1
|
Zhu Y, Liu Y, Yang K, Wu W, Cheng Y, Ding Y, Gu R, Liu H, Zhang X, Liu Y. Apoptotic vesicles inhibit bone marrow adiposity via wnt/β-catenin signaling. Regen Ther 2025; 29:262-270. [PMID: 40230357 PMCID: PMC11994938 DOI: 10.1016/j.reth.2025.03.012] [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: 09/29/2024] [Revised: 02/14/2025] [Accepted: 03/18/2025] [Indexed: 04/16/2025] Open
Abstract
Background There is currently increasing focus on aging-related diseases. Osteoporosis is a common disease the incidence of which increases with age. In older patients with osteoporosis, bone marrow mesenchymal stem cells (BMMSCs) have a decreased capacity for osteogenesis and an increased capacity for adipogenesis, causing excessive accumulation of adipose tissue in the bone marrow. Therefore, means of reducing bone marrow adiposity may have therapeutic potential for osteoporosis. Apoptotic vesicles (apoVs) participate in a wide range of physiological processes and have been shown to have therapeutic effects in a variety of diseases. The principal objective of this study was to examine the special properties and regulatory mechanisms of BMMSC-derived apoVs in the treatment of bone marrow adiposity. Results The results showed that apoVs could decrease bone marrow adiposity in osteoporotic mice and prevent adipogenic differentiation of MSCs by activating the Wnt/β-catenin pathway. Conclusion New apoV-based therapies have potential for the treatment of bone marrow adiposity in patients with aging-related osteoporosis and may be further applicable to the treatment of obesity and aging-related diseases.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- Department of Stomatology, Peking University Third Hospital, Beijing 100191, China
| | - Yaoshan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Kunkun Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Weiliang Wu
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, China
| | - Yawen Cheng
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Yanan Ding
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
| | - Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing 100081, China
- National Center of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
2
|
Wen RM, Wang HX. Effect of adipokines on bone marrow mesenchymal stem cell function. World J Stem Cells 2025; 17:106150. [DOI: 10.4252/wjsc.v17.i5.106150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/23/2025] [Accepted: 04/09/2025] [Indexed: 05/26/2025] Open
Abstract
During excessive adipose tissue accumulation, various adipokines such as visfatin, chemerin, vaspin, and adiponectin are released into systemic circulation, thereby influencing metabolic tissue function throughout the body. As multifunctional signaling molecules secreted by adipose tissue, adipokines play a pivotal role in metabolic regulation, inflammatory response, and tissue homeostasis. Recent studies have demonstrated that adipokines can influence skeletal system repair and regeneration by modulating bone marrow-derived mesenchymal stem cell (BMSC) proliferation, differentiation, migration, and immunomodulatory functions. However, different adipokines have distinct roles in regulating BMSC function, but their underlying molecular mechanisms are not fully understood. In this review, we systematically summarize the specific mechanisms of action and potential clinical applications of visfatin, chemerin, vaspin, and adiponectin on BMSC function in order to reveal new mechanisms of interaction between adipokines and BMSCs. The aim is to provide a theoretical basis for targeted treatment strategies for bone diseases targeting adipokines.
Collapse
Affiliation(s)
- Rui-Ming Wen
- School of Sports Health, Shenyang Sport University, Shenyang 110102, Liaoning Province, China
| | - Hai-Xia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang 110102, Liaoning Province, China
| |
Collapse
|
3
|
Tahoori M, Tafreshi AP, Naghshnejad F, Zeynali B. Transforming Growth Factor-β Signaling Inhibits the Osteogenic Differentiation of Mesenchymal Stem Cells via Activation of Wnt/β-Catenin Pathway. J Bone Metab 2025; 32:11-20. [PMID: 40098425 PMCID: PMC11960301 DOI: 10.11005/jbm.24.761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Due to the contradictory and temporally variable effects of transforming growth factor-β (TGF-β) and the Wnt/β-catenin pathways on osteogenic differentiation in different stem cell types, we sought to examine the activity of these pathways as well as their interaction during the osteogenic differentiation of the osteo-induced adiposederived mesenchymal stem cells (AD-MSCs). METHODS The osteo-induced AD-MSCs were treated with TGF-β1 (1 ng/mL) either alone or together with its antagonist SB- 431542 (10 μM) or that of the Wnt antagonist, inhibitor of Wnt production 2 (IWP2) (3 μM), every 3 days for 21 days. Cells were then analyzed for calcium deposit, bone matrix production, and the osteogenic markers gene expression. RESULTS Our results showed firstly that, either of the pathways is active since the mRNA expressions of their respective target genes, PAI-1 and Cyclin D1 were detectable although the latter was at a very low level. Secondly that, treatment with TGF-β1 decreased levels of calcium deposit, bone matrix production and the osteogenic markers gene expression. Accordingly, osteogenesis was induced in those treated with SB either alone or together with the TGF-β1, pointing to inhibitory effect of TGF-β pathway on osteogenic differentiation. Thirdly that following treatment with IWP2 and TGF-β1, the inhibitory effect of TGF-β1 on bone matrix production was reversed. Fourthly, there was constantly low expression of Wnt3amRNA but progressively increasing that of its endogenous antagonist Dkk-1mRNA throughout. CONCLUSIONS Together these results suggest that TGF-β1 requires the active Wnt/β-catenin signaling pathway to exert its inhibitory effects on osteogenic differentiation of AD-MSCs.
Collapse
Affiliation(s)
- Mahsa Tahoori
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran,
Iran
| | - Azita Parvaneh Tafreshi
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran,
Iran
- Department of Molecular Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran,
Iran
| | - Fatemeh Naghshnejad
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran,
Iran
| | - Bahman Zeynali
- Developmental Biology Laboratory, School of Biology, College of Science, University of Tehran, Tehran,
Iran
| |
Collapse
|
4
|
An F, Jia X, Shi Y, Xiao X, Yang F, Su J, Peng X, Geng G, Yan C. The ultimate microbial composition for correcting Th17/Treg cell imbalance and lipid metabolism disorders in osteoporosis. Int Immunopharmacol 2025; 144:113613. [PMID: 39571271 DOI: 10.1016/j.intimp.2024.113613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 12/15/2024]
Abstract
Osteoporosis is a systemic bone disease characterised by decreased bone mass and a deteriorated bone microstructure, leading to increased bone fragility and fracture risk. Disorders of the intestinal microbiota may be key inducers of osteoporosis. Furthermore, such disorders may contribute to osteoporosis by influencing immune function and lipid metabolism. Therefore, in this review, we aimed to summarise the molecular mechanisms through which the intestinal microbiota affect the onset and development of osteoporosis by regulating Th17/Treg imbalance and lipid metabolism disorders. We also discussed the regulatory mechanisms underlying the effect of intestinal microbiota-related modulators on Th17/Treg imbalance and lipid metabolism disorders in osteoporosis, to explore new molecular targets for its treatment and provide a theoretical basis for clinical management.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China.
| | - Xueru Jia
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Yangyang Shi
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Xiaolong Xiao
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Fan Yang
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Junchang Su
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Xia Peng
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Guangqin Geng
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China
| | - Chunlu Yan
- School of Tradional Chinese and Werstern Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu, China.
| |
Collapse
|
5
|
Zhao Y, Liu F, Pei Y, Lian F, Lin H. Involvement of the Wnt/β-catenin signalling pathway in heterotopic ossification and ossification-related diseases. J Cell Mol Med 2024; 28:e70113. [PMID: 39320014 PMCID: PMC11423343 DOI: 10.1111/jcmm.70113] [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: 03/13/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Heterotopic ossification (HO) is a pathological condition characterized by the formation of bone within soft tissues. The development of HO is a result of abnormal activation of the bone formation programs, where multiple signalling pathways, including Wnt/β-catenin, BMP and hedgehog signalling, are involved. The Wnt/β-catenin signalling pathway, a conserved pathway essential for various fundamental activities, has been found to play a significant role in pathological bone formation processes. It regulates angiogenesis, chondrocyte hypertrophy and osteoblast differentiation during the development of HO. More importantly, the crosstalk between Wnt signalling and other factors including BMP, Hedgehog signalling, YAP may contribute in a HO-favourable manner. Moreover, several miRNAs may also be involved in HO formation via the regulation of Wnt signalling. This review aims to summarize the role of Wnt/β-catenin signalling in the pathogenesis of HO, its interactions with related molecules, and potential preventive and therapeutic measures targeting Wnt/β-catenin signalling.
Collapse
Affiliation(s)
- Yike Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary school, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fangzhou Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary school, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yiran Pei
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary school, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fengyu Lian
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Queen Mary school, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hui Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
6
|
An F, Song J, Chang W, Zhang J, Gao P, Wang Y, Xiao Z, Yan C. Research Progress on the Mechanism of the SFRP-Mediated Wnt Signalling Pathway Involved in Bone Metabolism in Osteoporosis. Mol Biotechnol 2024; 66:975-990. [PMID: 38194214 DOI: 10.1007/s12033-023-01018-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024]
Abstract
Osteoporosis (OP) is a metabolic bone disease linked to an elevated fracture risk, primarily stemming from disruptions in bone metabolism. Present clinical treatments for OP merely alleviate symptoms. Hence, there exists a pressing need to identify novel targets for the clinical treatment of OP. Research indicates that the Wnt signalling pathway is modulated by serum-secreted frizzled-related protein 5 (SFRP5), potentially serving as a pivotal regulator in bone metabolism disorders. Moreover, studies confirm elevated SFRP5 expression in OP, with SFRP5 overexpression leading to the downregulation of Wnt and β-catenin proteins in the Wnt signalling pathway, as well as the expression of osteogenesis-related marker molecules such as RUNX2, ALP, and OPN. Conversely, the opposite has been reported when SFRP5 is knocked out, suggesting that SFRP5 may be a key factor involved in the regulation of bone metabolism via the Wnt signalling axis. However, the molecular mechanisms underlying the action of SFRP5-induced OP have yet to be comprehensively elucidated. This review focusses on the molecular structure and function of SFRP5 and the potential molecular mechanisms of the SFRP5-mediated Wnt signalling pathway involved in bone metabolism in OP, providing reasonable evidence for the targeted therapy of SFRP5 for the prevention and treatment of OP.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jiayi Song
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Weirong Chang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Yujie Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Zhipan Xiao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China
| | - Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
7
|
Rosen CJ, Horowitz MC. Nutrient regulation of bone marrow adipose tissue: skeletal implications of weight loss. Nat Rev Endocrinol 2023; 19:626-638. [PMID: 37587198 PMCID: PMC10592027 DOI: 10.1038/s41574-023-00879-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/18/2023]
Abstract
Adipose tissue is a dynamic component of the bone marrow, regulating skeletal remodelling and secreting paracrine and endocrine factors that can affect haematopoiesis, as well as potentially nourishing the bone marrow during periods of stress. Bone marrow adipose tissue is regulated by multiple factors, but particularly nutrient status. In this Review, we examine how bone marrow adipocytes originate, their function in normal and pathological states and how bone marrow adipose tissue modulates whole-body homoeostasis through actions on bone cells, haematopoietic stem cells and extra-medullary adipocytes during nutritional challenges. We focus on both rodent models and human studies to help understand the unique marrow adipocyte, its response to the external nutrient environment and its effects on the skeleton. We finish by addressing some critical questions that to date remain unanswered.
Collapse
Affiliation(s)
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Chatree K, Sriboonaied P, Phetkong C, Wattananit W, Chanchao C, Charoenpanich A. Distinctions in bone matrix nanostructure, composition, and formation between osteoblast-like cells, MG-63, and human mesenchymal stem cells, UE7T-13. Heliyon 2023; 9:e15556. [PMID: 37153435 PMCID: PMC10160763 DOI: 10.1016/j.heliyon.2023.e15556] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 05/09/2023] Open
Abstract
Osteoblast-like cells and human mesenchymal stem cells (hMSCs) are frequently employed as osteoprogenitor cell models for evaluating novel biomaterials in bone healing and tissue engineering. In this study, the characterization of UE7T-13 hMSCs and MG-63 human osteoblast-like cells was examined. Both cells can undergo osteogenesis and produce calcium extracellular matrix; however, calcium nodules produced by MG-63 lacked a central mass and appeared flatter than UE7T-13. The absence of growing calcium nodules in MG-63 was discovered by SEM-EDX to be associated with the formation of alternating layers of cells and calcium extracellular matrix. The nanostructure and composition analysis showed that UE7T-13 had a finer nanostructure of calcium nodules with a higher calcium/phosphate ratio than MG-63. Both cells expressed high intrinsic levels of collagen type I alpha 1 chain, while only UE7T-13 expressed high levels of alkaline phosphatase, biomineralization associated (ALPL). High ALP activity in UE7T-13 was not further enhanced by osteogenic induction, but in MG-63, low intrinsic ALP activity was greatly induced by osteogenic induction. These findings highlight the differences between the two immortal osteoprogenitor cell lines, along with some technical notes that should be considered while selecting and interpreting the pertinent in vitro model.
