2 Controls Osteogenic Potential of Bone Marrow-Derived Mesenchymal Stem Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:421-436. [PMID: 31896070 PMCID: PMC6940653 DOI: 10.1016/j.omtn.2019.12.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 01/13/2023]
Abstract
Methyltransferase-like 3 (METTL3) is the main enzyme for N6-methyladenosine (m6A)-based methylation of RNAs and it has been implicated in many biological and pathophysiological processes. In this study, we aimed to explore the potential involvement of METTL3 in osteoblast differentiation and decipher the underlying cellular and molecular mechanisms. We demonstrated that METTL3 is downregulated in human osteoporosis and the ovariectomized (OVX) mouse model, as well as during the osteogenic differentiation. Silence of METTL3 by short interfering RNA (siRNA) decreased m6A methylation levels and inhibited osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) and reduced bone mass, and similar effects were observed in METTL3+/- knockout mice. In contrast, adenovirus-mediated overexpression of METTL3 produced the opposite effects. In addition, METTL3 enhanced, whereas METTL3 silence or knockout suppressed, the m6A methylations of runt-related transcription factor 2 (RUNX2; a key transcription factor for osteoblast differentiation and bone formation) and precursor (pre-)miR-320. Moreover, downregulation of mature miR-320 rescued the decreased bone mass caused by METTL3 silence or METTL3+/- knockout. Therefore, METTL3-based m6A modification favors osteogenic differentiation of BMSCs through m6A-based direct and indirect regulation of RUNX2, and abnormal downregulation of METTL3 is likely one of the mechanisms underlying osteoporosis in patients and mice. Thus, METTL3 overexpression might be considered a new approach of replacement therapy for the treatment of human osteoporosis.
Collapse
Affiliation(s)
- Gege Yan
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Ye Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Mingyu He
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Rui Gong
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hong Lei
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Hongbao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Weijie Du
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Tianshuai Ma
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shenzhen Liu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Zihang Xu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Manqi Gao
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Meixi Yu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yu Bian
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Ping Pang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xin Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shuting Yu
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Fan Yang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Benzhi Cai
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| | - Lei Yang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
39
|
Stem cell-based bone and dental regeneration: a view of microenvironmental modulation. Int J Oral Sci 2019; 11:23. [PMID: 31423011 PMCID: PMC6802669 DOI: 10.1038/s41368-019-0060-3] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/28/2019] [Accepted: 06/12/2019] [Indexed: 02/06/2023] Open
Abstract
In modern medicine, bone and dental loss and defects are common and widespread morbidities, for which regenerative therapy has shown great promise. Mesenchymal stem cells, obtained from various sources and playing an essential role in organ development and postnatal repair, have exhibited enormous potential for regenerating bone and dental tissue. Currently, mesenchymal stem cells (MSCs)-based bone and dental regeneration mainly includes two strategies: the rescue or mobilization of endogenous MSCs and the application of exogenous MSCs in cytotherapy or tissue engineering. Nevertheless, the efficacy of MSC-based regeneration is not always fulfilled, especially in diseased microenvironments. Specifically, the diseased microenvironment not only impairs the regenerative potential of resident MSCs but also controls the therapeutic efficacy of exogenous MSCs, both as donors and recipients. Accordingly, approaches targeting a diseased microenvironment have been established, including improving the diseased niche to restore endogenous MSCs, enhancing MSC resistance to a diseased microenvironment and renormalizing the microenvironment to guarantee MSC-mediated therapies. Moreover, the application of extracellular vesicles (EVs) as cell-free therapy has emerged as a promising therapeutic strategy. In this review, we summarize current knowledge regarding the tactics of MSC-based bone and dental regeneration and the decisive role of the microenvironment, emphasizing the therapeutic potential of microenvironment-targeting strategies in bone and dental regenerative medicine.