Collapse
Affiliation(s)
- Kamonwan Chatree
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Patsawee Sriboonaied
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Chinnatam Phetkong
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Witoon Wattananit
- Scientific and Technological Equipment Centre, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| | - Chanpen Chanchao
- Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Adisri Charoenpanich
- Department of Biology, Faculty of Science, Silpakorn University, Nakhon Pathom, Thailand
| |
Collapse
|
9
|
Zhang H, Zhang Q, Yuan Z, Dong J. Non-coding RNAs in ossification of the posterior longitudinal ligament. Front Genet 2022; 13:1069575. [PMID: 36506306 PMCID: PMC9729789 DOI: 10.3389/fgene.2022.1069575] [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: 10/14/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Ossification of the posterior longitudinal ligament (OPLL) is a kind of disease that involves a variety of factors leading to ectopic bone deposition of the spinal ligament. Although the detailed mechanism is not clear, genetic factors play important roles in the development of this disease. Noncoding RNA (ncRNA) refers to an RNA molecule that is not translated into a protein but participates in the regulation of gene expression. Functionally important types of ncRNA associated with OPLL include long noncoding RNA, microRNA, and circular RNA. We listed the differentially expressed ncRNAs in OPLL patients and normal controls to find the ncRNAs most relevant to the pathogenesis of the disease. The potential regulatory networks of ncRNA in OPLL cells were analyzed based on their most abundant signal transduction pathway data. The analysis of the highly connected ncRNAs in the regulatory network suggests that they play an important role in OPLL. These findings provide new directions for the study of OPLL pathogenesis and therapeutic targets. In this paper, we reviewed and analyzed the literature on ncRNAs in OPLL published in recent years, aiming to help doctors better understand and treat this disease.
Collapse
|
10
|
Lineage-selective super enhancers mediate core regulatory circuitry during adipogenic and osteogenic differentiation of human mesenchymal stem cells. Cell Death Dis 2022; 13:866. [PMID: 36224171 PMCID: PMC9556616 DOI: 10.1038/s41419-022-05309-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Human mesenchymal stem cells (hMSCs) can be differentiated into osteoblasts and adipocytes. During these processes, super enhancers (SEs) play important roles. Here, we performed comprehensive characterization of the SEs changes associated with adipogenic and osteogenic differentiation of hMSCs, and revealed that SEs changed more dramatically compared with typical enhancers. We identified a set of lineage-selective SEs, whose target genes were enriched with cell type-specific functions. Functional experiments in lineage-selective SEs demonstrated their specific roles in directed differentiation of hMSCs. We also found that some key transcription factors regulated by lineage-selective SEs could form core regulatory circuitry (CRC) to regulate each other's expression and control the hMSCs fate determination. In addition, we found that GWAS SNPs of osteoporosis and obesity were significantly enriched in osteoblasts-selective SEs or adipocytes-selective SEs, respectively. Taken together, our studies unveiled important roles of lineage-selective SEs in hMSCs differentiation into osteoblasts and adipocytes.
Collapse
|
11
|
Du Y, Liu Y, Zhou Y, Zhang P. Knockdown of CDC20 promotes adipogenesis of bone marrow-derived stem cells by modulating β-catenin. Stem Cell Res Ther 2022; 13:443. [PMID: 36056439 PMCID: PMC9438178 DOI: 10.1186/s13287-022-03062-0] [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: 12/20/2021] [Accepted: 06/04/2022] [Indexed: 11/26/2022] Open
Abstract
Background Bone is a rigid organ that provides physical protection and support to vital organs of the body. Bone loss disorders are commonly associated with increased bone marrow adipose tissue. Bone marrow mesenchymal stromal/stem cells (BMSCs) are multipotent progenitors that can differentiate into osteoblasts, adipocytes, and chondrocytes. Cell division cycle 20 (CDC20) is a co-activator of anaphase promoting complex/cyclosome (APC/C), and is required for ubiquitin ligase activity. Our previous study showed that CDC20 promoted the osteogenic commitment of BMSCs and Cdc20 conditional knockout mice suggested a decline in bone mass. In this study, we found that knockdown of CDC20 promoted adipogenic differentiation of BMSCs by modulating β-catenin, which suggested a link between adipogenesis and osteogenesis. Methods Lentivirus containing a CDC20 shRNA was used for CDC20 knockdown in human BMSCs (hBMSCs). Primary mouse BMSCs (mBMSCs) were isolated from Cdc20f/f and Sp7-Cre;Cdc20f/f mice. Adipogenesis was examined using quantitative real-time reverse transcription PCR (qRT-PCR) and western blotting analysis of adipogenic regulators, Oil Red O staining, and transplantation into nude mice. CDC20 knockout efficiency was determined through immunochemistry, qRT-PCR, and western blotting of bone marrow. Accumulation of adiposity was measured through histology and staining of bone sections. Exploration of the molecular mechanism was determined through western blotting, Oil Red O staining, and qRT-PCR. Results CDC20 expression in hBMSCs was significantly decreased during adipogenic differentiation. CDC20 knockdown enhanced hBMSC adipogenic differentiation in vitro. CDC20-knockdown hBMSCs showed more adipose tissue-like constructs upon hematoxylin and eosin (H&E) and Oil Red O staining. Sp7-Cre;Cdc20f/f mice presented increased adipocytes in their bone marrow compared with the control mice. mBMSCs from Sp7-Cre;Cdc20f/f mice showed upregulated adipogenic differentiation. Knockdown of CDC20 led to decreased β-catenin levels, and a β-catenin pathway activator (lithium chloride) abolished the role of CDC20 in BMSC adipogenic differentiation. Conclusions Our findings showed that CDC20 knockdown enhanced adipogenesis of hBMSC and mBMSCs adipogenesis in vitro and in vivo. CDC20 regulates both adipogenesis and osteogenesis of BMSCs, and might lead to the development of new therapeutic targets for “fatty bone” and osteoporosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03062-0.
Collapse
Affiliation(s)
- Yangge Du
- Department of Prosthodontics, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yunsong Liu
- Department of Prosthodontics, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China
| | - Yongsheng Zhou
- Department of Prosthodontics, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.
| | - Ping Zhang
- Department of Prosthodontics, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, National Clinical Research Center for Oral Diseases, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing, 100081, People's Republic of China.
| |
Collapse
|
12
|
Costantini A, Mäkitie RE, Hartmann MA, Fratzl-Zelman N, Zillikens MC, Kornak U, Søe K, Mäkitie O. Early-Onset Osteoporosis: Rare Monogenic Forms Elucidate the Complexity of Disease Pathogenesis Beyond Type I Collagen. J Bone Miner Res 2022; 37:1623-1641. [PMID: 35949115 PMCID: PMC9542053 DOI: 10.1002/jbmr.4668] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 12/05/2022]
Abstract
Early-onset osteoporosis (EOOP), characterized by low bone mineral density (BMD) and fractures, affects children, premenopausal women and men aged <50 years. EOOP may be secondary to a chronic illness, long-term medication, nutritional deficiencies, etc. If no such cause is identified, EOOP is regarded primary and may then be related to rare variants in genes playing a pivotal role in bone homeostasis. If the cause remains unknown, EOOP is considered idiopathic. The scope of this review is to guide through clinical and genetic diagnostics of EOOP, summarize the present knowledge on rare monogenic forms of EOOP, and describe how analysis of bone biopsy samples can lead to a better understanding of the disease pathogenesis. The diagnostic pathway of EOOP is often complicated and extensive assessments may be needed to reliably exclude secondary causes. Due to the genetic heterogeneity and overlapping features in the various genetic forms of EOOP and other bone fragility disorders, the genetic diagnosis usually requires the use of next-generation sequencing to investigate several genes simultaneously. Recent discoveries have elucidated the complexity of disease pathogenesis both regarding genetic architecture and bone tissue-level pathology. Two rare monogenic forms of EOOP are due to defects in genes partaking in the canonical WNT pathway: LRP5 and WNT1. Variants in the genes encoding plastin-3 (PLS3) and sphingomyelin synthase 2 (SGMS2) have also been found in children and young adults with skeletal fragility. The molecular mechanisms leading from gene defects to clinical manifestations are often not fully understood. Detailed analysis of patient-derived transiliac bone biopsies gives valuable information to understand disease pathogenesis, distinguishes EOOP from other bone fragility disorders, and guides in patient management, but is not widely available in clinical settings. Despite the great advances in this field, EOOP remains an insufficiently explored entity and further research is needed to optimize diagnostic and therapeutic approaches. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Alice Costantini
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Paris Cité University, INSERM UMR1163, Institut Imagine, Paris, France
| | - Riikka E Mäkitie
- Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Otorhinolaryngology-Head and Neck Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Markus A Hartmann
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria.,Vienna Bone and Growth Center, Vienna, Austria
| | - Nadja Fratzl-Zelman
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria.,Vienna Bone and Growth Center, Vienna, Austria
| | - M Carola Zillikens
- Bone Center, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Uwe Kornak
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark.,Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Outi Mäkitie
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Children's Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
13
|
Liu J, Gan L, Ma B, He S, Wu P, Li H, Xiong J. Alterations in chromatin accessibility during osteoblast and adipocyte differentiation in human mesenchymal stem cells. BMC Med Genomics 2022; 15:17. [PMID: 35101056 PMCID: PMC8802426 DOI: 10.1186/s12920-022-01168-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
Although differential expression of genes is apparent during the adipogenic/osteogenic differentiation of marrow mesenchymal stem cells (MSCs), it is not known whether this is associated with changes in chromosomal structure. In this study, we used ATAC-sequencing technology to observe variations in chromatin assembly during the early stages of MSC differentiation. This showed significant changes in the number and distribution of chromosome accessibility at different time points of adipogenic/osteogenic differentiation. Sequencing of differential peaks indicated alterations in transcription factor motifs involved in MSC differentiation. Gene Ontology (GO) and pathway analysis indicated that changes in biological function resulted from the alterations in chromatin accessibility. We then integrated ATAC-seq and RNA-seq and found that only a small proportion of the overlapping genes were screened out from ATAC-seq and RNA-seq overlapping. Through GO and pathway analysis of these overlapped genes, we not only observed some known biological functions related to adipogenic/osteogenic differentiation but also noticed some unusual biological clustering during MSC differentiation. In summary, our work not only presents the landscape of chromatin accessibility of MSC during differentiation but also helps to further our understanding of the underlying mechanisms of gene expression in these processes.
Collapse
Affiliation(s)
- Jianyun Liu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Lijun Gan
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Baichen Ma
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Shan He
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Ping Wu
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Huiming Li
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China
| | - Jianjun Xiong
- Jiangxi Provincial Key Laboratory of Systems Biomedicine, Jiujiang University, Jiujiang, 332000, China.
| |
Collapse
|
14
|
Genome-wide identification and expression profiling analysis of Wnt family genes affecting adipocyte differentiation in cattle. Sci Rep 2022; 12:489. [PMID: 35017603 PMCID: PMC8752766 DOI: 10.1038/s41598-021-04468-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022] Open
Abstract
The Wnt family features conserved glycoproteins that play roles in tissue regeneration, animal development and cell proliferation and differentiation. For its functional diversity and importance, this family has been studied in several species, but not in the Bovinae. Herein we identified 19 Wnt genes in cattle, and seven other species of Bovinae, and described their corresponding protein properties. Phylogenetic analysis clustered the 149 Wnt proteins in Bovinae, and 38 Wnt proteins from the human and mouse into 12 major clades. Wnt genes from the same subfamilies shared similar protein motif compositions and exon–intron patterns. Chromosomal distribution and collinearity analysis revealed that they were conservative in cattle and five species of Bovinae. RNA-seq data analysis indicated that Wnt genes exhibited tissue-specific expression in cattle. qPCR analysis revealed a unique expression pattern of each gene during bovine adipocytes differentiation. Finally, the comprehensive analysis indicated that Wnt2B may regulate adipose differentiation by activating FZD5, which is worthy of further study. Our study presents the first genome-wide study of the Wnt gene family in Bovinae, and lays the foundation for further functional characterization of this family in bovine adipocytes differentiation.
Collapse
|
15
|
Tencerova M, Lundby L, Buntzen S, Norderval S, Hougaard HT, Pedersen BG, Kassem M. Molecular differences of adipose-derived mesenchymal stem cells between non-responders and responders in treatment of transphincteric perianal fistulas. Stem Cell Res Ther 2021; 12:586. [PMID: 34819138 PMCID: PMC8611942 DOI: 10.1186/s13287-021-02644-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/31/2021] [Indexed: 12/12/2022] Open
Abstract
Background Injection of autologous adipose tissue (AT) has recently been demonstrated to be an effective and safe treatment for anal fistulas. AT mesenchymal stem cells (AT-MSCs) mediate the healing process, but the relationship between molecular characteristics of AT-MSCs of the injected AT and fistula healing has not been adequately studied. Thus we aimed to characterize the molecular and functional properties of AT-MSCs isolated from autologous AT injected as a treatment of cryptogenic high transsphincteric perianal fistulas and correlate these findings to the healing process.
Methods 27 patients (age 45 ± 2 years) diagnosed with perianal fistula were enrolled in the study and treated with autologous AT injected around the anal fistula tract. AT-MSCs were isolated for cellular and molecular analyses. The fistula healing was evaluated by MRI scanning after 6 months of treatment. AT-MSC phenotype was compared between responders and non-responders with respect to fistula healing. Results 52% of all patients exhibited clinical healing of the fistulas as evaluated 6 months after last injection. Cultured AT-MSCs in the responder group had a lower short-term proliferation rate and higher osteoblast differentiation potential compared to non-responder AT-MSCs. On the other hand, adipocyte differentiation potential of AT-MSCs was higher in non-responder group. Interestingly, AT-MSCs of responders exhibited lower expression of inflammatory and senescence associated genes such as IL1B, NFKB, CDKN2A, TPB3,TGFB1. Conclusion Our data suggest that cellular quality of the injected AT-MSCs including cell proliferation, differentiation capacity and secretion of proinflammatory molecules may provide a possible mechanism underlying fistula healing. Furthermore, these biomarkers may be useful to predict a positive fistula healing outcome. Trial registration: NTC04834609, Registered 6 April 2021. https://clinicaltrials.gov/ct2/show/NCT04834609 Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02644-8.
Collapse
Affiliation(s)
- Michaela Tencerova
- Molecular Endocrinology and Stem Cell Research Unit, Department of Endocrinology and Metabolism, Odense University Hospital and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark. .,Molecular Physiology of Bone, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic.
| | - Lilli Lundby
- Department of Surgery, Pelvic Floor Unit, Aarhus University Hospital, Århus, Denmark
| | - Steen Buntzen
- Department of Surgery, Pelvic Floor Unit, Aarhus University Hospital, Århus, Denmark.,Department of Gastrointestinal Surgery, University Hospital of North Norway, Tromsoe, Norway.,Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsö, Norway
| | - Stig Norderval
- Department of Gastrointestinal Surgery, University Hospital of North Norway, Tromsoe, Norway.,Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsö, Norway
| | - Helene Tarri Hougaard
- Department of Surgery, Pelvic Floor Unit, Aarhus University Hospital, Århus, Denmark
| | | | - Moustapha Kassem
- Molecular Endocrinology and Stem Cell Research Unit, Department of Endocrinology and Metabolism, Odense University Hospital and Institute of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
16
|
Lin W, Chen Z, Mo X, Zhao S, Wen Z, Cheung WH, Fu D, Chen B. Phactr1 negatively regulates bone mass by inhibiting osteogenesis and promoting adipogenesis of BMSCs via RhoA/ROCK2. J Mol Histol 2021; 53:119-131. [PMID: 34709489 DOI: 10.1007/s10735-021-10031-z] [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: 08/19/2021] [Accepted: 10/13/2021] [Indexed: 12/01/2022]
Abstract
The imbalance between osteogenic and adipogenic differentiation of Bone marrow-derived mesenchymal stem cells (BMSCs) is involved in the occurrence and development of osteoporosis (OP). Previous studies have indicated the potential of phosphatase and actin regulator 1 (Phactr1) in regulating osteogenic and adipogenic differentiation of BMSCs. The present study aims to investigate the function and mechanism of Phactr1 in regulating osteogenic and adipogenic differentiation of BMSCs. Herein, the expression of Phactr1 in bone and adipose tissue of OP rats was determined by immunohistochemical. BMSCs were subjected to osteogenic and adipogenic differentiation, and transfected with Phactr1 overexpression lentivirus, small interference RNA (siRNA) and KD025 (selective ROCK2 inhibitor). The relationship between Phactr1 and ROCK2 was detected by Co-IP experiment. The expression of Phactr1, Runx2, C/EBPα, RhoA and ROCK2 was detected by Western blot. Calcium nodule and lipid droplets were determined by alizarin red and Oil red O staining. Interestingly, Phactr1 increased in both bone and adipose tissue of OP rats. During osteogenic differentiation, Phactr1 decreased and active RhoA, ROCK2 increased, while overexpression Phactr1 inhibits the increase of Runx2. Phactr1 increased and active RhoA decreased, ROCK2 did not changed during adipogenic differentiation. While, Knockdown Phactr1 inhibits the increase of C/EBPα. Phactr1 and ROCK2 were combined in osteogenic differentiation, but not in adipogenic differentiation. By using KD025, the decrease of Phactr1 and increase of Runx2 were inhibited respectively in osteogenic differentiation. Meanwhile, when ROCK2 was inhibited, Phactr1, C/EBPα were significantly increased in adipogenic differentiation. These findings indicated that Phactr1 negatively regulates bone mass by inhibiting osteogenesis and promoting adipogenesis of BMSCs by activating RhoA/ROCK2.
Collapse
Affiliation(s)
- Wei Lin
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zhipeng Chen
- Department of Orthopaedics, The Second Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyi Mo
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Shengli Zhao
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zhenxing Wen
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Wing Hoi Cheung
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Dan Fu
- Department of Orthopedic, Kiang Wu Hospital, Macau, 999078, China
| | - Bailing Chen
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China.
| |
Collapse
|
17
|
Zhou Y, Wei LL, Zhang RP, Han CW, Cao Y. Globular adiponectin inhibits osteoblastic differentiation of vascular smooth muscle cells through the PI3K/AKT and Wnt/β-catenin pathway. J Mol Histol 2021; 52:1067-1080. [PMID: 34398360 PMCID: PMC8487883 DOI: 10.1007/s10735-021-10012-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/10/2021] [Indexed: 01/20/2023]
Abstract
Lipid metabolism is closely related to the improvement of vascular calcification (VC) in chronic kidney disease (CKD). Globular adiponectin (gAd) has been reported to be involved in the development of VC in CKD, but the detailed regulatory role remains unclear. The present study is aimed to investigate the biological function and the underlying regulation mechanism of gAd in the process of VC during CKD. Vascular smooth muscle cells (VSMCs) calcification was determined by Alizarin Red S staining. Protein signaling related with VC was tested by western blotting. The expression and intracellular localization of runt-related transcription factor 2 (Runx2) was detected by immunofluorescence and uraemic rat with VC was established by a two-step nephrectomy. Combined with the results of Alizarin Red S staining, we discovered that β-glycerophosphate (β-Gp)-induced the osteoblastic differentiation of VSMCs was significantly reversed by gAd treatment. Along with the VSMCs calcification and the increase of Runx2 in β-Gp-exposed VSMCs, the activities of protein kinase B (AKT) and Wnt/β-catenin pathway were enhanced, but that were counteracted by the exposure of gAd in rat and human VSMCs. After administration with agonists of the Wnt (SKL2001) and AKT (SC79), there appeared more osteoblastic differentiation and higher expression of Runx2 in gAd-treated VSMCs, but showing lower impact in the presence of SC79 than that in the presence of SKL2001. In the in vivo experiments, intravenous injection of gAd also significantly inhibited VC and Runx2 level in uraemic rat in a dose-dependent manner, possibly through regulating Wnt/β-catenin pathway. This study demonstrates that gAd ameliorates osteoblastic differentiation of VSMCs possibly by blocking PI3K/AKT and Wnt/β-catenin signaling transduction. The findings provide an important foundation for gAd in treating VC in kidney diseases.
Collapse
Affiliation(s)
- Yun Zhou
- Laboratory Medicine, China-Japan Friendship Hospital, No.2 Yinghua Street, Chaoyang District, Beijing, 100029, China
| | - Li-Long Wei
- Laboratory Medicine, China-Japan Friendship Hospital, No.2 Yinghua Street, Chaoyang District, Beijing, 100029, China
| | - Rui-Ping Zhang
- Laboratory Medicine, China-Japan Friendship Hospital, No.2 Yinghua Street, Chaoyang District, Beijing, 100029, China
| | - Cheng-Wu Han
- Laboratory Medicine, China-Japan Friendship Hospital, No.2 Yinghua Street, Chaoyang District, Beijing, 100029, China
| | - Yongtong Cao
- Laboratory Medicine, China-Japan Friendship Hospital, No.2 Yinghua Street, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
18
|
Mollentze J, Durandt C, Pepper MS. An In Vitro and In Vivo Comparison of Osteogenic Differentiation of Human Mesenchymal Stromal/Stem Cells. Stem Cells Int 2021; 2021:9919361. [PMID: 34539793 PMCID: PMC8443361 DOI: 10.1155/2021/9919361] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/23/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
The use of stem cells in regenerative medicine, including tissue engineering and transplantation, has generated a great deal of enthusiasm. Mesenchymal stromal/stem cells (MSCs) can be isolated from various tissues, most commonly, bone marrow but more recently adipose tissue, dental pulp, and Wharton's jelly, to name a few. MSCs display varying phenotypic profiles and osteogenic differentiating capacity depending and their site of origin. MSCs have been successfully differentiated into osteoblasts both in vitro an in vivo but discrepancies exist when the two are compared: what happens in vitro does not necessarily happen in vivo, and it is therefore important to understand why these differences occur. The osteogenic process is a complex network of transcription factors, stimulators, inhibitors, proteins, etc., and in vivo experiments are helpful in evaluating the various aspects of this osteogenic process without distractions and confounding variables. With that in mind, the results of in vitro experiments need to be carefully considered and interpreted with caution as they do not perfectly replicate the conditions found within living organisms. This is where in vivo experiments help us better understand interactions that might occur in the osteogenic process that cannot be replicated in vitro. Potentially, these differences could also be exploited to develop an optimal MSC cell therapeutic product that can be used for bone disorders. There are many bone disorders, most of which cause a great deal of discomfort. Clinically acceptable protocols could be developed in which MSCs are used to aid in bone regeneration providing relief for patients with chronic pain. The aim of this review is to examine the differences between studies conducted in vitro and in vivo with regard to the osteogenic process to better define the gaps in current osteogenic research. By better understanding osteogenic differentiation, we can better define treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Jamie Mollentze
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology; SAMRC Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
19
|
Jung SJ, Choi YJ, Park TK, Woo SE, Kim BY, Yoon JS, Jang SY. Wnt signalling inhibits adipogenesis in orbital fibroblasts from patients with Graves' orbitopathy. Br J Ophthalmol 2021; 106:1019-1027. [PMID: 34193409 DOI: 10.1136/bjophthalmol-2020-316898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/07/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND/AIMS To investigate the role of Wnt signalling in adipogenesis using an in vitro model of Graves' orbitopathy (GO). METHODS Orbital fat was obtained from patients with GO and non-GO participants for primary orbital fibroblast (OF) culture. Expression levels of Wnt5a, Wnt10b, β-catenin, phospho-β-catenin and cyclin D1 were compared between GO and non-GO OFs. These expression levels were also determined during adipogenesis of GO and non-GO OFs. The effects of a stimulator and inhibitor of Wnt signalling on adipogenesis of GO and non-GO OFs were investigated. RESULTS Western blotting analysis showed significant reductions in β-catenin and cyclin D1 and significant enhancement of phospho-β-catenin in OFs from patients with GO, compared with OFs from non-GO participants (p<0.05). Expression of Wnt5a, Wnt10b, β-catenin and cyclin D1 in OFs was highest on day 0, and then gradually declined after induction of adipogenic differentiation. The expression levels of PPARγ, C/EBPα and C/EBPβ were reduced in Wnt stimulator-treated OFs in a dose-dependent manner. Oil red O staining confirmed that a stimulator of Wnt inhibited adipogenesis in GO OFs. CONCLUSION These results indicate that Wnt signalling inhibits adipogenesis in OFs from patients with GO and non-GO participants. Further studies are required to examine the potential of Wnt signalling as a target for therapeutic strategies.