Collapse
|
40
|
Soda N, Sakai N, Kato H, Takami M, Fujita T. Singleton-Merten Syndrome-like Skeletal Abnormalities in Mice with Constitutively Activated MDA5. THE JOURNAL OF IMMUNOLOGY 2019; 203:1356-1368. [PMID: 31366715 DOI: 10.4049/jimmunol.1900354] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/01/2019] [Indexed: 12/14/2022]
Abstract
Singleton-Merten syndrome (SMS) is a type I interferonopathy characterized by dental dysplasia, aortic calcification, skeletal abnormalities, glaucoma, and psoriasis. A missense mutation in IFIH1 encoding a cytoplasmic viral RNA sensor MDA5 has recently been identified in the SMS patients as well as in patients with a monogenic form of lupus. We previously reported that Ifih1gs/+ mice express a constitutively active MDA5 and spontaneously develop lupus-like nephritis. In this study, we demonstrate that the Ifih1gs/+ mice also exhibit SMS-like bone abnormalities, including decreased bone mineral density and thin cortical bone. Histological analysis revealed a low number of osteoclasts, low bone formation rate, and abnormal development of growth plate cartilages in Ifih1gs/+ mice. These abnormalities were not observed in Ifih1gs/+ ・Mavs-/- and Ifih1gs/+ ・Ifnar1-/- mice, indicating the critical role of type I IFNs induced by MDA5/MAVS-dependent signaling in the bone pathogenesis of Ifih1gs/+ mice, affecting bone turnover. Taken together, our findings suggest the inhibition of type I IFN signaling as a possible effective therapeutic strategy for bone disorders in SMS patients.
Collapse
Affiliation(s)
- Nobumasa Soda
- Laboratory of Molecular Genetics, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto, 606-8507 Japan.,Laboratory of Molecular and Cellular Immunology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501 Japan
| | - Nobuhiro Sakai
- Department of Pharmacology, School of Dentistry, Showa University, Tokyo, 142-8555 Japan; and
| | - Hiroki Kato
- Laboratory of Molecular Genetics, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto, 606-8507 Japan.,Institute of Cardiovascular Immunology, University Hospital Bonn, University of Bonn, Bonn, 53127 Germany
| | - Masamichi Takami
- Department of Pharmacology, School of Dentistry, Showa University, Tokyo, 142-8555 Japan; and
| | - Takashi Fujita
- Laboratory of Molecular Genetics, Institute for Frontier Life and Medical Science, Kyoto University, Kyoto, 606-8507 Japan; .,Laboratory of Molecular and Cellular Immunology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501 Japan
| |
Collapse
|
41
|
Narayanan A, Srinaath N, Rohini M, Selvamurugan N. Regulation of Runx2 by MicroRNAs in osteoblast differentiation. Life Sci 2019; 232:116676. [PMID: 31340165 DOI: 10.1016/j.lfs.2019.116676] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/12/2019] [Accepted: 07/20/2019] [Indexed: 12/21/2022]
Abstract
Bone is one of the most dynamic organs in the body that continuously undergoes remodeling through bone formation and resorption. A cascade of molecules and pathways results in the osteoblast differentiation that is attributed to osteogenesis, or bone formation. The process of osteogenesis is achieved through participation of the Wnt pathway, FGFs, BMPs/TGF-β, and transcription factors such as Runx2 and Osx. The activity and function of the master transcription factor, Runx2, is of utmost significance as it can induce the function of osteoblast differentiation markers. A number of microRNAs [miRNAs] have been recently identified in the regulation of Runx2 expression/activity, thus affecting the process of osteogenesis. miRNAs that target Runx2 corepressors favor osteogenesis, while miRNAs that target Runx2 coactivators inhibit osteogenesis. In this review, we focus on the regulation of Runx2 by miRNAs in osteoblast differentiation and their potential for treating bone and bone-related diseases.
Collapse
Affiliation(s)
- Akshaya Narayanan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Srinaath
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - M Rohini
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
42
|
Wildman BJ, Godfrey TC, Rehan M, Chen Y, Afreen LH, Hassan Q. MICROmanagement of Runx2 Function in Skeletal Cells. ACTA ACUST UNITED AC 2019; 5:55-64. [PMID: 31289715 DOI: 10.1007/s40610-019-0115-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose of Review- Precise and temporal expression of Runx2 and its regulatory transcriptional network is a key determinant for the intricate cellular and developmental processes in adult bone tissue formation. This review analyzes how microRNA functions to regulate this network, and how dysregulation results in bone disorders. Recent Findings- Similar to other biologic processes, microRNA (miRNA/miR) regulation is undeniably indispensable to bone synthesis and maintenance. There exists a miRNA-RUNX2 network where RUNX2 regulates the transcription of miRs, or is post transcriptionally regulated by a class of miRs, forming a variety of miR-RUNX2 regulatory pathways which regulate osteogenesis. Summary- The current review provides insights to understand transcriptional-post transcriptional regulatory network governed by Runx2 and osteogenic miRs, and is based largely from in vitro and in vivo studies. When taken together, this article discusses a new regulatory layer of bone tissue specific gene expression by RUNX2 influenced via miRNA.