Collapse
Affiliation(s)
- Sang Joon Jung
- Department of Ophthalmology, Korea Army Training Center, Republic of Korea Army, Nonsan-si, Republic of Korea
| | - Yeon Jeong Choi
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Tae Kwann Park
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Sang Earn Woo
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Bo-Yeon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Jin Sook Yoon
- Department of Ophthalmology, Severance Hospital, The Institute of Vision Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Young Jang
- Department of Ophthalmology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| |
Collapse
|
20
|
7-HYB, a Phenolic Compound Isolated from Myristica fragrans Houtt Increases Cell Migration, Osteoblast Differentiation, and Mineralization through BMP2 and β-catenin Signaling. Int J Mol Sci 2020; 21:ijms21218059. [PMID: 33137925 PMCID: PMC7663243 DOI: 10.3390/ijms21218059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022] Open
Abstract
The seeds (nutmegs) of Myristica fragrans Houtt have been used as popular spices and traditional medicine to treat a variety of diseases. A phenolic compound, ((7S)-8′-(benzo[3′,4′]dioxol-1′-yl)-7-hydroxypropyl)benzene-2,4-diol (7-HYB) was isolated from the seeds of M. fragrans. This study aimed to investigate the anabolic effects of 7-HYB in osteogenesis and bone mineralization. In the present study, 7-HYB promotes the early and late differentiation of MC3T3-E1 preosteoblasts. 7-HYB also elevated cell migration rate during differentiation of the preosteoblasts with the increased phosphorylation of mitogen-activated protein kinases (MAPKs) including ERK1/2, p38, and JNK. In addition, 7-HYB induced the protein level of BMP2, the phosphorylation of Smad1/5/8, and the expression of RUNX2. 7-HYB also inhibited GSK3β and subsequently increased the level of β-catenin. However, in bone marrow macrophages (BMMs), 7-HYB has no biological effects in cell viability, TRAP-positive multinuclear osteoclasts, and gene expression (c-Fos and NF-ATc1) in receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis. Our findings suggest that 7-HYB plays an important role in osteoblast differentiation through the BMP2 and β-catenin signaling pathway. It also indicates that 7-HYB might have a therapeutic effect for the treatment of bone diseases such as osteoporosis and periodontitis.
Collapse
|
21
|
Bertrand AA, Malapati SH, Yamaguchi DT, Lee JC. The Intersection of Mechanotransduction and Regenerative Osteogenic Materials. Adv Healthc Mater 2020; 9:e2000709. [PMID: 32940024 PMCID: PMC7864218 DOI: 10.1002/adhm.202000709] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/14/2020] [Indexed: 12/23/2022]
Abstract
Mechanical signals play a central role in cell fate determination and differentiation in both physiologic and pathologic circumstances. Such signals may be delivered using materials to generate discrete microenvironments for the purposes of tissue regeneration and have garnered increasing attention in recent years. Unlike the addition of progenitor cells or growth factors, delivery of a microenvironment is particularly attractive in that it may reduce the known untoward consequences of the former two strategies, such as excessive proliferation and potential malignant transformation. Additionally, the ability to spatially modulate the fabrication of materials allows for the creation of multiple microenvironments, particularly attractive for regenerating complex tissues. While many regenerative materials have been developed and tested for augmentation of specific cellular responses, the intersection between cell biology and material interactions have been difficult to dissect due to the complexity of both physical and chemical interactions. Specifically, modulating materials to target individual signaling pathways is an avenue of interdisciplinary research that may lead to a more effective method of optimizing regenerative materials. In this work, the aim is to summarize the major mechanotransduction pathways for osteogenic differentiation and to consolidate the known materials and material properties that activate such pathways.
Collapse
Affiliation(s)
- Anthony A. Bertrand
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Sri Harshini Malapati
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
| | - Dean T. Yamaguchi
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, California
| | - Justine C. Lee
- Division of Plastic and Reconstructive Surgery, University of California Los Angeles David Geffen School of Medicine, Los Angeles, California
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, California
- UCLA Molecular Biology Institute, Los Angeles, California
| |
Collapse
|
22
|
Li X, Ren G, Cai C, Yang X, Nie L, Jing X, Li C. TNF‑α regulates the osteogenic differentiation of bone morphogenetic factor 9 adenovirus‑transduced rat follicle stem cells via Wnt signaling. Mol Med Rep 2020; 22:3141-3150. [PMID: 32945435 PMCID: PMC7453510 DOI: 10.3892/mmr.2020.11439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022] Open
Abstract
Periodontitis is a chronic infectious disease that alters the cellular microenvironment and promotes bone absorption. Bone morphogenetic protein 9 (BMP9) serves an important role in proliferation and differentiation, and tumor necrosis factor‑alpha (TNF‑α) is an important contributor to bone resorption. The present study aimed to investigate the effect of osteogenic differentiation in the presence of BMP9 and TNF‑α in rat follicle stem cells (rDFCs). rDFCs were transfected with adenoviruses expressing BMP9 (AdBMP9) and the expression levels of important proteins [BMP9, β‑catenin, glycogen synthase kinase 3β (GSK3β), phosphorylated‑GSK3β, calcium/calmodulin dependent protein kinase II and nemo like kinase] were determined using western blotting. The effect of osteogenesis was analyzed using reverse transcription‑quantitative PCR, in addition to alkaline phosphatase, Alizarin Red S, and hematoxylin and eosin staining methods. The results of the present study revealed that TNF‑α activated the canonical Wnt signaling pathway and suppressed osteogenesis. High concentrations of Dickkopf 1 (DKK1) reduced the osteogenic differentiation of AdBMP9‑transduced rDFCs, whereas low concentrations of DKK1 promoted BMP9‑induced bone formation, which was discovered to partially act via the canonical and non‑canonical Wnt signaling pathways. In conclusion, the findings of the present study suggested that the enhanced promoting effect of BMP9 alongside the treatment with low concentrations of DKK1 may be useful for treating periodontitis bone absorption.
Collapse
Affiliation(s)
- Xinyue Li
- Department of Outpatients, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Ge Ren
- Department of Outpatients, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Changjun Cai
- Department of Outpatients, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Xia Yang
- Department of Outpatients, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Li Nie
- Department of Outpatients, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Xueqin Jing
- Department of Outpatients, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
| | - Conghua Li
- Department of Outpatients, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 401147, P.R. China
| |
Collapse
|
23
|
Generation of Inducible CRISPRi and CRISPRa Human Stromal/Stem Cell Lines for Controlled Target Gene Transcription during Lineage Differentiation. Stem Cells Int 2020; 2020:8857344. [PMID: 32922451 PMCID: PMC7453244 DOI: 10.1155/2020/8857344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/20/2020] [Accepted: 08/04/2020] [Indexed: 01/12/2023] Open
Abstract
Background Human bone marrow stromal/stem cells (hMSCs, also known as the skeletal stem cells or mesenchymal stem cells) are being employed to study lineage fate determination to osteoblasts, adipocytes, and chondrocytes. However, mechanistic studies employing hMSC have been hampered by the difficulty of deriving genetically modified cell lines due to the low and unstable transfection efficiency. Methods We infected hMSC with a CRISPR/Cas9 lentivirus system, with specific inducible dCas9-coupled transcription activator or repressor: dCas9-KRAB or dCas9-VP64, respectively, and established two hMSC lines (hMSC-CRISPRi and hMSC-CRISPRa) that can inhibit or activate gene expression, respectively. The two cell lines showed similar cell morphology, cell growth kinetics, and similar lineage differentiation potentials as the parental hMSC line. The expression of KRAB-dCas9 or VP64-dCas9 was controlled by the presence or absence of doxycycline (Dox) in the cell culturing medium. To demonstrate the functionality of the dCas9-effector hMSC system, we tested controlled expression of alkaline phosphatase (ALP) gene through transfection with the same single ALP sgRNA. Results In the presence of Dox, the expression of ALP showed 60-90% inhibition in hMSC-CRISPRi while ALP showed more than 20-fold increased expression in hMSC-CRISPRa. As expected, the ALP was functionally active and the cells showed evidence for inhibition or enhancement of in vitro osteoblast differentiation, respectively. Conclusion hMSC-CRISPRi and hMSC-CRISPRa are useful resources to study genes and genetic pathways regulating lineage-specific differentiation of hMSC.
Collapse
|
24
|
TMARg, a Novel Anthraquinone Isolated from Rubia cordifolia Nakai, Increases Osteogenesis and Mineralization through BMP2 and β-Catenin Signaling. Int J Mol Sci 2020; 21:ijms21155332. [PMID: 32727092 PMCID: PMC7432489 DOI: 10.3390/ijms21155332] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Plant extracts have long been regarded as useful medicines in the treatment of human diseases. Rubia cordifolia Nakai has been used as a traditional medicine, as it has pharmacological properties such as antioxidant and anti-inflammatory activity. However, the biological functions of TMARg, isolated from the roots of R. cordifolia, in osteoblast differentiation remain unknown. This study was performed to investigate the pharmacological effects and intracellular signaling of TMARg in the osteoblast differentiation of pre-osteoblast MC3T3-E1 cells and mesenchymal precursor C2C12 cells. Methods: Cell viability was evaluated using an MTT assay. Early and late osteoblast differentiation was examined by analyzing the activity of alkaline phosphatase (ALP), and by staining it with Alizarin red S (ARS). Cell migration was determined by using migration assays. Western blot analysis and immunocytochemical analysis were used to examine the intracellular signaling pathways and differentiation proteins. Results: In the present study, TMARg showed no cytotoxicity and increased the osteoblast differentiation in pre-osteoblasts, as assessed from the alkaline phosphate (ALP) staining and activity and ARS staining. TMARg also induced BMP2 expression and increased the p-smad1/5/8-RUNX2 and β-catenin pathways in both MC3T3-E1 and C2C12 cells. Furthermore, TMARg activated mitogen-activated protein kinases (MAPKs) and increased the cell migration rate. In addition, the TMARg-mediated osteoblast differentiation was suppressed by BMP and Wnt inhibitors with the downregulation of BMP2 expression. Conclusion: These findings demonstrate that TMARg exerts pharmacological and biological effects on osteoblast differentiation through the activation of BMP2 and β-catenin signaling pathways, and suggest that TMARg might be a potential phytomedicine for the treatment of bone diseases.
Collapse
|
25
|
Tencerova M, Frost M, Figeac F, Nielsen TK, Ali D, Lauterlein JJL, Andersen TL, Haakonsson AK, Rauch A, Madsen JS, Ejersted C, Højlund K, Kassem M. Obesity-Associated Hypermetabolism and Accelerated Senescence of Bone Marrow Stromal Stem Cells Suggest a Potential Mechanism for Bone Fragility. Cell Rep 2020; 27:2050-2062.e6. [PMID: 31091445 DOI: 10.1016/j.celrep.2019.04.066] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/06/2019] [Accepted: 04/12/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is associated with increased risk for fragility fractures. However, the cellular mechanisms are unknown. Using a translational approach combining RNA sequencing and cellular analyses, we investigated bone marrow stromal stem cells (BM-MSCs) of 54 men divided into lean, overweight, and obese groups on the basis of BMI. Compared with BM-MSCs obtained from lean, obese BM-MSCs exhibited a shift of molecular phenotype toward committed adipocytic progenitors and increased expression of metabolic genes involved in glycolytic and oxidoreductase activity. Interestingly, compared with paired samples of peripheral adipose tissue-derived stromal cells (AT-MSCs), insulin signaling of obese BM-MSCs was enhanced and accompanied by increased abundance of insulin receptor positive (IR+) and leptin receptor positive (LEPR+) cells in BM-MSC cultures. Their hyper-activated metabolic state was accompanied by an accelerated senescence phenotype. Our data provide a plausible explanation for the bone fragility in obesity caused by enhanced insulin signaling leading to accelerated metabolic senescence of BM-MSCs.