Collapse
Affiliation(s)
- Benjamin J Wildman
- RNA Biology and Epigenetics Laboratory, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama, Birmingham AL, 35294
| | - Tanner C Godfrey
- RNA Biology and Epigenetics Laboratory, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama, Birmingham AL, 35294
| | - Mohammad Rehan
- RNA Biology and Epigenetics Laboratory, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama, Birmingham AL, 35294
| | - Yuechuan Chen
- RNA Biology and Epigenetics Laboratory, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama, Birmingham AL, 35294
| | - Lubana H Afreen
- RNA Biology and Epigenetics Laboratory, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama, Birmingham AL, 35294
| | - Quamarul Hassan
- RNA Biology and Epigenetics Laboratory, Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama, Birmingham AL, 35294
| |
Collapse
|
43
|
Bellavia D, De Luca A, Carina V, Costa V, Raimondi L, Salamanna F, Alessandro R, Fini M, Giavaresi G. Deregulated miRNAs in bone health: Epigenetic roles in osteoporosis. Bone 2019; 122:52-75. [PMID: 30772601 DOI: 10.1016/j.bone.2019.02.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 02/07/2023]
Abstract
MicroRNA (miRNA) has shown to enhance or inhibit cell proliferation, differentiation and activity of different cell types in bone tissue. The discovery of miRNA actions and their targets has helped to identify them as novel regulations actors in bone. Various studies have shown that miRNA deregulation mediates the progression of bone-related pathologies, such as osteoporosis. The present review intends to give an exhaustive overview of miRNAs with experimentally validated targets involved in bone homeostasis and highlight their possible role in osteoporosis development. Moreover, the review analyzes miRNAs identified in clinical trials and involved in osteoporosis.
Collapse
Affiliation(s)
- D Bellavia
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - A De Luca
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - V Carina
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - V Costa
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - L Raimondi
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - F Salamanna
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - R Alessandro
- Department of Biopathology and Medical Biotechnologies, Section of Biology and Genetics, University of Palermo, Palermo 90133, Italy; Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council, Palermo, Italy
| | - M Fini
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| | - G Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, Bologna, Italy
| |
Collapse
|
44
|
Shuai Y, Yang R, Mu R, Yu Y, Rong L, Jin L. MiR-199a-3p mediates the adipogenic differentiation of bone marrow-derived mesenchymal stem cells by regulating KDM6A/WNT signaling. Life Sci 2019; 220:84-91. [DOI: 10.1016/j.lfs.2019.01.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/16/2022]
|
45
|
Yang XH, Yang K, An YL, Wang LB, Luo G, Hu XH. MicroRNA-705 regulates the differentiation of mouse mandible bone marrow mesenchymal stem cells. PeerJ 2019; 7:e6279. [PMID: 30648022 PMCID: PMC6330203 DOI: 10.7717/peerj.6279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
The craniofacial skeleton is the foundation of most stomatological treatments, including prosthodontics and maxillofacial surgery. Although histologically similar to the appendicular skeleton, the craniofacial skeleton manifests many unique properties in response to external stimuli and signals. However, the mandibular or maxillary bone marrow mesenchyme, which is the intrinsic foundation of the functions of craniofacial skeleton, has not been well studied, and its homeostasis mechanism remains elusive. Osteoporosis is a systemic disease that affects all skeletons and is characterized by bone mass loss. Osteoporotic bone marrow mesenchymal stem cells (BMMSCs) exhibit disturbed homeostasis and distorted lineage commitment. Many reports have shown that microRNAs (miRNAs) play important roles in regulating MSCs homeostasis. Here, to obtain a better understanding of mandibular bone marrow MSCs homeostasis, we isolated and cultured mandible marrow MSCs from mouse mandibles. Using miR-705 mimics and an inhibitor, we demonstrated that miR-705 played a vital role in shifting the mandibular MSCs lineage commitment in vitro. Utilizing an osteoporosis mouse model, we demonstrated that MSCs from ovariectomized (OVX) mouse mandibular bone marrow exhibited impaired osteogenic and excessive adipogenic differentiation. miR-705 was found overexpressed in OVX mandibular MSCs. The knock down of miR-705 in vitro partially attenuated the differentiation disorder of the OVX mandibular MSCs by upregulating the expression of osteogenic marker genes but suppressing adipogenic genes. Taken together, our findings provide a better understanding of the homeostasis mechanism of mandibular BMMSCs and a novel potential therapeutic target for treating mandibular osteoporosis.