Collapse
Affiliation(s)
- Michaela Tencerova
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark.
| | - Morten Frost
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark
| | - Florence Figeac
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Tina Kamilla Nielsen
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Dalia Ali
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jens-Jacob Lindegaard Lauterlein
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Thomas Levin Andersen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark; Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Anders Kristian Haakonsson
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Alexander Rauch
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark
| | - Jonna Skov Madsen
- Institute of Regional Health Science, University of Southern Denmark, 5000 Odense C, Denmark; Department of Biochemistry and Immunology, Lillebaelt Hospital, 7100 Vejle, Denmark
| | - Charlotte Ejersted
- Department of Endocrinology, Odense University Hospital, 5000 Odense C, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Moustapha Kassem
- Department of Molecular Endocrinology, KMEB, University of Southern Denmark and Odense University Hospital, 5000 Odense C, Denmark; Department of Cellular and Molecular Medicine, DanStem (Danish Stem Cell Center), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
26
|
Hong F, Wu S, Zhang C, Li L, Chen J, Fu Y, Wang J. TRPM7 Upregulate the Activity of SMAD1 through PLC Signaling Way to Promote Osteogenesis of hBMSCs. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9458983. [PMID: 32596398 PMCID: PMC7294393 DOI: 10.1155/2020/9458983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/18/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
TRPM7 is a member of the transient receptor potential cation channel (TRP channel) subfamily M and possesses both an ion channel domain and a functional serine/threonine α-kinase domain. It has been proven to play an essential role in the osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs). However, the signaling pathway and molecular mechanism for TRPM7 in regulating osteogenic differentiation remain largely unknown. In this study, the potential role and mechanism of TRPM7 in the osteogenic differentiation of hBMSCs were investigated. The results showed that the expression of TRPM7 mRNA and protein increased, as did the osteogenic induction time. Upregulation or inhibition of TRPM7 could promote or inhibit the osteogenic differentiation of hBMSCs for 14 days. It was also found that the upregulation or inhibition of TRPM7 promoted or inhibited the activity of PLC and SMAD1, respectively, during osteogenic differentiation. PLC could promote osteogenic differentiation by upregulating the activity of SMAD1. However, inhibition of PLC alone could reduce the activity of SMAD1 but not inhibit completely the activation of SMAD1. Therefore, we inferred that it is an important signaling pathway for TRPM7 to upregulate the activity of SMAD1 through PLC and thereby promote the osteogenic differentiation of hBMSCs, but it is not a singular pathway. TRPM7 may also regulate the activation of SMAD1 through other ways, except for PLC, during osteogenic differentiation of hBMSCs.
Collapse
Affiliation(s)
- Fanfan Hong
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shali Wu
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Cui Zhang
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Liang Li
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jianling Chen
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yong Fu
- Department of ENT, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jinfu Wang
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of ENT, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Sangeetha KN, Vennila R, Secunda R, Sakthivel S, Pathak S, Jeswanth S, Surendran R. Functional variations between Mesenchymal Stem Cells of different tissue origins: A comparative gene expression profiling. Biotechnol Lett 2020; 42:1287-1304. [PMID: 32372268 DOI: 10.1007/s10529-020-02898-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 04/24/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Mesenchymal Stem Cells (MSCs), regardless of the tissue sources, are considered as excellent candidates for cellular therapy as they are immune-privileged cells containing a multitude of therapeutic functions that aid in tissue regeneration and repair. For the effective application of these cells in cell therapy, it is important to understand and characterize their biological functions. OBJECTIVES The present study attempts to characterize the variations in multipotent function such as cell surface antigen levels, proliferation, differentiation and stemness (pluripotency) potential of MSCs isolated from foetal [wharton's jelly (WJ), foetal and maternal side of placenta (PF and PM)] and adult tissue sources [bone marrow (BM) and adipose tissue (AT)] using gene expression by real time PCR (qRT-PCR). RESULTS Amongst the different tissue sources, PM, PF and AT-MSCs exhibited significant increase (p < 0.001, p < 0.001 and p < 0.01 respectively) in CD 73 expression and therefore could have a role in immunomodulation. WJ-MSCs exhibited superior proliferation potential based on growth curve, PCNA and Wnt gene expression. BM-MSCs were superior in exhibiting trilineage differentiation. Enhanced stemness potential (Oct 4 and Nanog) was observed for both BM and WJ-MSCs. In addition, BM and WJ-MSCs expressed high levels of CD 90 making them suitable in bone repair and regeneration. CONCLUSION Thus to conclude, out of the five different sources tested, BM an adult source and WJ-MSCs a foetal source were superior in exhibiting most of the biological functions indicating that these sources may be suitable candidates for cell repair and regeneration studies.
Collapse
Affiliation(s)
- K N Sangeetha
- Stem Cell Research Centre, Government Stanley Hospital, Chennai, Tamilnadu, 600001, India
| | | | - R Secunda
- Stem Cell Research Centre, Government Stanley Hospital, Chennai, Tamilnadu, 600001, India.
| | - S Sakthivel
- Stem Cell Research Centre, Government Stanley Hospital, Chennai, Tamilnadu, 600001, India
| | - Surajit Pathak
- Chettinad Academy of Research and Education, Chettinad Hospital & Research Institute, Chennai, Tamilnadu, India
| | - S Jeswanth
- Stem Cell Research Centre, Government Stanley Hospital, Chennai, Tamilnadu, 600001, India
| | - R Surendran
- Hepato-Pancreato-Biliary Centre for Surgery & Transplantation, MIOT International, Chennai, Tamilnadu, India
| |
Collapse
|
28
|
Yang F, Jia Y, Sun Q, Zheng C, Liu C, Wang W, Du L, Kang S, Niu X, Li J. Raloxifene improves TNF-α-induced osteogenic differentiation inhibition of bone marrow mesenchymal stem cells and alleviates osteoporosis. Exp Ther Med 2020; 20:309-314. [PMID: 32550885 PMCID: PMC7296296 DOI: 10.3892/etm.2020.8689] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/09/2019] [Indexed: 01/01/2023] Open
Abstract
Effect of raloxifene (RLF) on the improvement of inhibited osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) resulted from tumor necrosis factor-α (TNF-α) induction, thus alleviating the progression of osteoporosis (OP), was investigated. An in vivo OP rat model was constructed by performing the procedures of ovariectomy (OVX). Rats were randomly divided into sham group, OVX group and RLF+OVX group. BMSCs were extracted from healthy rats, and randomly divided into control group, TNF-α group, RLF group and TNF-α+RLF group. Viability and cellular calcification ability in each group were detected. The relative levels of osteocalcin (OCN), Runx2 and NF-κB in cells with different treatments were determined. The body weight of rats in the OVX group and RLF+OVX group gradually increased compared with that in the sham group on the 8th week. No significant difference in body weight was observed between the rats of the OVX group and RLF+OVX group. Bone metabolism index (BMD) in the rats of the RLF+OVX group was higher than that of the OVX group, and lower compared with that of the sham group. Compared with the sham group, the elastic/max radial degree and elastic/max load of femora were reduced in the OVX group and RLF+OVX group, especially in the OVX group. The relative levels of OCN and Runx2, as well as the ALP activity and calcification ability, were decreased in the OVX group compared with the sham group, and the effect was partially reversed by the RLF treatment. After osteogenic differentiation of BMSCs, the viability and calcification ability were markedly reduced in TNF-α group, which was reversed by RLF treatment. Moreover, TNF-α induction downregulated the relative levels of OCN and Runx2, and RLF treatment could enhance their levels. The upregulated NF-κB protein level, induced by TNF-α, was reduced after RLF treatment. TNF-α induction inhibits osteogenic differentiation of BMSCs, which could be remarkably alleviated by RLF. It is suggested that RLF contributes to the alleviation of OP progression.
Collapse
Affiliation(s)
- Fenghe Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China.,Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Yusong Jia
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Qi Sun
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Chenying Zheng
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Chuyin Liu
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Wei Wang
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Li Du
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Shengqian Kang
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| | - Xufeng Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, P.R. China.,Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing 100083, P.R. China
| | - Jinyu Li
- Department of Orthopaedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, P.R. China
| |
Collapse
|
29
|
Zhu Y, Zhang X, Gu R, Liu X, Wang S, Xia D, Li Z, Lian X, Zhang P, Liu Y, Zhou Y. LAMA2 regulates the fate commitment of mesenchymal stem cells via hedgehog signaling. Stem Cell Res Ther 2020; 11:135. [PMID: 32213190 PMCID: PMC7093965 DOI: 10.1186/s13287-020-01631-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone defects are a common clinical condition that has gained an increasing amount of attention in recent years. Causes of bone defect include tumors, inflammation, and fractures. Bone tissue engineering is a novel treatment of bone defect, and human mesenchymal stem cells (hMSCs) are the ideal seed cells for bone tissue engineering due to their multi-lineage differentiation potential and immunogenicity. The laminin α2 (LAMA2) gene encodes the α2 subunit of laminins. Mutations in this gene have been reported to cause muscular dystrophy, but thus far no studies have elucidated the role of LAMA2 in the fate choices of MSCs. Here, we aimed to investigate the critical role of LAMA2 in the osteogenesis and adipogenesis of mesenchymal stem cells (MSCs). METHODS We investigated LAMA2 function in osteogenic and adipogenic differentiation of MSCs in vitro and in vivo through loss- and gain-of-function experiments. In addition, molecular mechanism was clarified by Western blot and siRNA. RESULTS Our results demonstrated that LAMA2 was a critical regulator for fate commitment of MSCs. Both in vitro and in vivo studies indicate that LAMA2 inhibits osteogenesis and promotes adipogenesis. Mechanistically, we found that LAMA2 regulated osteogenesis and adipogenesis of MSCs by modulating the hedgehog signaling pathway. CONCLUSIONS The present work confirms that LAMA2 is a new molecular target for MSC-based bone regeneration.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Ranli Gu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Xuenan Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Siyi Wang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Dandan Xia
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, 100871, People's Republic of China
| | - Zheng Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Xiaomin Lian
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China.
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China.
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, National Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, National Clinical Research Center for Oral Diseases, 22 Zhongguancun South Avenue, Beijing, 100081, People's Republic of China
| |
Collapse
|
30
|
Metabolic programming determines the lineage-differentiation fate of murine bone marrow stromal progenitor cells. Bone Res 2019; 7:35. [PMID: 31754546 PMCID: PMC6856123 DOI: 10.1038/s41413-019-0076-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/22/2019] [Accepted: 08/18/2019] [Indexed: 12/30/2022] Open
Abstract
Enhanced bone marrow adipogenesis and impaired osteoblastogenesis have been observed in obesity, suggesting that the metabolic microenvironment regulates bone marrow adipocyte and osteoblast progenitor differentiation fate. To determine the molecular mechanisms, we studied two immortalized murine cell lines of adipocyte or osteoblast progenitors (BMSCsadipo and BMSCsosteo, respectively) under basal and adipogenic culture conditions. At baseline, BMSCsadipo, and BMSCsosteo exhibit a distinct metabolic program evidenced by the presence of specific global gene expression, cellular bioenergetics, and metabolomic signatures that are dependent on insulin signaling and glycolysis in BMSCsosteo versus oxidative phosphorylation in BMSCsadipo. To test the flexibility of the metabolic program, we treated BMSCsadipo with parathyroid hormone, S961 (an inhibitor of insulin signaling) and oligomycin (an inhibitor of oxidative phosphorylation). The treatment induced significant changes in cellular bioenergetics that were associated with decreased adipocytic differentiation. Similarly, 12 weeks of a high-fat diet in mice led to the expansion of adipocyte progenitors, enhanced adipocyte differentiation and insulin signaling in cultured BMSCs. Our data demonstrate that BMSC progenitors possess a distinct metabolic program and are poised to respond to exogenous metabolic cues that regulate their differentiation fate.
Collapse
|
31
|
Notch Signaling Inhibition by LY411575 Attenuates Osteoblast Differentiation and Decreased Ectopic Bone Formation Capacity of Human Skeletal (Mesenchymal) Stem Cells. Stem Cells Int 2019; 2019:3041262. [PMID: 31534459 PMCID: PMC6724428 DOI: 10.1155/2019/3041262] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 06/23/2019] [Indexed: 11/19/2022] Open
Abstract
Background Chemical biology approaches using small molecule inhibitors targeting specific signaling pathways are useful tools to dissect the molecular mechanisms governing stem cell differentiation and for their possible use in therapeutic interventions. Methods Stem cell signaling small molecule library functional screen was performed employing human bone marrow skeletal (mesenchymal) stem cells (hBMSCs). Alkaline phosphatase (ALP) activity and formation of mineralized matrix visualized by Alizarin red staining were employed as markers for osteoblastic differentiation. Global gene expression profiling was conducted using the Agilent microarray platform, and data normalization and bioinformatics were performed using GeneSpring software. Pathway analyses were conducted using the Ingenuity Pathway Analysis (IPA) tool. In vivo ectopic bone formation was performed using hBMSC mixed with hydroxyapatite–tricalcium phosphate granules that were implanted subcutaneously in 8-week-old female nude mice. Hematoxylin and eosin staining and Sirius red staining were performed to identify bone formation in vivo. Results Among the tested molecules, LY411575, a potent γ-secretase and Notch signaling inhibitor, exhibited significant inhibitory effects on osteoblastic differentiation of hBMSCs manifested by reduced ALP activity, mineralized matrix formation, and decreased osteoblast-specific gene expression as well as in vivo ectopic bone formation. Global gene expression profiling of LY411575-treated cells revealed changes in multiple signaling pathways, including focal adhesion, insulin, TGFβ, IL6, and Notch signaling, and decreased the expression of genes associated with functional categories of tissue development. Among the affected signaling networks were TGFβ1, SPP1, and ERK regulatory networks. Conclusions We identified γ-secretase inhibitor (LY411575) as a potent regulator of osteoblastic differentiation of hBMSC that may be useful as a therapeutic option for treating conditions associated with ectopic bone formation.
Collapse
|
32
|
Esche J, Shi L, Hartmann MF, Schönau E, Wudy SA, Remer T. Glucocorticoids and Body Fat Inversely Associate With Bone Marrow Density of the Distal Radius in Healthy Youths. J Clin Endocrinol Metab 2019; 104:2250-2256. [PMID: 30715368 DOI: 10.1210/jc.2018-02108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/25/2019] [Indexed: 01/28/2023]
Abstract
CONTEXT Elevated bone marrow adipose tissue (BMAT) is associated with lower bone quality, higher fracture rates, and an unfavorable overall metabolic profile. Apart from age, particularly glucocorticoids (GC), body fat, and diet are discussed to influence BMAT. We hypothesized that already in healthy youths, higher fat intake, higher fat mass index (FMI), and higher GC secretion, still within the normal range, may associate with increased BMAT. DESIGN In a subsample of healthy 6- to 18-year-old participants of the Dortmund Nutritional and Anthropometric Longitudinally Designed Study, peripheral quantitative CT of the nondominant proximal forearm was used to determine bone marrow density of the distal radius as an inverse surrogate parameter for BMAT. In those participants (n = 172) who had collected two, 24-hour urines within around one year before bone measurement, major urinary GC metabolites were measured by gas chromatography-mass spectrometry and summed up to assess daily adrenal GC secretion (ΣC21). Dietary intake was assessed by 3-day weighed dietary records. FMI was anthropometrically calculated. Separate multiple linear regression models were used to analyze the relationships of ΣC21, FMI, and fat intake with BMAT. RESULTS After controlling for confounders, such as age, energy intake, and forearm muscle area, ΣC21 (β = -0.042) and FMI (β = -0.002) showed inverse relationships with bone marrow density (P < 0.05), whereas fat intake did not associate significantly. CONCLUSION Our results indicate that already a moderately elevated GC secretion and higher body fatness during adolescence may adversely impact BMAT, an indicator for long-term bone health.
Collapse
Affiliation(s)
- Jonas Esche
- Dortmund Nutritional and Anthropometric Longitudinally Designed Study Center, Institute of Nutrition and Food Science, Nutritional Epidemiology, University of Bonn, Dortmund, Germany
| | - Lijie Shi
- Dortmund Nutritional and Anthropometric Longitudinally Designed Study Center, Institute of Nutrition and Food Science, Nutritional Epidemiology, University of Bonn, Dortmund, Germany
| | - Michaela F Hartmann
- Steroid Research and Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Justus-Liebig-University, Giessen, Germany
| | - Eckhard Schönau
- Children's Hospital, University of Cologne, Cologne, Germany
| | - Stefan A Wudy
- Steroid Research and Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Justus-Liebig-University, Giessen, Germany
| | - Thomas Remer
- Dortmund Nutritional and Anthropometric Longitudinally Designed Study Center, Institute of Nutrition and Food Science, Nutritional Epidemiology, University of Bonn, Dortmund, Germany
| |
Collapse
|
33
|
Liang X, Hou Z, Xie Y, Yan F, Li S, Zhu X, Cai L. Icariin promotes osteogenic differentiation of bone marrow stromal cells and prevents bone loss in OVX mice via activating autophagy. J Cell Biochem 2019; 120:13121-13132. [PMID: 30887562 DOI: 10.1002/jcb.28585] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/27/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Xiaoxiao Liang
- Department of Orthopaedic Surgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Zhiqiang Hou
- Department of Orthopaedic Surgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Yuanlong Xie
- Department of Orthopaedic Surgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Feifei Yan
- Department of Orthopaedic Surgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Sisi Li
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science Huazhong University of Science and Technology Wuhan China
| | - Xiaobin Zhu
- Department of Orthopaedic Surgery Zhongnan Hospital of Wuhan University Wuhan China
| | - Lin Cai
- Department of Orthopaedic Surgery Zhongnan Hospital of Wuhan University Wuhan China
| |
Collapse
|
34
|
Zeng Q, Wang Y, Gao J, Yan Z, Li Z, Zou X, Li Y, Wang J, Guo Y. miR-29b-3p regulated osteoblast differentiation via regulating IGF-1 secretion of mechanically stimulated osteocytes. Cell Mol Biol Lett 2019; 24:11. [PMID: 30915127 PMCID: PMC6416934 DOI: 10.1186/s11658-019-0136-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022] Open
Abstract
Background Mechanical loading is an essential factor for bone formation. A previous study indicated that mechanical tensile strain of 2500 microstrain (με) at 0.5 Hz for 8 h promoted osteogenesis and corresponding mechanoresponsive microRNAs (miRs) were identified in osteoblasts. However, in osteocytes, it has not been identified which miRs respond to the mechanical strain, and it is not fully understood how the mechanoresponsive miRs regulate osteoblast differentiation. Methods Mouse MLO-Y4 osteocytes were applied to the same mechanical tensile strain in vitro. Using molecular and biochemical methods, IGF-1 (insulin-like growth factor-1), PGE2 (prostaglandin E2), OPG (osteoprotegerin) and NOS (nitric oxide synthase) activities of the cells were assayed. MiR microarray and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assays were applied to select and validate differentially expressed miRs, and the target genes of these miRs were then predicted. MC3T3-E1 osteoblasts were stimulated by the mechanical tensile strain, and the miR-29b-3p expression was detected with miR microarray and RT-qPCR. Additionally, the effect of miR-29b-3p on IFG-1 secretion of osteocytes and the influence of conditioned medium of osteocytes transfected with miR-29b-3p on osteoblast differentiation were investigated. Results The mechanical strain increased secretions of IGF-1 and PGE2, elevated OPG expression and NOS activities, and resulted in altered expression of the ten miRs, and possible target genes for these differentially expressed miRs were revealed through bioinformatics. Among the ten miRs, miR-29b-3p were down-regulated, and miR-29b-3p overexpression decreased the IGF-1 secretion of osteocytes. The mechanical strain did not change expression of osteoblasts' miR-29b-3p. In addition, the conditioned medium of mechanically strained osteocytes promoted osteoblast differentiation, and the conditioned medium of osteocytes transfected with miR-29b-3p mimic inhibited osteoblast differentiation. Conclusions In osteocytes (but not osteoblasts), miR-29b-3p was responsive to the mechanical tensile strain and regulated osteoblast differentiation via regulating IGF-1 secretion of mechanically strained osteocytes.
Collapse
Affiliation(s)
- Qiangcheng Zeng
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China
| | - Yang Wang
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China.,3Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044 China
| | - Jie Gao
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China.,Medical Department, Secondary Renmin Hospital of Dezhou, Dezhou, 253023 Shangdong China
| | - Zhixiong Yan
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Zhenghua Li
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China
| | - Xianqiong Zou
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Yanan Li
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Jiahui Wang
- 2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| | - Yong Guo
- 1key laboratory of Functional Bioresource Utilization in University of Shandong, Shandong Key Laboratory of Biophysics, Dezhou University, Dezhou, 253023 China.,2Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin City, 541100 Guangxi China
| |
Collapse
|
35
|
Ali H, Zmuda JM, Cvejkus RK, Kershaw EE, Kuipers AL, Oczypok EA, Wheeler V, Bunker CH, Miljkovic I. Wnt Pathway Inhibitor DKK1: A Potential Novel Biomarker for Adiposity. J Endocr Soc 2019; 3:488-495. [PMID: 30746507 PMCID: PMC6364619 DOI: 10.1210/js.2018-00325] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/28/2018] [Indexed: 12/25/2022] Open
Abstract
Emerging evidence indicates that ectopic skeletal muscle adiposity may be a risk factor for type 2 diabetes (T2D), especially in persons of African ancestry. In vitro studies suggest that a Wnt pathway inhibitor, Dickkopf-related protein 1 (DKK1), plays a role in adiposity regulation and could be a biomarker for adiposity in humans. The objective of this study was to test whether serum DKK1 levels relate to adiposity measures in a cohort from an African ancestry population at high risk for T2D. Fasting serum DKK1 was measured in a sample of 159 men of African ancestry aged ≥40 years (mean age ± SD, 63.5 ± 8.2 years; mean body mass index, 27.8 ± 4.5 kg/m2). Anthropometrics included total-body and trunk adiposity measured by dual-energy x-ray absorptiometry and lower-leg skeletal muscle density measured by CT [which reflects the intramuscular adiposity content (mg/cm3)]. Serum DKK1 was positively correlated with BMI (r = 0.20; P = 0.01), waist circumference (r = 0.15; P = 0.046), DXA total-body adiposity (r = 0.24; P = 0.003), and DXA trunk adiposity (r = 0.21; P = 0.009), independent of age and height. In addition, serum DKK1 was inversely correlated with skeletal muscle density (r = −0.25; P = 0.002), independent of age, BMI, and calf muscle area. No significant correlation was found between serum DKK1 and fasting serum glucose or insulin levels or insulin resistance estimated by homeostasis model assessment. These findings suggest that higher levels of serum DKK1 may be associated with greater overall, central, and ectopic skeletal muscle adiposity. Further studies are needed to unravel the potential role of DKK1 in the regulation of adiposity in humans.
Collapse
Affiliation(s)
- Hira Ali
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph M Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ryan K Cvejkus
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Allison L Kuipers
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Elizabeth A Oczypok
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Victor Wheeler
- Tobago Health Studies Office, Scarborough, Tobago, Trinidad & Tobago, West Indies
| | - Clareann H Bunker
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Iva Miljkovic
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Artsi H, Gurt I, El-Haj M, Müller R, Kuhn GA, Ben Shalom G, Cohen-Kfir E, Abramowitz E, Kandel L, Safran O, Dresner-Pollak R. Sirt1 Promotes a Thermogenic Gene Program in Bone Marrow Adipocytes: From Mice to (Wo)Men. Front Endocrinol (Lausanne) 2019; 10:126. [PMID: 30873124 PMCID: PMC6403178 DOI: 10.3389/fendo.2019.00126] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/11/2019] [Indexed: 12/22/2022] Open
Abstract
Bone marrow adipose tissue (MAT) is influenced by nutritional cues, and participates in whole body energy metabolism. To investigate the role of Sirtuin1 (Sirt1), a key player in metabolism, in MAT, marrow adiposity was evaluated in inbred 5-month-old 129/Sv Sirt1 haplo-insufficient (Sirt1 Δ/+) and wild type (WT) mice. Decreased expression of the thermogenic genes: Prdm16, Pgc1α, Foxc2, Dio2, and β3AR was detected in whole tibiae derived from Sirt1 Δ/+ compared to WT female mice. Similarly, decreased expression of Prdm16 and Pgc1α was observed in primary bone marrow mesenchymal stem cell (BM-MSC) cultures obtained from Sirt1 Δ/+ compared to WT female mice, suggesting a cell autonomous effect of Sirt1 in BM-MSCs. In vitro, Sirt1 over-expression in the mesenchymal embryonic fibroblast stem cell line C3HT101/2 increased Pgc1α and Prdm16 protein level. Similarly, pharmacologic activation of Sirt1 by SRT3025 increased Foxc2, Pgc1α, Dio2, Tfam, and Cyc1 expression while inhibition of Sirt1 by EX527 down-regulated UCP1 in C3HT101/2 cells. Importantly, in human femoral BM-MSCs obtained from female patients undergoing hip operations for fracture or osteoarthritis, Sirt1 activation by SRT3025 increased PGC1α mRNA and protein level. Blocking sclerostin, an inhibitor of the WNT pathway and a Sirt1 target, by the monoclonal humanized antibody (Sc-AbII), stimulated β3AR, PRDM16, and UCP1 gene expression, and increased PGC1α protein level. These results show that Sirt1 stimulates a thermogenic gene program in marrow adipocytes in mice and humans via PGC1α activation and sclerostin inhibition. The implications of these findings to bone health, hematopoiesis and whole body energy metabolism remain to be investigated.