Collapse
Affiliation(s)
- Xiao Hong Yang
- Department of Prosthetics, the Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China
| | - Kun Yang
- Department of Periodontology, the Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi Medical university, Zunyi, Guizhou, China
| | - Yu Lin An
- Department of Stomatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Li Bo Wang
- Department of Prosthetics, the Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi Medical University, Zunyi, Guizhou, China.,Department of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Guo Luo
- Department of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiao Hua Hu
- Department of Oral and Maxillofacial Surgery, the Affiliated Stomatology Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
46
|
Lo PK, Wolfson B, Zhou Q. Adipogenesis and Noncoding RNAs. HANDBOOK OF NUTRITION, DIET, AND EPIGENETICS 2019:623-645. [DOI: 10.1007/978-3-319-55530-0_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
47
|
Gender-independent efficacy of mesenchymal stem cell therapy in sex hormone-deficient bone loss via immunosuppression and resident stem cell recovery. Exp Mol Med 2018; 50:1-14. [PMID: 30559383 PMCID: PMC6297134 DOI: 10.1038/s12276-018-0192-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis develops with high prevalence in both postmenopausal women and hypogonadal men. Osteoporosis results in significant morbidity, but no cure has been established. Mesenchymal stem cells (MSCs) critically contribute to bone homeostasis and possess potent immunomodulatory/anti-inflammatory capability. Here, we investigated the therapeutic efficacy of using an infusion of MSCs to treat sex hormone-deficient bone loss and its underlying mechanisms. In particular, we compared the impacts of MSC cytotherapy in the two genders with the aim of examining potential gender differences. Using the gonadectomy (GNX) model, we confirmed that the osteoporotic phenotypes were substantially consistent between female and male mice. Importantly, systemic MSC transplantation (MSCT) not only rescued trabecular bone loss in GNX mice but also restored cortical bone mass and bone quality. Unexpectedly, no differences were detected between the genders. Furthermore, MSCT demonstrated an equal efficiency in rectifying the bone remodeling balance in both genders of GNX animals, as proven by the comparable recovery of bone formation and parallel normalization of bone resorption. Mechanistically, using green fluorescent protein (GFP)-based cell-tracing, we demonstrated rapid engraftment but poor inhabitation of donor MSCs in the GNX recipient bone marrow of each gender. Alternatively, MSCT uniformly reduced the CD3+T-cell population and suppressed the serum levels of inflammatory cytokines in reversing female and male GNX osteoporosis, which was attributed to the ability of the MSC to induce T-cell apoptosis. Immunosuppression in the microenvironment eventually led to functional recovery of endogenous MSCs, which resulted in restored osteogenesis and normalized behavior to modulate osteoclastogenesis. Collectively, these data revealed recipient sexually monomorphic responses to MSC therapy in gonadal steroid deficiency-induced osteoporosis via immunosuppression/anti-inflammation and resident stem cell recovery.
Collapse
|
48
|
Mechanisms of Zuogui Pill in Treating Osteoporosis: Perspective from Bone Marrow Mesenchymal Stem Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:3717391. [PMID: 30327678 PMCID: PMC6169217 DOI: 10.1155/2018/3717391] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/04/2018] [Indexed: 01/08/2023]
Abstract
The current treatment strategies for osteoporosis (OP) involve promoting osteogenic differentiation and inhibiting adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). According to a theory of traditional Chinese medicine (TCM), the kidneys contain an “essence” that regulate bone metabolism and generate marrow. Kidney disorders are therefore considered to be a major cause of OP as per the principles of TCM, which recommends kidney-tonifying treatments for OP. The Zuogui pill (ZGP) is a classic kidney-tonifying medication that effectively improves OP symptoms. Studies have shown that ZGP can promote the osteogenic differentiation of BMSCs, providing scientific evidence for the TCM theory linking kidneys with bone metabolism. In this review, we have provided an overview of recent studies that examined the underlying mechanisms of ZGP mediated regulation of BMSC osteogenic and adipogenic differentiation.