Collapse
Affiliation(s)
- Hanna Artsi
- Division of Medicine, Department of Endocrinology and Metabolism, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Irina Gurt
- Division of Medicine, Department of Endocrinology and Metabolism, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Madi El-Haj
- Department of Orthopedics, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ralph Müller
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Gisela A. Kuhn
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Gal Ben Shalom
- Division of Medicine, Department of Endocrinology and Metabolism, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Einav Cohen-Kfir
- Division of Medicine, Department of Endocrinology and Metabolism, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eva Abramowitz
- Division of Medicine, Department of Endocrinology and Metabolism, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leonid Kandel
- Department of Orthopedics, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ori Safran
- Department of Orthopedics, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rivka Dresner-Pollak
- Division of Medicine, Department of Endocrinology and Metabolism, Faculty of Medicine, The Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
- *Correspondence: Rivka Dresner-Pollak
| |
Collapse
|
37
|
Li Y, Meng Y, Yu X. The Unique Metabolic Characteristics of Bone Marrow Adipose Tissue. Front Endocrinol (Lausanne) 2019; 10:69. [PMID: 30800100 PMCID: PMC6375842 DOI: 10.3389/fendo.2019.00069] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/24/2019] [Indexed: 02/05/2023] Open
Abstract
Bone marrow adipose tissue (MAT) is distinct from white adipose tissue (WAT) or brown adipose tissue (BAT) for its location, feature and function. As a largely ignored adipose depot, it is situated in bone marrow space and resided with bone tissue side-by-side. MAT is considered not only as a regulator of bone metabolism through paracrine, but also as a functionally particular adipose tissue that may contribute to global metabolism. Adipokines, inflammatory factors and other molecules derived from bone marrow adipocytes may exert systematic effects. In this review, we summary the evidence from several aspects including development, distribution, histological features and phenotype to elaborate the basic characteristics of MAT. We discuss the association between bone metabolism and MAT, and highlight our current understanding of this special adipose tissue. We further demonstrate the probable relationship between MAT and energy metabolism, as well as glucose metabolism. On the basis of preliminary results from animal model and clinical studies, we propose that MAT has its unique secretory and metabolic function, although there is no in-depth study at present.
Collapse
Affiliation(s)
- Yujue Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Meng
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xijie Yu ;
| |
Collapse
|
38
|
Woods GN, Ewing SK, Sigurdsson S, Kado DM, Ix JH, Hue TF, Eiriksdottir G, Xu K, Gudnason V, Lang TF, Vittinghoff E, Harris TB, Rosen CJ, Li X, Schwartz AV. Chronic Kidney Disease Is Associated With Greater Bone Marrow Adiposity. J Bone Miner Res 2018; 33:2158-2164. [PMID: 30075054 PMCID: PMC6702945 DOI: 10.1002/jbmr.3562] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 01/06/2023]
Abstract
Bone marrow adiposity is associated with aging, osteoporosis, and reduced hematopoiesis, as well as anorexia nervosa, but little is known about the underlying mechanisms that affect marrow adiposity. Chronic kidney disease (CKD) may influence bone marrow adipose tissue (BMAT), possibly through loss of lean mass or higher circulating levels of sclerostin. To test these hypotheses, we investigated the cross-sectional association between estimated glomerular filtration rate (eGFR) as a measure of kidney function and 1 H-MRS-based measurement of vertebral BMAT (L1 to L4) in 475 older adults from the Age Gene/Environment Susceptibility (AGES)-Reykjavik study. Mean BMAT was compared in those with eGFR >60 (n = 297) versus those with eGFR 45 to 60 (n = 120) or eGFR <45 (n = 58) using linear regression models. Participants had a mean age of 81.5 (SD 4.1) years, mean eGFR of 64.3 (SD 16.1) mL/min/1.734 cm2 , mean BMAT of 54.5% (SD 8.5); 48.2% were women. In unadjusted and adjusted models (age, visit window, gender, diabetes and visceral adipose tissue), BMAT was higher in those with eGFR <45 (adjusted mean 58.5%; 95% CI, 56.2 to 60.7) compared with those with eGFR >60 (adjusted mean 53.8%; 95% CI, 52.8 to 54.8) (p = 0.0002). BMAT did not differ in those with eGFR 45 to 60 (adjusted mean 54.3%; 95% CI, 52.8 to 55.9) compared with those with eGFR >60 (p = 0.58). In a subgroup of participants with serum sclerostin available (n = 253), additional adjustment for sclerostin attenuated the difference in adjusted mean vertebral BMAT between those with eGFR <45 versus >60 from 3.7% (p = 0.04) to 2.4% (p = 0.20). CKD stage 3b or worse was associated with greater bone marrow adiposity; this association may be partially mediated by sclerostin. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Gina N Woods
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,VA San Diego Healthcare System, San Diego, CA, USA
| | - Susan K Ewing
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | | | - Deborah M Kado
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,Department of Family Medicine & Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Joachim H Ix
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,VA San Diego Healthcare System, San Diego, CA, USA
| | - Trisha F Hue
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | | | - Kaipin Xu
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Thomas F Lang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Eric Vittinghoff
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Tamara B Harris
- Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | | | - Xiaojuan Li
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, USA
| | - Ann V Schwartz
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| |
Collapse
|
39
|
AlMuraikhi N, Ali D, Alshanwani A, Vishnubalaji R, Manikandan M, Atteya M, Siyal A, Alfayez M, Aldahmash A, Kassem M, Alajez NM. Stem cell library screen identified ruxolitinib as regulator of osteoblastic differentiation of human skeletal stem cells. Stem Cell Res Ther 2018; 9:319. [PMID: 30463599 PMCID: PMC6249887 DOI: 10.1186/s13287-018-1068-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/18/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
Background Better understanding of the signaling pathways that regulate human bone marrow stromal stem cell (hBMSC) differentiation into bone-forming osteoblasts is crucial for their clinical use in regenerative medicine. Chemical biology approaches using small molecules targeting specific signaling pathways are increasingly employed to manipulate stem cell differentiation fate. Methods We employed alkaline phosphatase activity and staining assays to assess osteoblast differentiation and Alizarin R staining to assess mineralized matrix formation of cultured hBMSCs. Changes in gene expression were assessed using an Agilent microarray platform, and data normalization and bioinformatics were performed using GeneSpring software. For in vivo ectopic bone formation experiments, hMSCs were mixed with hydroxyapatite–tricalcium phosphate granules and implanted subcutaneously into the dorsal surface of 8-week-old female nude mice. Hematoxylin and eosin staining and Sirius Red staining were used to detect bone formation in vivo. Results We identified several compounds which inhibited osteoblastic differentiation of hMSCs. In particular, we identified ruxolitinib (INCB018424) (3 μM), an inhibitor of JAK-STAT signaling that inhibited osteoblastic differentiation and matrix mineralization of hMSCs in vitro and reduced ectopic bone formation in vivo. Global gene expression profiling of ruxolitinib-treated cells identified 847 upregulated and 822 downregulated mRNA transcripts, compared to vehicle-treated control cells. Bioinformatic analysis revealed differential regulation of multiple genetic pathways, including TGFβ and insulin signaling, endochondral ossification, and focal adhesion. Conclusions We identified ruxolitinib as an important regulator of osteoblast differentiation of hMSCs. It is plausible that inhibition of osteoblast differentiation by ruxolitinib may represent a novel therapeutic strategy for the treatment of pathological conditions caused by accelerated osteoblast differentiation and mineralization. Electronic supplementary material The online version of this article (10.1186/s13287-018-1068-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nihal AlMuraikhi
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia
| | - Dalia Ali
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia.,Molecular Endocrinology Unit (KMEB), Department of Endocrinology, University Hospital of Odense and University of Southern Denmark, Odense, Denmark
| | - Aliah Alshanwani
- Department of Physiology, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia
| | - Radhakrishnan Vishnubalaji
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia
| | - Muthurangan Manikandan
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia
| | - Muhammad Atteya
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia.,Histology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Abdulaziz Siyal
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia
| | - Musaad Alfayez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia
| | - Abdullah Aldahmash
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia.,Prince Naif Health Research Center, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia
| | - Moustapha Kassem
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia.,Molecular Endocrinology Unit (KMEB), Department of Endocrinology, University Hospital of Odense and University of Southern Denmark, Odense, Denmark.,Department of Cellular and Molecular Medicine, Danish Stem Cell Center (DanStem), University of Copenhagen, 2200, Copenhagen, Denmark
| | - Nehad M Alajez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, 11461, Kingdom of Saudi Arabia. .,Cancer Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar.
| |
Collapse
|
40
|
Chen D, Xie R, Shu B, Landay AL, Wei C, Reiser J, Spagnoli A, Torquati A, Forsyth CB, Keshavarzian A, Sumner DR. Wnt signaling in bone, kidney, intestine, and adipose tissue and interorgan interaction in aging. Ann N Y Acad Sci 2018; 1442:48-60. [PMID: 30101565 DOI: 10.1111/nyas.13945] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/02/2018] [Accepted: 07/11/2018] [Indexed: 12/12/2022]
Abstract
Over the last two decades, it has become increasingly apparent that Wnt signaling plays a critical role in development and adult tissue homeostasis in multiple organs and in the pathogenesis of many diseases. In particular, a crucial role for Wnt signaling in bone development and bone tissue homeostasis has been well recognized. Numerous genome-wide association studies confirmed the importance of Wnt signaling in controlling bone mass. Moreover, ample evidence suggests that Wnt signaling is essential for kidney, intestine, and adipose tissue development and homeostasis. Recent emerging evidence demonstrates that Wnt signaling may play a fundamental role in the aging process of those organs. New discoveries show that bone is not only the major reservoir for calcium and phosphate storage, but also the largest organ with multiple functions, including mineral and energy metabolism. The interactions among bone, kidney, intestine, and adipose tissue are controlled and regulated by several endocrine signals, including FGF23, klotho, sclerostin, osteocalcin, vitamin D, and leptin. Since the aging process is characterized by structural and functional decline in almost all tissues and organs, understanding the Wnt signaling-related interactions among bone, kidney, intestine, and adipose tissue in aging may shed light on the pathogenesis of age-related diseases.
Collapse
Affiliation(s)
- Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois
| | - Rong Xie
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois
| | - Bing Shu
- Spine Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Alan L Landay
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, Illinois
| | - Changli Wei
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Jochen Reiser
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Anna Spagnoli
- Department of Pediatrics, Rush University Medical Center, Chicago, Illinois
| | - Alfonso Torquati
- Department of Surgery, Rush University Medical Center, Chicago, Illinois
| | | | - Ali Keshavarzian
- Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - D Rick Sumner
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
41
|
Clonal Analysis Delineates Transcriptional Programs of Osteogenic and Adipogenic Lineages of Adult Mouse Skeletal Progenitors. Stem Cell Reports 2018; 11:212-227. [PMID: 29937146 PMCID: PMC6067065 DOI: 10.1016/j.stemcr.2018.05.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/23/2022] Open
Abstract
Bone, cartilage, and marrow adipocytes are generated by skeletal progenitors, but the relationships between lineages and mechanisms controlling their differentiation are poorly understood. We established mouse clonal skeletal progenitors with distinct differentiation properties and analyzed their transcriptome. Unipotent osteogenic and adipogenic cells expressed specific transcriptional programs, whereas bipotent clones combined expression of those genes and did not show a unique signature. We tested potential regulators of lineage commitment and found that in the presence of interferon-γ (IFNγ) adipogenic clones can be induced to osteogenesis and that their adipogenic capacity is inhibited. Analysis of IFNγ-regulated genes showed that lineage signatures and fate commitment of skeletal progenitors were controlled by EGR1 and EGR2. Knockdown experiments revealed that EGR1 is a positive regulator of the adipogenic transcriptional program and differentiation capacity, whereas EGR2 inhibits the osteogenic program and potency. Therefore, our work revealed transcriptional signatures of osteogenic and adipogenic lineages and mechanism triggering cell fate. Bone marrow osteo- and adipogenic progenitors have specific transcriptional profiles Bipotent progenitors combine expression of osteogenic and adipogenic programs IFNγ inhibits adipogenesis and induces osteogenesis via downregulation of Egr1/Egr2 Egr1 maintains adipogenic and Egr2 suppresses osteogenic lineage commitment
Collapse
|
42
|
Romidepsin Promotes Osteogenic and Adipocytic Differentiation of Human Mesenchymal Stem Cells through Inhibition of Histondeacetylase Activity. Stem Cells Int 2018; 2018:2379546. [PMID: 29731773 PMCID: PMC5872662 DOI: 10.1155/2018/2379546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/13/2017] [Accepted: 12/31/2017] [Indexed: 12/17/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are adult multipotent stem cells that can differentiate into mesodermal lineage cells, including adipocytes and osteoblasts. However, the epigenetic mechanisms governing the lineage-specific commitment of BMSCs into adipocytes or osteoblasts are under investigation. Herein, we investigated the epigenetic effect of romidepsin, a small molecule dual inhibitor targeting HDAC1 and HDAC2 identified through an epigenetic library functional screen. BMSCs exposed to romidepsin (5 nM) exhibited enhanced adipocytic and osteoblastic differentiation. Global gene expression and signaling pathway analyses of differentially expressed genes revealed a strong enrichment of genes involved in adipogenesis and osteogenesis in romidepsin-treated BMSCs during induction into adipocytes or osteoblasts, respectively. Pharmacological inhibition of FAK signaling during adipogenesis or inhibition of FAK or TGFβ signaling during osteogenesis diminished the biological effects of romidepsin on BMSCs. The results of chromatin immunoprecipitation combined with quantitative polymerase chain reaction indicated a significant increase in H3K9Ac epigenetic markers in the promoter regions of peroxisome proliferator-activated receptor gamma (PPARγ) and KLF15 (related to adipogenesis) or SP7 (Osterix) and alkaline phosphatase (ALP) (related to osteogenesis) in romidepsin-treated BMSCs. Our data indicated that romidepsin is a novel in vitro modulator of adipocytic and osteoblastic differentiation of BMSCs.
Collapse
|
43
|
Wang Y, Pan J, Zhang Y, Li X, Zhang Z, Wang P, Qin Z, Li J. Wnt and Notch signaling pathways in calcium phosphate‐enhanced osteogenic differentiation: A pilot study. J Biomed Mater Res B Appl Biomater 2018; 107:149-160. [PMID: 29569393 DOI: 10.1002/jbm.b.34105] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 02/12/2018] [Accepted: 02/18/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Yu Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| | - Jianwei Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| | - Yiqun Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| | - Xiang Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| | - Zhen Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| | - Peng Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| | - Zhifan Qin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| | - Jihua Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research, Platforms & Department of Oral and Maxillofacial Surgery, West China College of StomatologySichuan UniversityChengdu610041 China
| |
Collapse
|
44
|
Rubin J, Styner M, Uzer G. Physical Signals May Affect Mesenchymal Stem Cell Differentiation via Epigenetic Controls. Exerc Sport Sci Rev 2018; 46:42-47. [PMID: 28795956 DOI: 10.1249/jes.0000000000000129] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Marrow mesenchymal stem cells supply bone osteoblasts and adipocytes. Exercise effects to increase bone and decrease fat involve transfer of signals from the cytoplasm into the nucleus to regulate gene expression. We propose that exercise control of stem cell fate relies on structural connections that terminate in the nucleus and involve intranuclear actin structures that regulate epigenetic gene expression.
Collapse
Affiliation(s)
- Janet Rubin
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Maya Styner
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Gunes Uzer
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
45
|
Rotherham M, Henstock JR, Qutachi O, El Haj AJ. Remote regulation of magnetic particle targeted Wnt signaling for bone tissue engineering. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:173-184. [DOI: 10.1016/j.nano.2017.09.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 08/14/2017] [Accepted: 09/15/2017] [Indexed: 01/18/2023]
|
46
|
Icariin Regulates the Bidirectional Differentiation of Bone Marrow Mesenchymal Stem Cells through Canonical Wnt Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:8085325. [PMID: 29445413 PMCID: PMC5763109 DOI: 10.1155/2017/8085325] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/28/2017] [Accepted: 09/12/2017] [Indexed: 01/13/2023]
Abstract
Fat infiltration within the bone marrow is easily observed in some postmenopausal women. Those fats are mainly derived from bone marrow mesenchymal stem cells (BMMSCs). The increment of adipocytes derived from BMMSCs leads to decreased osteoblasts derived from BMMSCs, so the bidirectional differentiation of BMMSCs significantly contributes to osteoporosis. Icariin is the main extractive of Herba Epimedii which is widely used in traditional Chinese medicine. In this experiment, we investigated the effect of icariin on the bidirectional differentiation of BMMSCs through quantitative real-time PCR, immunofluorescence, western blot, and tissue sections in vitro and in vivo. We found that icariin obviously promotes osteogenesis and inhibits adipogenesis through detecting staining and gene expression. Micro-CT analysis showed that icariin treatment alleviated the loss of cancellous bone of the distal femur in ovariectomized (OVX) mice. H&E staining analysis showed that icariin-treated OVX mice obtained higher bone mass and fewer bone marrow lipid droplets than OVX mice. Western blot and immunofluorescence showed that icariin regulates the bidirectional differentiation of BMMSCs via canonical Wnt signaling. This study demonstrates that icariin exerts its antiosteoporotic effect by regulating the bidirectional differentiation of BMMSCs through the canonical Wnt signaling pathway.
Collapse
|
47
|
Li KC, Chang YH, Hsu MN, Lo SC, Li WH, Hu YC. Baculovirus-Mediated miR-214 Knockdown Shifts Osteoporotic ASCs Differentiation and Improves Osteoporotic Bone Defects Repair. Sci Rep 2017; 7:16225. [PMID: 29176755 PMCID: PMC5701180 DOI: 10.1038/s41598-017-16547-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022] Open
Abstract
Osteoporotic patients often suffer from bone fracture but its healing is compromised due to impaired osteogenesis potential of bone marrow-derived mesenchymal stem cells (BMSCs). Here we aimed to exploit adipose-derived stem cells from ovariectomized rats (OVX-ASCs) for bone healing. We unraveled that OVX-ASCs highly expressed miR-214 and identified 2 miR-214 targets: CTNNB1 (β-catenin) and TAB2. We demonstrated that miR-214 targeting of these two genes blocked the Wnt pathway, led to preferable adipogenesis and hindered osteogenesis. As a result, OVX-ASCs implantation into OVX rats failed to heal critical-size metaphyseal bone defects. We further engineered the OVX-ASCs with a novel Cre/loxP-based hybrid baculovirus vector that conferred prolonged expression of miR-214 sponge. Gene delivery for miR-214 sponge expression successfully downregulated miR-214 levels, activated the Wnt pathway, upregulated osteogenic factors β-catenin/Runx2, downregulated adipogenic factors PPAR-γ and C/EBP-α, shifted the differentiation propensity towards osteogenic lineage, enhanced the osteogenesis of co-cultured OVX-BMSCs, elevated BMP7/osteoprotegerin secretion and hindered exosomal miR-214/osteopontin release. Consequently, implanting the miR-214 sponge-expressing OVX-ASCs tremendously improved bone healing in OVX rats. Co-expression of miR-214 sponge and BMP2 further synergized the OVX-ASCs-mediated bone regeneration in OVX rats. This study implicates the potential of suppressing miR-214 by baculovirus-mediated gene delivery in osteoporotic ASCs for regenerative medicine.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Yu-Han Chang
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
- Department of Orthopaedic, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Mu-Nung Hsu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Shih-Chun Lo
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Wan-Hua Li
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan.
| |
Collapse
|
48
|
Moon JS, Ko HM, Park JI, Kim JH, Kim SH, Kim MS. Inhibition of human mesenchymal stem cell proliferation via Wnt signaling activation. J Cell Biochem 2017; 119:1670-1678. [PMID: 28776719 DOI: 10.1002/jcb.26326] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/02/2017] [Indexed: 01/09/2023]
Abstract
Human mesenchymal stem cells (hMSCs), characterized by rapid in vitro expandability and multi-differentiation potential, have been widely used in the clinical field of tissue engineering. Recent studies have shown that various signaling networks are involved in the growth and differentiation of hMSCs. Although Wnts and their downstream signaling components have been implicated in the regulation of hMSCs, the role of Wnt signaling in hMSC self-renewal is still controversial. Here, it was observed that activation of endogenous canonical Wnt signaling with LiCl, which decreased β-catenin phosphorylation, leads to a decrease in hMSC proliferation. The fact that this growth arrest is not linked to apoptosis was verified by annexin V-FITC/propidium iodide assay. It was associated with sealing off of the cells in the G1 phase of the cell cycle accompanied by changes in expression of cell cycle-associated genes such as cyclin A and D. In addition, activation of Wnt signaling during hMSC proliferation seemed to reduce their clonogenic potential. On the contrary, Wnt signaling activation during hMSC proliferation had little effect on the osteogenic differentiation capability of cells. These findings show that canonical Wnt signaling is a critical regulator of hMSC proliferation and clonogenicity.
Collapse
Affiliation(s)
| | - Hyun-Mi Ko
- Department of Microbiology, College of Medicine, Seonam University, Namwon, Korea
| | - Ji-Il Park
- Department of Dental Hygiene, Gwangju Health College, Gwangju, Korea
| | - Jae-Hyung Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Sun-Hun Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Min-Seok Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, Korea
| |
Collapse
|
49
|
Zhang C, Hong FF, Wang CC, Li L, Chen JL, Liu F, Quan RF, Wang JF. TRIB3 inhibits proliferation and promotes osteogenesis in hBMSCs by regulating the ERK1/2 signaling pathway. Sci Rep 2017; 7:10342. [PMID: 28871113 PMCID: PMC5583332 DOI: 10.1038/s41598-017-10601-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 08/10/2017] [Indexed: 12/20/2022] Open
Abstract
Osteogenic differentiation in human bone marrow-derived mesenchymal stem cells (hBMSCs) is regulated by various factors, including bone morphogenetic proteins (BMPs), Notch, growth hormones and mitogen-activated protein kinases (MAPKs). Tribbles homolog 3 (TRIB3), a pseudokinase, plays an important role in cancer cells and adipocytes. However, TRIB3 function in osteogenic differentiation is unknown, although it is involved in regulating signaling pathways associated with osteogenic differentiation. Here, we found that TRIB3 was highly expressed during osteogenic differentiation in hBMSCs. Inhibition of focal adhesion kinase (FAK) or phosphatidylinositol 3-kinase (PI3K) resulted in a significant decrease in TRIB3 expression, and expression of TRIB3 was restored by increasing insulin-like growth factor-1 (IGF-1) via activating phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signaling. TRIB3 knock-down enhanced proliferation and decreased osteogenic differentiation at the middle stage of differentiation, and these effects were reversed by inhibiting the activation of extracellular signal-regulated kinase (ERK)-1/2. In conclusion, TRIB3 plays an important role in proliferation and osteogenic differentiation by regulating ERK1/2 activity at the middle stage of differentiation, and expression of TRIB3 is regulated by FAK in a PI3K/AKT-dependent manner.
Collapse
Affiliation(s)
- Cui Zhang
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Fan-Fan Hong
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Cui-Cui Wang
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Liang Li
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Jian-Ling Chen
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China
| | - Fei Liu
- Institute of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, 311200, P. R. China
| | - Ren-Fu Quan
- Institute of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, Zhejiang, 311200, P. R. China.
| | - Jin-Fu Wang
- Institute of Cell and Development Biology, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang, 310058, P. R. China.
| |
Collapse
|
50
|
Wang Y, Zhang X, Shao J, Liu H, Liu X, Luo E. Adiponectin regulates BMSC osteogenic differentiation and osteogenesis through the Wnt/β-catenin pathway. Sci Rep 2017. [PMID: 28623357 PMCID: PMC5473871 DOI: 10.1038/s41598-017-03899-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Recent studies have demonstrated the stimulatory effects of adiponectin on bone formation, but the mechanism underlying these effects remains unclear. The Wnt/β-catenin pathway, one of the most important pathways in osteogenesis, has rarely been associated with the osteogenic effects of adiponectin in previous studies. The present study was designed to investigate the effects of adiponectin on bone mesenchymal stem cell (BMSC) osteogenic differentiation and bone formation through the Wnt/β-catenin pathway. We detected adiponectin receptor expression in BMSCs, constructed a recombinant adenovirus containing the human adiponectin gene, and then used the adenovirus to transfect BMSCs in vitro or injected the adenovirus into bone defect areas in animal models. Wnt/β-catenin pathway and osteogenesis were detected by real-time PCR, western blotting, immunofluorescence, HE staining and micro-CT. In both our in vivo and in vitro experiments, we detected higher gene and protein expression levels of the Wnt/β-catenin pathway-related factors β-catenin and cyclinD1 in adiponectin transgenic BMSCs and rats. Similar results were noted regarding the gene and protein expression levels of osteogenesis-related genes. In addition, more new bone formation was observed in the adiponectin-treated groups. Our results indicate that adiponectin could facilitate BMSC osteogenic differentiation and osteogenesis, and the Wnt/β-catenin pathway was involved in the osteogenic effect of adiponectin.
Collapse
Affiliation(s)
- Yiyao Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xiaohui Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jun Shao
- Department of Stomatology, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, 510800, People's Republic of China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xian Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - En Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|