Collapse
|
49
|
Rarani FZ, Borhani F, Rashidi B. Endometrial pinopode biomarkers: Molecules and microRNAs. J Cell Physiol 2018; 233:9145-9158. [PMID: 29968908 DOI: 10.1002/jcp.26852] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/10/2018] [Indexed: 12/12/2022]
Abstract
Ultrastructural changes on the apical surface of the luminal epithelium of the uterus are known as pinopodes. Their morphology in species and in special species is associated with different results about size, duration, and percentage of surface area covered by pinopodes. The content of pinopodes is different in rodents and humans. In mice and rats pinopodes have many vacuoles and no organelle that extends to the actin stalk above the microvilli. Human pinopodes do not have a large vacuole and contain the golgi complex, a rough endoplasmic reticulum, secretory vesicles, and mitochondria that extend from the entire cell surface. It has been suggested that pinopodes are good markers of endometrial receptivity and implantation window. There are several molecular markers related to the presence of pinopodes, including integrins, leukemia inhibiting factor (LIF), l-selectin, HOXA10, glutaredoxin, glycodelinA, heparin-binding epidermal growth factor, mucins, and microRNAs (miRNAs). Multiple lines of evidence have indicated that miRNAs could affect the expression of LIF and pinopodes in the endometrium and these molecules play key roles in implantation window processes. Here, we have summarized the morphology and function of pinopodes. Moreover, we have highlighted several molecules in relation to pinopodes that could be used as biomarkers.
Collapse
Affiliation(s)
- Fahimeh Zamani Rarani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fatemeh Borhani
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.,Department of Basic Sciences, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahman Rashidi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
50
|
Chen X, Zhi X, Yin Z, Li X, Qin L, Qiu Z, Su J. 18β-Glycyrrhetinic Acid Inhibits Osteoclastogenesis In Vivo and In Vitro by Blocking RANKL-Mediated RANK-TRAF6 Interactions and NF-κB and MAPK Signaling Pathways. Front Pharmacol 2018; 9:647. [PMID: 29973878 PMCID: PMC6019442 DOI: 10.3389/fphar.2018.00647] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/30/2018] [Indexed: 12/21/2022] Open
Abstract
Bone metabolism is determined by a delicate balance between bone resorption by osteoclasts and bone formation by osteoblasts. The imbalance due to over-activated osteoclasts plays an important role in various diseases. Activation of NF-κB and MAPK signaling pathways by receptor activator of nuclear factor -κB ligand (RANKL) is vital for osteoclastogenesis. Here, we for the first time explored the effects of 18β-glycyrrhetinic acid (18β-GA), a pentacyclic triterpenoid found in the Glycyrrhiza glabra L roots, on RANKL-induced osteoclastogenesis, osteoclast functions and signaling pathways in vitro and in vivo. In bone marrow monocytes (BMMs) and RAW264.7 cells, 18β-GA inhibited osteoclastogenesis, decreased expression of TRAP, cathepsin K, CTR and MMP-9, blocked actin ring formation and compromised osteoclasts functions in a dose-dependent manner at an early stage with minimal effects on osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). For underlying molecular mechanisms, 18β-GA inhibited RANKL-induced phosphorylation of p65, p50, and IκB, blocked p65 nuclear translocation and decreased the DNA-binding activity of NF-κB. Besides, 18β-GA inhibited the activation of the MAPK pathways. Co-immunoprecipitation showed that 18β-GA treatment blocked RANK-TRAF6 association at an upstream site. In vivo, 18β-GA treatment inhibited ovariectomy-induced osteoclastogenesis and reduced bone loss in mice. Overall, our results demonstrated that 18β-GA inhibited RANKL-induced osteoclastogenesis by inhibiting RANK expression in preosteoclasts and blocking the binding of RANK and TRAF6 which lead to the inhibition of NF-κB and MAPK signaling pathways. 18β-GA is a promising novel candidate in the treatment of osteoclast-related diseases such as postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
- China-South Korea Bioengineering Center, Shanghai, China
| | - Xin Zhi
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Xiaoqun Li
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Longjuan Qin
- Orthopedic Basic and Translational Research Center, Jiangyin, China
| | - Zili Qiu
- Jinling High School, Nanjing, China
| | - Jiacan Su
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
- China-South Korea Bioengineering Center, Shanghai, China
| |
Collapse
|